Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Sci Signal ; 17(829): eadk8249, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38530880

ABSTRACT

Mutations in the gene encoding the lysosomal enzyme glucocerebrosidase (GCase) are responsible for Gaucher disease (GD) and are considered the strongest genetic risk factor for Parkinson's disease (PD) and Lewy body dementia (LBD). GCase deficiency leads to extensive accumulation of glucosylceramides (GCs) in cells and contributes to the neuropathology of GD, PD, and LBD by triggering chronic neuroinflammation. Here, we investigated the mechanisms by which GC accumulation induces neuroinflammation. We found that GC accumulation within microglia induced by pharmacological inhibition of GCase triggered STING-dependent inflammation, which contributed to neuronal loss both in vitro and in vivo. GC accumulation in microglia induced mitochondrial DNA (mtDNA) leakage to the cytosol to trigger STING-dependent inflammation. Rapamycin, a compound that promotes lysosomal activity, improved mitochondrial function, thereby decreasing STING signaling. Furthermore, lysosomal damage caused by GC accumulation led to defects in the degradation of activated STING, further exacerbating inflammation mediated by microglia. Thus, limiting STING activity may be a strategy to suppress neuroinflammation caused by GCase deficiency.


Subject(s)
Gaucher Disease , Parkinson Disease , Animals , Mice , alpha-Synuclein/metabolism , Gaucher Disease/genetics , Gaucher Disease/pathology , Glucosylceramides/metabolism , Inflammation/metabolism , Lysosomes/metabolism , Microglia/metabolism , Neuroinflammatory Diseases , Parkinson Disease/metabolism
2.
Cell Mol Life Sci ; 81(1): 128, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38472451

ABSTRACT

Epigenetic dysregulation that leads to alterations in gene expression and is suggested to be one of the key pathophysiological factors of Parkinson's disease (PD). Here, we found that α-synuclein preformed fibrils (PFFs) induced histone H3 dimethylation at lysine 9 (H3K9me2) and increased the euchromatic histone methyltransferases EHMT1 and EHMT2, which were accompanied by neuronal synaptic damage, including loss of synapses and diminished expression levels of synaptic-related proteins. Furthermore, the levels of H3K9me2 at promoters in genes that encode the synaptic-related proteins SNAP25, PSD95, Synapsin 1 and vGLUT1 were increased in primary neurons after PFF treatment, which suggests a linkage between H3K9 dimethylation and synaptic dysfunction. Inhibition of EHMT1/2 with the specific inhibitor A-366 or shRNA suppressed histone methylation and alleviated synaptic damage in primary neurons that were treated with PFFs. In addition, the synaptic damage and motor impairment in mice that were injected with PFFs were repressed by treatment with the EHMT1/2 inhibitor A-366. Thus, our findings reveal the role of histone H3 modification by EHMT1/2 in synaptic damage and motor impairment in a PFF animal model, suggesting the involvement of epigenetic dysregulation in PD pathogenesis.


Subject(s)
Motor Disorders , Parkinson Disease , Animals , Mice , Histones/metabolism , Methylation , Neurons/metabolism , alpha-Synuclein/metabolism
3.
Hum Mol Genet ; 33(1): 64-77, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37756636

ABSTRACT

GGGGCC (G4C2) hexanucleotide repeat expansion (HRE) in the first intron of the chromosome 9 open reading frame 72 (C9ORF72) gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Among the five dipeptide repeat proteins translated from G4C2 HRE, arginine-rich poly-PR (proline:arginine) is extremely toxic. However, the molecular mechanism responsible for poly-PR-induced cell toxicity remains incompletely understood. Here, we found that poly-PR overexpression triggers severe DNA damage in cultured cells, primary cortical neurons, and the motor cortex of a poly-PR transgenic mouse model. Interestingly, we identified a linkage between poly-PR and RNA-binding protein fused in sarcoma (FUS), another ALS-related gene product associated with DNA repair. Poly-PR interacts with FUS both in vitro and in vivo, phase separates with FUS in a poly-PR concentration-dependent manner, and impairs the fluidity of FUS droplets in vitro and in cells. Moreover, poly-PR impedes the recruitment of FUS and its downstream protein XRCC1 to DNA damage foci after microirradiation. Importantly, overexpression of FUS significantly decreased the level of DNA damage and dramatically reduced poly-PR-induced cell death. Our data suggest the severe DNA damage caused by poly-PR and highlight the interconnection between poly-PR and FUS, enlightening the potential therapeutic role of FUS in alleviating poly-PR-induced cell toxicity.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Animals , Mice , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , DNA Repeat Expansion , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Proteins/genetics , DNA Damage/genetics , Arginine/genetics , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Dipeptides/genetics
4.
Eur Spine J ; 30(10): 3115-3127, 2021 10.
Article in English | MEDLINE | ID: mdl-34392419

ABSTRACT

OBJECTIVE: We focus on providing the first comprehensive national dataset on the incidence, injury aetiology and mortality of TSCI in China. METHODS: A multi-stage stratified cluster sampling method was used. We included TSCI cases from all hospitals in three regions, nine provinces and 27 cities in China via search of electronic medical records and retrospectively analysed the characteristics of TSCI in China from 2009 to 2018. We estimated the incidence of TSCI in the total population and subgroups. RESULTS: There were 5954 actual cases in 2009, corresponding to a total estimated TSCI incidence of 45.1 cases per million population (95% CI, 44.0-46.3). There were 10,074 actual cases in 2018, corresponding to a total estimated TSCI incidence of 66.5 cases per million population (95% CI, 65.2-67.8) (P < 0.001; annual average percentage change (AAPC), 4.4%). From 2009 to 2018, the incidence of almost all sex/age groups showed an increasing trend over time (P < 0.001; AAPC, 0.7-8.8%). The elderly population (aged 65-74) displayed the highest incidence of TSCI (with an average annual incidence of 127.1 cases per million [95% CI, 119.8-134.3]). CONCLUSIONS: The TSCI incidence increased significantly from 2009 to 2018. The incidence in the elderly populations was consistently high and continues to increase over time. The mortality of TSCI patients in hospitals is relatively low and continues to decrease each year, but elderly individuals remain at a high risk of hospital death.


Subject(s)
Spinal Cord Injuries , Aged , China/epidemiology , Humans , Incidence , Research Design , Retrospective Studies , Spinal Cord Injuries/epidemiology
5.
J Cell Physiol ; 235(2): 869-879, 2020 02.
Article in English | MEDLINE | ID: mdl-31232473

ABSTRACT

Lack of dopamine production and neurodegeneration of dopaminergic neurons in the substantia nigra are considered as the major characteristics of Parkinson's disease, a prevalent movement disorder worldwide. DJ-1 mutation leading to loss of its protein functions is a genetic factor of PD. In this study, our results illustrated that DJ-1 can directly interact with Ca2+ /calmodulin-dependent protein kinase kinase ß (CaMKKß) and modifies the cAMP-responsive element binding protein 1 (CREB1) activity, thus regulates tyrosine hydroxylase (TH) expression. In Dj-1 knockout mouse substantia nigra, the levels of TH and the phosphorylation of CREB1 Ser133 are significantly decreased. Moreover, Dj-1 deficiency suppresses the phosphorylation of CaMKIV (Thr196/200) and CREB1 (Ser133), subsequently inhibits TH expression in vitro. Furthermore, Knockdown of Creb1 abolishes the effects of DJ-1 on TH regulation. Our data reveal a novel pathway in which DJ-1 regulates CaMKKß/CaMKIV/CREB1 activities to facilitate TH expression.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Parkinson Disease/pathology , Protein Deglycase DJ-1/metabolism , Tyrosine 3-Monooxygenase/biosynthesis , Animals , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/genetics , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Knockout , Phosphorylation , Signal Transduction , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
6.
Nat Commun ; 10(1): 2906, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31266945

ABSTRACT

A GGGGCC hexanucleotide repeat expansion in intron 1 of chromosome 9 open reading frame 72 (C9ORF72) gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. Repeat-associated non-ATG translation of dipeptide repeat proteins (DPRs) contributes to the neuropathological features of c9FTD/ALS. Among the five DPRs, arginine-rich poly-PR are reported to be the most toxic. Here, we generate a transgenic mouse line that expresses poly-PR (GFP-PR28) specifically in neurons. GFP-PR28 homozygous mice show decreased survival time, while the heterozygous mice show motor imbalance, decreased brain weight, loss of Purkinje cells and lower motor neurons, and inflammation in the cerebellum and spinal cord. Transcriptional analysis shows that in the cerebellum, GFP-PR28 heterozygous mice show differential expression of genes related to synaptic transmission. Our findings show that GFP-PR28 transgenic mice partly model neuropathological features of c9FTD/ALS, and show a role for poly-PR in neurodegeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , C9orf72 Protein/genetics , Dipeptides/genetics , Disease Models, Animal , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , C9orf72 Protein/metabolism , Dipeptides/metabolism , Dipeptides/toxicity , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Motor Activity
7.
Neurosci Bull ; 35(5): 889-900, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31148094

ABSTRACT

GGGGCC repeat expansions in the C9ORF72 gene are the most common cause of amyotrophic lateral sclerosis and frontotemporal dementia (c9ALS/FTD). It has been reported that hexanucleotide repeat expansions in C9ORF72 produce five dipeptide repeat (DPR) proteins by an unconventional repeat-associated non-ATG (RAN) translation. Within the five DPR proteins, poly-PR and poly-GR that contain arginine are more toxic than the other DPRs (poly-GA, poly-GP, and poly-PA). Here, we demonstrated that poly-PR peptides transferred into cells by endocytosis in a clathrin-dependent manner, leading to endoplasmic reticulum stress and cell death. In SH-SY5Y cells and primary cortical neurons, poly-PR activated JUN amino-terminal kinase (JNK) and increased the levels of p53 and Bax. The uptake of poly-PR peptides by cells was significantly inhibited by knockdown of clathrin or by chlorpromazine, an inhibitor that blocks clathrin-mediated endocytosis. Inhibition of clathrin-dependent endocytosis by chlorpromazine significantly blocked the transfer of poly-PR peptides into cells, and attenuated poly-PR-induced JNK activation and cell death. Our data revealed that the uptake of poly-PR undergoes clathrin-dependent endocytosis and blockade of this process prevents the toxic effects of synthetic poly-PR peptides.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Clathrin/deficiency , Dipeptides/metabolism , Endocytosis/physiology , Frontotemporal Dementia/metabolism , Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein , Cell Line, Tumor , Clathrin/genetics , Dipeptides/genetics , Endoplasmic Reticulum Stress/physiology , Frontotemporal Dementia/genetics , Gene Knockdown Techniques/methods , Humans
8.
Acta Pharmacol Sin ; 40(1): 26-34, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29950615

ABSTRACT

REV-ERBα, the NR1D1 (nuclear receptor subfamily 1, group D, member 1) gene product, is a dominant transcriptional silencer that represses the expression of genes involved in numerous physiological functions, including circadian rhythm, inflammation, and metabolism, and plays a crucial role in maintaining immune functions. Microglia-mediated neuroinflammation is tightly associated with various neurodegenerative diseases and psychiatric disorders. However, the role of REV-ERBα in neuroinflammation is largely unclear. In this study, we investigated whether and how pharmacological activation of REV-ERBα affected lipopolysaccharide (LPS)-induced neuroinflammation in mouse microglia in vitro and in vivo. In BV2 cells or primary mouse cultured microglia, application of REV-ERBα agonist GSK4112 or SR9011 dose-dependently suppressed LPS-induced microglial activation through the nuclear factor kappa B (NF-κB) pathway. In BV2 cells, pretreatment with GSK4112 inhibited LPS-induced phosphorylation of the inhibitor of NF-κB alpha (IκBα) kinase (IκK), thus restraining the phosphorylation and degradation of IκBα, and blocked the nuclear translocation of p65, a NF-κB subunit, thereby suppressing the expression and secretion of the proinflammatory cytokines, such as interleukin 6 (IL-6) and tumor necrosis factor α (TNFα). Moreover, REV-ERBα agonist-induced inhibition on neuroinflammation protected neurons from microglial activation-induced damage, which were also demonstrated in mice with their ventral midbrain microinjected with GSK4112, and then stimulated with LPS. Our results reveal that enhanced REV-ERBα activity suppresses microglial activation through the NF-κB pathway in the central nervous system.


Subject(s)
Glycine/analogs & derivatives , Microglia/drug effects , Nuclear Receptor Subfamily 1, Group D, Member 1/agonists , Pyrrolidines/therapeutic use , Signal Transduction/drug effects , Thiophenes/therapeutic use , Transcription Factor RelA/metabolism , Animals , Cell Line, Tumor , Glycine/pharmacology , Glycine/therapeutic use , HEK293 Cells , Humans , Inflammation/drug therapy , Male , Mesencephalon/physiopathology , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Pyrrolidines/pharmacology , Thiophenes/pharmacology
9.
Front Cell Neurosci ; 12: 400, 2018.
Article in English | MEDLINE | ID: mdl-30455635

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder. Although its pathogenesis remains unclear, growing evidencce suggests that microglia-mediated neuroinflammation contributes greatly to the progression of PD. P7C3, an aminopropyl carbazole, possesses significant neuroprotective effects in several neurodegenerative disease animal models, including PD. In this study, we designed to investigate the effects of P7C3 on neuroinflammation. We showed that P7C3 specially suppressed the expression of lipopolysaccharide (LPS)-induced pro-inflammatory factors but not influenced the anti-inflammatory factors in microglia. The inhibition of the nuclear factor κB (NF-κB) signaling pathway was involved in the mechanisms of the anti-inflammatory effects by P7C3. LPS-induced activation of IκB kinase (IKK), degradation of the inhibitory κB alpha (IκBα) and nuclear translocation of NF-κB can be attenuated by the pretreatment of P7C3 in microglia. Furthermore, in LPS-treated microglia, P7C3-pretreatment decreased the toxicity of conditioned media to MES23.5 cells (a dopaminergic (DA) cell line). Most importantly, the anti-inflammatory effects of P7C3 were observed in LPS-stimulated mouse model. In general, our study demonstrates that P7C3 inhibits LPS-induced microglial activation through repressing the NF-κB pathway both in vivo and in vitro, providing a theoretical basis for P7C3 in anti-inflammation.

10.
J Biol Chem ; 293(14): 5090-5101, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29449373

ABSTRACT

Abelson helper integration site 1 (AHI1) is associated with several neuropsychiatric and brain developmental disorders, such as schizophrenia, depression, autism, and Joubert syndrome. Ahi1 deficiency in mice leads to behaviors typical of depression. However, the mechanisms by which AHI1 regulates behavior remain to be elucidated. Here, we found that down-regulation of expression of the rate-limiting enzyme in dopamine biosynthesis, tyrosine hydroxylase (TH), in the midbrains of Ahi1-knockout (KO) mice is responsible for Ahi1-deficiency-mediated depressive symptoms. We also found that Rev-Erbα, a TH transcriptional repressor and circadian regulator, is up-regulated in the Ahi1-KO mouse midbrains and Ahi1-knockdown Neuro-2a cells. Moreover, brain and muscle Arnt-like protein 1 (BMAL1), the Rev-Erbα transcriptional regulator, is also increased in the Ahi1-KO mouse midbrains and Ahi1-knockdown cells. Our results further revealed that AHI1 decreases BMAL1/Rev-Erbα expression by interacting with and repressing retinoic acid receptor-related orphan receptor α, a nuclear receptor and transcriptional regulator of circadian genes. Of note, Bmal1 deficiency reversed the reduction in TH expression induced by Ahi1 deficiency. Moreover, microinfusion of the Rev-Erbα inhibitor SR8278 into the ventral midbrain of Ahi1-KO mice significantly increased TH expression in the ventral tegmental area and improved their depressive symptoms. These findings provide a mechanistic explanation for a link between AHI1-related behaviors and the circadian clock pathway, indicating an involvement of circadian regulatory proteins in AHI1-regulated mood and behavior.


Subject(s)
Circadian Clocks , Depression/genetics , Down-Regulation , Proto-Oncogene Proteins/genetics , Signal Transduction , Tyrosine 3-Monooxygenase/genetics , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Adaptor Proteins, Vesicular Transport , Affect , Animals , Depression/metabolism , Gene Deletion , Mesencephalon/physiology , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Proto-Oncogene Proteins/metabolism , Tyrosine 3-Monooxygenase/metabolism
11.
Acta Pharmacol Sin ; 39(4): 597-606, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29219947

ABSTRACT

Dendritic cell nuclear protein-1 (DCNP1) is a protein associated with major depression. In the brains of depression patients, DCNP1 is up-regulated. However, how DCNP1 participates in the pathogenesis of major depression remains unknown. In this study, we first transfected HEK293 cells with EGFP-DCNP1 and demonstrated that the full-length DCNP1 protein was localized in the nucleus, and RRK (the residues 117-119) composed its nuclear localization signal (NLS). An RRK-deletion form of DCNP1 (DCNP1ΔRRK) and truncated form (DCNP11-116), each lacking the RRK residues, did not show the specific nuclear localization like full-length DCNP1 in the cells. A rat glioma cell line C6 can synthesize melatonin, a hormone that plays important roles in both sleep and depression. We then revealed that transfection of C6 cells with full-length DCNP1 but not DCNP1ΔRRK or DCNP11-116 significantly decreased the levels of melatonin. Furthermore, overexpression of full-length DCNP1, but not DCNP1ΔRRK or DCNP11-116, in C6 cells significantly decreased both the mRNA and protein levels of N-acetyltransferase (NAT), a key enzyme in melatonin synthesis. Full-length DCNP1 but not DCNP1ΔRRK or DCNP11-116 was detected to interact with the Nat promoter and inhibited its activity through its E-box motif. Furthermore, full-length DCNP1 but not the mutants interacted with and repressed the transcriptional activity of BMAL1, a transcription factor that transactivates Nat through the E-box motif. In conclusion, we have shown that RRK (the residues 117-119) are the NLS responsible for DCNP1 nuclear localization. Nuclear DCNP1 represses NAT expression and melatonin biosynthesis by interacting with BMAL1 and repressing its transcriptional activity. Our study reveals a connection between the major depression candidate protein DCNP1, circadian system and melatonin biosynthesis, which may contribute to the pathogenesis of depression.


Subject(s)
ARNTL Transcription Factors/metabolism , Acetyltransferases/antagonists & inhibitors , Melatonin/biosynthesis , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , ARNTL Transcription Factors/genetics , Acetyltransferases/genetics , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Humans , Nuclear Localization Signals , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/metabolism , Rats , Repressor Proteins/genetics , Sequence Deletion , Transcription, Genetic
12.
Mol Neurodegener ; 7: 40, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22892098

ABSTRACT

BACKGROUND: Mutations or deletions in DJ-1/PARK7 gene are causative for recessive forms of early onset Parkinson's disease (PD). Wild-type DJ-1 has cytoprotective roles against cell death through multiple pathways. The most commonly studied mutant DJ-1(L166P) shifts its subcellular distribution to mitochondria and renders cells more susceptible to cell death under stress stimuli. We previously reported that wild-type DJ-1 binds to Bcl-XL and stabilizes it against ultraviolet B (UVB) irradiation-induced rapid degradation. However, the mechanisms by which mitochondrial DJ-1(L166P) promotes cell death under death stimuli are largely unknown. RESULTS: We show that DJ-1(L166P) is more prone to localize in mitochondria and it binds to Bcl-XL more strongly than wild-type DJ-1. In addition, UVB irradiation significantly promotes DJ-1(L166P) translocation to mitochondria and binding to Bcl-XL. DJ-1(L166P) but not wild-type DJ-1 dissociates Bax from Bcl-XL, thereby leading to Bax enrichment at outer mitochondrial membrane and promoting mitochondrial apoptosis pathway in response to UVB irradiation. CONCLUSION: Our findings suggest that wild-type DJ-1 protects cells and DJ-1(L166P) impairs cells by differentially regulating mitochondrial Bax/Bcl-XL functions.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Mitochondria/metabolism , Mutation , Oncogene Proteins/genetics , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , Cell Death/physiology , Gene Knockdown Techniques , HEK293 Cells , Humans , Immunohistochemistry , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/metabolism , Oncogene Proteins/metabolism , Protein Deglycase DJ-1 , bcl-2-Associated X Protein/genetics , bcl-X Protein/genetics
13.
Sci Signal ; 5(238): ra61, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22912494

ABSTRACT

Inflammation in Parkinson's disease is closely associated with disease pathogenesis. Mutations in Omi, which encodes the protease Omi, are linked to neurodegeneration and Parkinson's disease in humans and in mouse models. The severe neurodegeneration and neuroinflammation that occur in mnd2 (motor neuron degeneration 2) mice result from loss of the protease activity of Omi by the point mutation S276C; however, the substrates of Omi that induce neurodegeneration are unknown. We showed that Omi was required for the production of inflammatory molecules by microglia, which are the resident macrophages in the central nervous system. Omi suppressed the activation of the mitogen-activated protein kinases (MAPKs) extracellular signal-regulated kinase 1 and 2 (ERK1/2) by cleaving the upstream kinase MEK1 (mitogen-activated or extracellular signal-regulated protein kinase kinase 1). Knockdown of Omi in microglial cell lines led to activation of ERK1/2 and resulted in degradation of IκBα [α inhibitor of nuclear factor κB (NF-κB)], resulting in NF-κB activation and the expression of genes encoding inflammatory molecules, such as tumor necrosis factor-α and inducible nitric oxide synthase. The production of inflammatory molecules induced by the knockdown of Omi was blocked by the MEK1-specific inhibitor U0126. Furthermore, expression of the protease-deficient S276C Omi mutant in a microglial cell line had no effect on MEK1 cleavage or ERK1/2 activation. In the brains of mnd2 mice, we observed increased transcription of several genes encoding inflammatory molecules, as well as activation of astrocytes and microglia. Therefore, our study demonstrates that Omi is an intrinsic cellular factor that inhibits neuroinflammation.


Subject(s)
MAP Kinase Kinase 1/metabolism , Microglia/metabolism , Mitochondrial Proteins/metabolism , Neurodegenerative Diseases/metabolism , Serine Endopeptidases/metabolism , Animals , Blotting, Western , Butadienes/pharmacology , Cell Line , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , High-Temperature Requirement A Serine Peptidase 2 , Humans , I-kappa B Proteins/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , MAP Kinase Kinase 1/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/drug effects , Mitochondrial Proteins/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitriles/pharmacology , Point Mutation , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
J Biol Chem ; 286(40): 35308-17, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21852238

ABSTRACT

Parkinson disease (PD)- and cancer-associated protein, DJ-1, mediates cellular protection via many signaling pathways. Deletions or mutations in the DJ-1 gene are directly linked to autosomal recessive early-onset PD. DJ-1 has potential roles in mitochondria. Here, we show that DJ-1 increases its mitochondrial distribution in response to ultraviolet B (UVB) irradiation and binds to Bcl-X(L). The interactions between DJ-1 and Bcl-X(L) are oxidation-dependent. DJ-1(C106A), a mutant form of DJ-1 that is unable to be oxidized, binds Bcl-X(L) much less than DJ-1 does. Moreover, DJ-1 stabilizes Bcl-X(L) protein level by inhibiting its ubiquitination and degradation through ubiquitin proteasome system (UPS) in response to UVB irradiation. Furthermore, under UVB irradiation, knockdown of DJ-1 leads to increases of Bcl-X(L) ubiquitination and degradation upon UVB irradiation, thereby increasing mitochondrial Bax, caspase-3 activation and PARP cleavage. These data suggest that DJ-1 protects cells against UVB-induced cell death dependent on its oxidation and its association with mitochondrial Bcl-X(L).


Subject(s)
Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/metabolism , Oncogene Proteins/metabolism , bcl-X Protein/metabolism , Caspase 3/metabolism , Cell Line , Glutathione Transferase/metabolism , Humans , Mitochondria/metabolism , Neoplasms/metabolism , Parkinson Disease/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Protein Deglycase DJ-1 , Subcellular Fractions/metabolism , Ubiquitin/chemistry , Ultraviolet Rays
15.
Cancer Lett ; 297(1): 101-8, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20510502

ABSTRACT

Autophagy mediates cellular self-digestion to degrade cytoplasmic proteins and organelles and plays important roles in tumorigenesis. DJ-1 is an oncogene product in association with cancers and tumorigenesis. In this study, we show that knockdown of DJ-1 induces autophagy through activating JNK pathway to promote Beclin 1 transcription, whereas overexpression of DJ-1 inhibits these processes. Moreover, inhibition of JNK pathway by SP600125 blocks autophagy activation and p62 degradation induced by knockdown of DJ-1. Our findings suggest that DJ-1 regulates autophagy in a JNK-dependent manner. Thus, the involvement of DJ-1 in autophagy regulation may be involved in tumorigenesis.


Subject(s)
Autophagy , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Neoplasms/enzymology , Oncogene Proteins/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Anthracenes/pharmacology , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Beclin-1 , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Membrane Proteins/metabolism , Neoplasms/genetics , Neoplasms/pathology , Oncogene Proteins/genetics , Protein Deglycase DJ-1 , Protein Kinase Inhibitors/pharmacology , RNA Interference , Sequestosome-1 Protein , Signal Transduction/drug effects , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...