Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
2.
Autophagy ; 12(8): 1215-28, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27171370

ABSTRACT

As a selective degradation system, chaperone-mediated autophagy (CMA) is essential for maintaining cellular homeostasis and survival under stress conditions. Increasing evidence points to an important role for the dysfunction of CMA in the pathogenesis of Parkinson disease (PD). However, the mechanisms by which CMA regulates neuronal survival under stress and its role in neurodegenerative diseases are not fully understood. PARK7/DJ-1 is an autosomal recessive familial PD gene. PARK7 plays a critical role in antioxidative response and its dysfunction leads to mitochondrial defects. In the current study, we showed that CMA mediated the lysosome-dependent degradation of PARK7. Importantly, CMA preferentially removed the oxidatively damaged nonfunctional PARK7 protein. Furthermore, CMA protected cells from mitochondrial toxin MPP(+)-induced changes in mitochondrial morphology and function, and increased cell viability. These protective effects were lost under PARK7-deficiency conditions. Conversely, overexpression of PARK7 significantly attenuated the mitochondrial dysfunction and cell death exacerbated by blocking CMA under oxidative stress. Thus, our findings reveal a mechanism by which CMA protects mitochondrial function by degrading nonfunctional PARK7 and maintaining its homeostasis, and dysregulation of this pathway may contribute to the neuronal stress and death in PD pathogenesis.


Subject(s)
Autophagy , Mitochondria/metabolism , Molecular Chaperones/metabolism , Protein Deglycase DJ-1/metabolism , Amino Acid Motifs , Animals , Antioxidants/metabolism , Cell Death , Cell Survival , Gene Expression Regulation , HEK293 Cells , Homeostasis , Humans , Lysosomes/metabolism , Male , Mice , Mice, Transgenic , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Oxidative Stress , Parkinson Disease/metabolism , Rats, Sprague-Dawley
3.
Neurosci Bull ; 31(4): 452-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26206599

ABSTRACT

Chaperone-mediated autophagy (CMA), one of the main pathways of lysosomal proteolysis, is characterized by the selective targeting and direct translocation into the lysosomal lumen of substrate proteins containing a targeting motif biochemically related to the pentapeptide KFERQ. Along with the other two lysosomal pathways, macro- and micro-autophagy, CMA is essential for maintaining cellular homeostasis and survival by selectively degrading misfolded, oxidized, or damaged cytosolic proteins. CMA plays an important role in pathologies such as cancer, kidney disorders, and neurodegenerative diseases. Neurons are post-mitotic and highly susceptible to dysfunction of cellular quality-control systems. Maintaining a balance between protein synthesis and degradation is critical for neuronal functions and homeostasis. Recent studies have revealed several new mechanisms by which CMA protects neurons through regulating factors critical for their viability and homeostasis. In the current review, we summarize recent advances in the understanding of the regulation and physiology of CMA with a specific focus on its possible roles in neuroprotection.


Subject(s)
Autophagy/physiology , Lysosomes/physiology , Neurons/physiology , Alzheimer Disease/metabolism , Animals , Humans , Huntington Disease/metabolism , Lysosomes/metabolism , Molecular Chaperones/metabolism , Molecular Chaperones/physiology , Neurons/metabolism , Parkinson Disease/metabolism
4.
J Neuroinflammation ; 12: 33, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25890150

ABSTRACT

BACKGROUND: Neuroinflammatory responses have been recognized as an important aspect in the pathogenesis of Parkinson's disease (PD). Transcriptional regulation plays a critical role in the process of inflammation. Transcription factor myocyte enhancer factor 2D (MEF2D) is identified as a central factor in transmission of extracellular signals and activation of the genetic programs in response to a wide range of stimuli in several cell types, including neurons. But its presence and function in microglia have not been reported. We therefore investigated the effect of MEF2D in activated microglia on the progress of neuroinflammation and the survival of neurons. METHODS: BV2 cells and primary cultured glial cells were stimulated with lipopolysaccharide (LPS). Samples from cells were examined for MEF2D expression, interleukin-10 (IL-10), and tumor necrosis factor alpha (TNF-α) by immunoblotting, quantitative real-time PCR (qPCR) or enzyme-linked immunosorbent assay (ELISA). The activity of MEF2D was examined by electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation assay (ChIP). Recombinant lentivirus expressing shRNA specific to MEF2D was used to silence MEF2D expression in BV2 cells. The role of IL-10 transcriptionally induced by MEF2D on neuronal survival was assessed by anti-IL-10 neutralizing antibody. The survival of neurons was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining. Male C57bl/6 mice were used to establish an acute PD model. Brain sections and cell slides were tested by immunofluorescence. RESULTS: We demonstrated that MEF2D was present in microglia. Activation of microglia was associated with an increase in MEF2D level and activity in response to different stimuli in vivo and in vitro. MEF2D bound to a MEF2 consensus site in the promoter region of IL-10 gene and stimulated IL-10 transcription. Silencing MEF2D decreased the level of IL-10, increased the TNF-α mRNA, and promoted inflammation-induced cytotoxicity, consistent with the result of inhibiting IL-10 activity with an anti-IL-10 neutralizing antibody. CONCLUSIONS: Our study identifies MEF2D as a critical regulator of IL-10 gene expression that negatively controls microglia inflammation response and prevents inflammation-mediated cytotoxicity.


Subject(s)
Apoptosis/physiology , Interleukin-10/metabolism , MEF2 Transcription Factors/metabolism , Microglia/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Brain/cytology , Calcium-Binding Proteins/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Interleukin-10/genetics , Lipopolysaccharides/toxicity , MEF2 Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microglia/drug effects , RNA, Messenger/metabolism , Time Factors , Tyrosine 3-Monooxygenase/metabolism
5.
Biochem Biophys Res Commun ; 458(2): 227-33, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25623535

ABSTRACT

Oxidative stress and mitochondrial dysfunction have been linked to Parkinson's disease. DJ-1 is a recessive familial PD gene involved in antioxidative function and mitochondrial maintenance. Myricitrin, a flavanoid isolated from the root bark of Myrica cerifera, has potent antioxidative properties. In the present study, we investigated the protective effects of myricitrin against MPP(+)-induced mitochondrial dysfunction in SN4741 cells and attempted to elucidate the mechanisms underlying this protection. The results showed that incubating SN4741 cells with myricitrin significantly reduced cell death induced by the neurotoxin MPP(+). Furthermore, myricitrin protected cells from MPP(+)-induced effects on mitochondrial morphology and function. However, these protective effects were lost under DJ-1-deficient conditions. Thus, our results suggest that myricitrin alleviates MPP(+)-induced mitochondrial dysfunction and increases cell viability via DJ-1, indicating that myricitrin is a potential beneficial agent for age-related neurodegenerative diseases, particularly Parkinson's disease.


Subject(s)
1-Methyl-4-phenylpyridinium/administration & dosage , Flavonoids/administration & dosage , Mitochondria/physiology , Neurons/physiology , Oncogene Proteins/metabolism , Peroxiredoxins/metabolism , Animals , Antioxidants/administration & dosage , Apoptosis/drug effects , Apoptosis/physiology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Interactions , Mice , Mitochondria/drug effects , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Neurotoxins/administration & dosage , Protein Deglycase DJ-1
6.
J Neurochem ; 130(5): 720-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24848448

ABSTRACT

Progressive loss of dopaminergic (DA) neurons in the substantial nigra pars compacta (SNc) is an important pathological feature in Parkinson's disease (PD). Loss of transcription factor myocyte enhancer factor 2D (MEF2D), a key neuronal survival factor, has been shown to underlie the loss of DA neurons in SNc and the pathogenic process of PD. It is known that PD-associated neurotoxins reduce the level of MEF2D protein to trigger neuronal death. Although neurotoxins clearly destabilize MEF2D by post-translational mechanisms, it is not known whether regulation of MEF2D mRNA contributes to neurotoxin-induced decrease in MEF2D protein. In this work, we showed that MPP(+), the toxic metabolite of MPTP, caused a significant decrease in the half-life and total level of MEF2D mRNA in a DA neuronal cell line, SN4741 cells. Quantitative PCR analysis of the SNc DA neurons captured by immune-laser capture microdissection showed that exposure to MPTP led to a marked reduction in the level of MEF2D mRNA in SNc DA neurons compared to controls. Down-regulation of MEF2D mRNA alone reduced the viability of SN4741 cells and sensitized the cells to MPP(+)-induced toxicity. These results suggest that destabilization and reduction in MEF2D mRNA is in part responsible for neurotoxin-induced decrease in MEF2D protein and neuronal viability. Myocyte enhancer factor 2D (MEF2D) plays an important role in neuronal survival. How MEF2D mRNA is deregulated under toxic stress is unclear. We found that PD-associated neurotoxins destabilize MEF2D mRNA and reduce its level in vitro and in vivo. Reduction in MEF2D mRNA is sufficient to sensitize model cells to neurotoxin-induced toxicity, suggesting that destabilization of MEF2D mRNA is part of the mechanism by which neurotoxins trigger deregulation of neuronal survival.


Subject(s)
MPTP Poisoning/metabolism , Neurotoxins/pharmacology , RNA Stability/drug effects , RNA, Messenger/drug effects , Animals , Apoptosis/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Laser Capture Microdissection , MEF2 Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL