Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Neurochem ; 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38994800

ABSTRACT

Oxidative stress is one of the major culprits causing dopaminergic neuron loss in Parkinson's disease (PD). DJ-1 is a protein with multiple actions against oxidative stress, apoptosis, neuroinflammation, etc. DJ-1 expression is decreased in sporadic PD, therefore increasing DJ-1 expression might be beneficial in PD treatment. However, drugs known to upregulate DJ-1 are still lacking. In this study, we identified a novel DJ-1-elevating compound called ChemJ through luciferase assay-based high-throughput compound screening in SH-SY5Y cells and confirmed that ChemJ upregulated DJ-1 in SH-SY5Y cell line and primary cortical neurons. DJ-1 upregulation by ChemJ alleviated MPP+-induced oxidative stress. In exploring the underlying mechanisms, we found that the transcription factor CREB1 bound to DJ-1 promoter and positively regulated its expression under both unstressed and 1-methyl-4-phenylpyridinium-induced oxidative stress conditions and that ChemJ promoted DJ-1 expression via activating PKA/CREB1 pathway in SH-SY5Y cells. Our results demonstrated that ChemJ alleviated the MPP+-induced oxidative stress through a PKA/CREB1-mediated regulation of DJ-1 expression, thus offering a novel and promising avenue for PD treatment.

2.
Brain ; 142(5): 1349-1364, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30938419

ABSTRACT

Amyotrophic lateral sclerosis is a deleterious neurodegenerative disease without effective treatment options. Recent studies have indicated the involvement of the dysregulation of RNA metabolism in the pathogenesis of amyotrophic lateral sclerosis. Among the various RNA regulatory machineries, nonsense-mediated mRNA decay (NMD) is a stress responsive cellular surveillance system that degrades selected mRNA substrates to prevent the translation of defective or harmful proteins. Whether this pathway is affected in neurodegenerative diseases is unclear. Here we report the inhibition of NMD by arginine-rich dipeptide repeats derived from C9orf72 hexanucleotide repeat expansion, the most common cause of familial amyotrophic lateral sclerosis. Bioinformatic analysis of multiple transcriptome profiles revealed significant overlap of upregulated genes in NMD-defective cells with those in the brain tissues, micro-dissected motor neurons, or induced pluripotent stem cell-derived motor neurons specifically from amyotrophic lateral sclerosis patients carrying C9orf72 hexanucleotide repeat expansion, suggesting the suppression of NMD pathway in these patients. Using Drosophila as a model, we have validated that the C9orf72 hexanucleotide repeat expansion products could lead to the accumulation of the NMD substrates and identified arginine-rich dipeptide repeats, including poly glycine-arginine and poly proline-arginine, as the main culprits of NMD inhibition. Furthermore, in human SH-SY5Y neuroblastoma cells and in mouse brains, expression of glycine-arginine with 36 repeats (GR36) was sufficient to cause NMD inhibition. In cells expressing GR36, stress granule accumulation was accompanied by decreased processing body formation, which contributed to the inhibition of NMD. Remarkably, expression of UPF1, a core gene in the NMD pathway, efficiently blocked neurotoxicity caused by arginine-rich dipeptide repeats in both cellular and Drosophila models. Although not as effective as UPF1, expression of another NMD gene UPF2 also ameliorated the degenerative phenotypes in dipeptide repeat-expressing flies, indicating that genetically reactivating the NMD pathway could suppress dipeptide repeat toxicity. Finally, after validating tranilast as an NMD-activating drug, we demonstrated the therapeutic potential of this asthma drug in cellular and Drosophila models of C9orf72 dipeptide repeat neurotoxicity. Therefore, our study has revealed a cellular mechanism whereby arginine-rich C9orf72 dipeptide repeats could inhibit NMD activities by reducing the abundance of processing bodies. Furthermore, our results suggested that activation of the NMD pathway could be a potential therapeutic strategy for amyotrophic lateral sclerosis with defective RNA metabolism.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Nonsense Mediated mRNA Decay/physiology , Amyotrophic Lateral Sclerosis/drug therapy , Animals , Animals, Genetically Modified , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cell Line, Tumor , Dipeptides/genetics , Dipeptides/metabolism , Drosophila , Female , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Nonsense Mediated mRNA Decay/drug effects , ortho-Aminobenzoates/pharmacology , ortho-Aminobenzoates/therapeutic use
3.
J Biol Chem ; 291(27): 14373-14384, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27226613

ABSTRACT

Valosin-containing protein/p97(VCP) is a hexameric ATPase vital to protein degradation during endoplasmic reticulum stress. It regulates diverse cellular functions including autophagy, chromatin remodeling, and DNA repair. In addition, mutations in VCP cause inclusion body myopathy, Paget disease of the bone, and frontotemporal dementia (IBMPFD), as well as amyotrophic lateral sclerosis. Nevertheless, how the VCP activities were regulated and how the pathogenic mutations affect the function of VCP during stress are not unclear. Here we show that the small ubiquitin-like modifier (SUMO)-ylation of VCP is a normal stress response inhibited by the disease-causing mutations in the N-domain. Under oxidative and endoplasmic reticulum stress conditions, the SUMOylation of VCP facilitates the distribution of VCP to stress granules and nucleus, and promotes the VCP hexamer assembly. In contrast, pathogenic mutations in the VCP N-domain lead to reduced SUMOylation and weakened VCP hexamer formation upon stress. Defective SUMOylation of VCP also causes altered co-factor binding and attenuated endoplasmic reticulum-associated protein degradation. Furthermore, SUMO-defective VCP fails to protect against stress-induced toxicity in Drosophila Therefore, our results have revealed SUMOylation as a molecular signaling switch to regulate the distribution and functions of VCP during stress response, and suggest that deficiency in VCP SUMOylation caused by pathogenic mutations will render cells vulnerable to stress insults.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Endoplasmic Reticulum Stress , Mutation, Missense , Sumoylation , Adenosine Triphosphatases/genetics , Amino Acid Substitution , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Drosophila Proteins/genetics , Drosophila melanogaster , Humans , Male , Middle Aged , Protein Structure, Tertiary , Valosin Containing Protein
4.
Cell Death Dis ; 10(3): 215, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30833545

ABSTRACT

Stress granule formation is important for stress response in normal cells and could lead to chemotherapy resistance in cancer cells. Aberrant stress granule dynamics are also known to disrupt proteostasis, affect RNA metabolism, and contribute to neuronal cell death. Meanwhile, circadian abnormality is an aging-related risk factor for cancer and neurodegeneration. Whether stress granule dynamics are circadian regulated is entirely unknown. Here we show that the formation of stress granules varied by zeitgeber time in mouse liver. Moreover, altering circadian regulation by silencing the core circadian gene Bmal1 in a cell line expressing an endogenous GFP-tagged G3BP1 significantly increased stress granule dynamics, while the overexpression of Bmal1 decreased them. Surprisingly, increased stress granule dynamics and formation by transient decrease of BMAL1 coincided with increased resistance to stress-induced cell death. The circadian regulation of stress granules was mediated by oscillating eIF2α expression. At zeitgeber time when BMAL1 and eIF2α were at nadir, reduction of unphosphorylated eIF2α could significantly alter the ratio of phosphorylated/total eIF2α and quickly lead to increased formation of stress granules. Therefore, diurnal oscillating eIF2α connects the circadian cue to a cellular stress response mechanism that is vital for both neurodegeneration and cancer.


Subject(s)
Circadian Rhythm/genetics , Eukaryotic Initiation Factor-2/physiology , Stress, Physiological , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , ARNTL Transcription Factors/physiology , Animals , Drug Resistance, Neoplasm , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation , Male , Mice, Inbred C57BL
5.
Transl Neurodegener ; 7: 24, 2018.
Article in English | MEDLINE | ID: mdl-30338063

ABSTRACT

BACKGROUND: Circadian rhythms are oscillating physiological and behavioral changes governed by an internal molecular clock, and dysfunctions in circadian rhythms have been associated with ageing and various neurodegenerative diseases. However, the evidence directly connecting the neurodegeneration-associated proteins to circadian control at the molecular level remains sparse. METHODS: Using meta-analysis, synchronized animals and cell lines, cells and tissues from FUS R521C knock-in rats, we examined the role of FUS in circadian gene expression regulation. RESULTS: We found that FUS, an oscillating expressed nuclear protein implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), exerted a novel feedback route to regulate circadian gene expression. Nr1d1-encoded core circadian protein REV-ERBα bound the Fus promoter and regulated the expression of Fus. Meanwhile, FUS was in the same complex as PER/CRY, and repressed the expression of E box-containing core circadian genes, such as Per2, by mediating the promoter occupancy of PSF-HDAC1. Remarkably, a common pathogenic mutant FUS (R521C) showed increased binding to PSF, and caused decreased expression of Per2. CONCLUSIONS: Therefore, we have demonstrated FUS as a modulator of circadian gene expression, and provided novel mechanistic insights into the mutual influence between circadian control and neurodegeneration-associated proteins.

6.
Neurobiol Aging ; 72: 159-170, 2018 12.
Article in English | MEDLINE | ID: mdl-30273830

ABSTRACT

Mutations in fused in sarcoma (Fus) cause familial amyotrophic lateral sclerosis (ALS) and occasionally frontotemporal dementia. Here we report the establishment and characterization of a novel knockin (KI) rat model expressing a Fus point mutation (R521C) via CRISPR/Cas9. The mutant animals developed adult-onset learning and memory behavioral deficits, with reduced spine density in hippocampal neurons. Remarkably, sleep-wake cycle and circadian abnormalities preceded the onset of cognitive deficit. RNA-seq study further demonstrated altered expression of some key sleep and circadian regulators, such as orexin/hypocretin receptor type 2 and casein kinase 1 epsilon, in the mutant rats. Therefore, we have established a rodent model expressing physiological level of a pathogenic mutant FUS, and we found cognitive impairment as a main behavioral deficit at mid age. Furthermore, we have revealed a new role of FUS in sleep and circadian regulation and demonstrated that functional change in FUS could cause sleep-wake and circadian disturbance as early symptoms.


Subject(s)
Behavior, Animal , Chronobiology Disorders/genetics , Cognitive Dysfunction/genetics , RNA-Binding Protein FUS/metabolism , Sleep/genetics , Wakefulness/genetics , Age Factors , Animals , Cells, Cultured , Disease Models, Animal , Electroencephalography , Electromyography , Embryo, Mammalian , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Sleep Disorders, Circadian Rhythm/genetics
7.
PLoS One ; 12(7): e0180892, 2017.
Article in English | MEDLINE | ID: mdl-28700687

ABSTRACT

Autophagy-lysosomal pathway is a cellular protective system to remove aggregated proteins and damaged organelles. Meanwhile, exosome secretion has emerged as a mode to selectively clear the neurotoxic proteins, such as α-synuclein. Mounting evidence suggests that these two cellular processes are coordinated to facilitate the clearance of toxic cellular waste; however the regulators for the transition between these two processes are unclear. Here we show that SCAMP5, a secretory carrier membrane protein significantly induced in the brains of Huntington's disease patients, is quickly and transiently induced by protein stress and autophagic stimulation, and is regulated by the master autophagy transcriptional regulator TFEB. Ironically, SCAMP5 inhibits autophagy flux by blocking the fusion of autophagosomes and lysosomes. Although autophagy is blocked, SCAMP5 does not cause significant protein aggregation in cells. Instead, it promotes the Golgi fragmentation and stimulates the unconventional secretion of the co-localizing α-synuclein via exosome as an exosome component. Therefore, we have identified SCAMP5 as a novel coordinator of autophagy and exosome secretion, which is induced upon protein stress to channel the efficient clearance of toxic proteins via the exosomes rather than autophagy-lysosomal pathway.


Subject(s)
Exosomes/metabolism , alpha-Synuclein/metabolism , Autophagy/genetics , Autophagy/physiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Exosomes/genetics , Fluorescent Antibody Technique , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Immunoblotting , Immunoprecipitation , Lysosomes/genetics , Lysosomes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , RNA, Small Interfering/genetics , alpha-Synuclein/genetics
8.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 25(9): 794-7, 801, 2009 Sep.
Article in Zh | MEDLINE | ID: mdl-19737463

ABSTRACT

AIM: To investigate the tropism of bone marrow mesenchymal stem cells (BMSCs) in neurogenic differentiation for glioma and SDF-1alpha. METHODS: BHA, bFGF, and DMSO were used to induce the neural differentiation of BMSCs, and the expression of neural markers, Nestin, beta-III-Tubulin and NSE were analyzed by immunocytochemistry. We investigated, by using the Dunn chamber, the migration of BMSCs in neurogenic differentiation towards glioma and SDF-1alpha. RESULTS: BMSCs could be induced to differentiate into neuron-like cells, while the control remained the morphology of BMSCs. Dunn chamber analysis revealed that both the migration speed and efficiency of cells induced by C6 conditioned medium and SDF-1alpha were higher than control, demonstrating the chemotactic effect of both C6 conditioned medium and SDF-1alpha on BMSCs. These results were confimed by examination of migration tracks of individual cells. Moreover, BMSCs at different states of differentiation showed different degree of tropism for C6 conditioned medium and SDF-1alpha. CONCLUSION: The directed migration of BMSCs is closely related to the differentiation states of these cells.


Subject(s)
Cell Differentiation , Chemokine CXCL12/metabolism , Mesenchymal Stem Cells/cytology , Neurons/cytology , Animals , Cells, Cultured , Culture Media, Conditioned/metabolism , Mesenchymal Stem Cells/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL