Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 95
1.
Neurobiol Dis ; : 106528, 2024 May 11.
Article En | MEDLINE | ID: mdl-38740348

BACKGROUND: Brain injury has been suggested as a risk factor for neurodegenerative diseases. Accordingly, defects in the brain's intrinsic capacity to repair injury may result in the accumulation of damage and a progressive loss of brain function. The G2019S (GS) mutation in LRRK2 (leucine rich repeat kinase 2) is the most prevalent genetic alteration in Parkinson's disease (PD). Here, we sought to investigate how this LRRK2-GS mutation affects repair of the injured brain. METHODS: Brain injury was induced by stereotaxic injection of ATP, a damage-associated molecular pattern (DAMP) component, into the striatum of wild-type (WT) and LRRK2-GS mice. Effects of the LRRK2-GS mutation on brain injury and the recovery from injury were examined by analyzing the molecular and cellular behavior of neurons, astrocytes, and monocytes. RESULTS: Damaged neurons express osteopontin (OPN), a factor associated with brain repair. Following ATP-induced damage, monocytes entered injured brains, phagocytosing damaged neurons and producing exosome-like vesicles (EVs) containing OPN through activation of the inflammasome and subsequent pyroptosis. Following EV production, neurons and astrocytes processes elongated towards injured cores. In LRRK2-GS mice, OPN expression and monocytic pyroptosis were decreased compared with that in WT mice, resulting in diminished release of OPN-containing EVs and attenuated elongation of neuron and astrocyte processes. In addition, exosomes prepared from injured LRRK2-GS brains induced neurite outgrowth less efficiently than those from injured WT brains. CONCLUSIONS: The LRRK2-GS mutation delays repair of injured brains through reduced expression of OPN and diminished release of OPN-containing EVs from monocytes. These findings suggest that the LRRK2-GS mutation may promote the development of PD by delaying the repair of brain injury.

2.
Nanoscale ; 16(2): 833-847, 2024 Jan 03.
Article En | MEDLINE | ID: mdl-38093712

Astrocytes are highly activated following brain injuries, and their activation influences neuronal survival. Additionally, SOX9 expression is known to increase in reactive astrocytes. However, the role of SOX9 in activated astrocytes following ischemic brain damage has not been clearly elucidated yet. Therefore, in the present study, we investigated the role of SOX9 in reactive astrocytes using a poly-lactic-co-glycolic acid (PLGA) nanoparticle plasmid delivery system in a photothrombotic stroke animal model. We designed PLGA nanoparticles to exclusively enhance SOX9 gene expression in glial fibrillary acidic protein (GFAP)-immunoreactive astrocytes. Our observations indicate that PLGA nanoparticles encapsulated with GFAP:SOX9:tdTOM reduce ischemia-induced neurological deficits and infarct volume through the prostaglandin D2 pathway. Thus, the astrocyte-targeting PLGA nanoparticle plasmid delivery system provides a potential opportunity for stroke treatment. Since the only effective treatment currently available is reinstating the blood supply, cell-specific gene therapy using PLGA nanoparticles will open a new therapeutic paradigm for brain injury patients in the future.


Brain Injuries , Nanoparticles , Stroke , Humans , Animals , Astrocytes/metabolism , Stroke/therapy , Stroke/genetics , Stroke/metabolism , Brain Injuries/metabolism , Peptides/pharmacology , Brain/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/pharmacology
3.
Microbes Infect ; 25(8): 105211, 2023.
Article En | MEDLINE | ID: mdl-37574181

Viral myocarditis is an inflammatory disease of the myocardium, often leads to cardiac dysfunction and death. PARKIN (PRKN) and PINK1, well known as Parkinson's disease-associated genes, have been reported to be involved in innate immunity and mitochondrial damage control. Therefore, we investigated the role of parkin and PINK1 in coxsackievirus B3 (CVB3)-induced viral myocarditis because the etiology of myocarditis is related to abnormal immune response to viral infection and mitochondrial damage. After viral infection, the survival was significantly lower and myocardial damage was more severe in parkin knockout (KO) and PINK1 KO mice compared to wild-type (WT) mice. Parkin KO and PINK1 KO showed defective immune cell recruitment and impaired production of antiviral cytokines such as interferon-gamma, allowing increased viral replication. In addition, parkin KO and PINK1 KO mice were more susceptible to CVB3-induced mitochondrial damage than WT mice, resulting in susceptibility to viral-induced cardiac damage. Finally, using publicly available RNA-seq data, we found that pathogenic mutants of the PRKN gene are more common in patients with dilated cardiomyopathy and myocarditis than in controls or the general population. This study will help elucidate the molecular mechanism of CVB3-induced viral myocarditis.


Coxsackievirus Infections , Myocarditis , Virus Diseases , Animals , Humans , Mice , Coxsackievirus Infections/genetics , Coxsackievirus Infections/pathology , Disease Models, Animal , Enterovirus B, Human/genetics , Mice, Knockout , Myocarditis/genetics , Myocarditis/pathology , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics
4.
Theranostics ; 12(13): 5856-5876, 2022.
Article En | MEDLINE | ID: mdl-35966584

Preconditioning nerve injury enhances axonal regeneration of dorsal root ganglia (DRG) neurons in part by driving pro-regenerative perineuronal macrophage activation. How these macrophages influence the neuronal capacity of axon regeneration remains elusive. We report that oncomodulin (ONCM) is produced from the regeneration-associated macrophages and strongly influences regeneration of DRG sensory axons. We also attempted to promote sensory axon regeneration by nanogel-mediated delivery of ONCM to DRGs. Methods:In vitro neuron-macrophage interaction model and preconditioning sciatic nerve injury were used to verify the necessity of ONCM in preconditioning injury-induced neurite outgrowth. We developed a nanogel-mediated delivery system in which electrostatic encapsulation of ONCM by a reducible epsilon-poly(L-lysine)-nanogel (REPL-NG) enabled a controlled release of ONCM. Results: Sciatic nerve injury upregulated ONCM in DRG macrophages. ONCM in macrophages was necessary to produce pro-regenerative macrophages in the in vitro model of neuron-macrophage interaction and played an essential role in preconditioning-induced neurite outgrowth. ONCM increased neurite outgrowth in cultured DRG neurons by activating a distinct gene set, particularly neuropeptide-related genes. Increasing extracellularly secreted ONCM in DRGs sufficiently enhanced the capacity of neurite outgrowth. Intraganglionic injection of REPL-NG/ONCM complex allowed sustained ONCM activity in DRG tissue and achieved a remarkable long-range regeneration of dorsal column sensory axons beyond spinal cord lesion. Conclusion: NG-mediated ONCM delivery could be exploited as a therapeutic strategy for promoting sensory axon regeneration following spinal cord injury.


Axons , Peripheral Nerve Injuries , Axons/physiology , Calcium-Binding Proteins , Humans , Macrophages/physiology , Nanogels , Nerve Regeneration/physiology , Peripheral Nerve Injuries/pathology , Spinal Cord
5.
FEBS J ; 289(19): 5914-5932, 2022 10.
Article En | MEDLINE | ID: mdl-35429212

Interleukin-6 (IL-6) is involved in many inflammatory diseases. IL-6 binds to membrane-bound IL-6 receptor α (IL-6Rα) (classic signaling) or soluble IL-6Rα (trans-signaling); this complex then associates with the signal-transducing membrane protein gp130. IL-6Rα and gp130 float on membrane (i.e., lipid) rafts; however, how membrane rafts regulate IL-6 signaling remains unclear. Here, we demonstrate that both IL-6 classic signaling and trans-signaling depend on membrane cholesterol, an essential raft component. Super-resolution fluorescence imaging using perfringolysin O D4 fragments that selectively bind to high cholesterol concentrations revealed that IL-6 and hyper-IL-6, a fusion protein of IL-6 and soluble IL-6Rα, induce the alteration of membrane rafts. IL-6 and hyper-IL-6 induced D4-positive raft (D4 raft) formation without affecting cholera toxin subunit B (CTB)-positive rafts (CTB rafts). Receptor clustering of IL-6Rα and gp130 and STAT3 phosphorylation occurred in D4 rafts. These results indicate that D4 rafts serve as platforms for the assembly of functional IL-6 receptor complexes. We found that Eps15 homology domain-containing protein 1 (EHD1) mediates the formation of functional IL-6 receptor complexes through D4 rafts. Overall, we uncover a novel regulatory mechanism of the EHD1-mediated alteration of membrane raft in IL-6 signaling.


Cholera Toxin , Interleukin-6 , Cholera Toxin/metabolism , Cholesterol/metabolism , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism
6.
Exp Neurol ; 348: 113922, 2022 02.
Article En | MEDLINE | ID: mdl-34780772

Patients with diabetes suffer more severe ischemic stroke. A combination of metformin and dipeptidyl peptide-4 inhibitors is commonly prescribed to treat diabetes. Therefore, we aimed to determine if pretreatment with a combination of metformin and evogliptin, a dipeptidyl peptidase-4 inhibitor, could reduce cerebral infarct volume in rats with streptozotocin-induced diabetes. After confirming diabetes induction, the rats were treated with vehicle, evogliptin, metformin, or evogliptin/metformin combination for 30 days. Then, stroke was induced by transient middle cerebral artery occlusion (tMCAO). Infarct volume, oxidative stress, levels of methylglyoxal-modified protein, glucagon-like peptide-1 receptor (GLP-1R), AMPK, and Akt/PI3K pathway-related proteins, and post-stroke pancreatic islet cell volume were evaluated. Compared to vehicle, only the co-administration group had significantly reduced infarct volume from the effects of tMCAO; the regimen also improved glycemic control, whereas the individual treatments did not. Co-administration also significantly reduced methylglyoxal-modified protein level in the core of the brain cortex, and the expression of 4-HNE and 8-OHdG was reduced. Co-administration increased p-Akt levels in the ischemic core and mitigated the suppression of Bcl-2 expression. Plasma GLP-1 and dipeptidyl peptidase-4 levels and brain GLP-1R expression remained unaltered. In the pancreas, islet cell damage was reduced by co-administration. These results reveal that metformin and evogliptin co-administration ameliorates cerebral infarction associated with prolonged glycemic control and pancreatic beta cell sparing. Other potential protective mechanisms may be upregulation of insulin receptor signaling or reduction of methylglyoxal-induced neurotoxicity. The combination of metformin and evogliptin should be tested further for its potential against focal cerebral ischemia in diabetes patients.


Cerebral Infarction/prevention & control , Diabetes Mellitus, Experimental/complications , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Piperazines/therapeutic use , Animals , Brain Chemistry , Cerebral Infarction/etiology , Cerebral Infarction/pathology , Cerebrovascular Circulation , Diabetes Mellitus, Experimental/diagnostic imaging , Diabetes Mellitus, Experimental/drug therapy , Drug Therapy, Combination , Infarction, Middle Cerebral Artery/etiology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/prevention & control , Insulin-Secreting Cells/pathology , Magnetic Resonance Imaging , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Stroke/diagnostic imaging , Stroke/drug therapy , Stroke/etiology
7.
Korean J Physiol Pharmacol ; 25(6): 565-574, 2021 Nov 01.
Article En | MEDLINE | ID: mdl-34697267

Astrocytes are activated in response to brain damage. Here, we found that expression of Kir4.1, a major potassium channel in astrocytes, is increased in activated astrocytes in the injured brain together with upregulation of the neural stem cell markers, Sox2 and Nestin. Expression of Kir4.1 was also increased together with that of Nestin and Sox2 in neurospheres formed from dissociated P7 mouse brains. Using the Kir4.1 blocker BaCl2 to determine whether Kir4.1 is involved in acquisition of stemness, we found that inhibition of Kir4.1 activity caused a concentration-dependent increase in sphere size and Sox2 levels, but had little effect on Nestin levels. Moreover, induction of differentiation of cultured neural stem cells by withdrawing epidermal growth factor and fibroblast growth factor from the culture medium caused a sharp initial increase in Kir4.1 expression followed by a decrease, whereas Sox2 and Nestin levels continuously decreased. Inhibition of Kir4.1 had no effect on expression levels of Sox2 or Nestin, or the astrocyte and neuron markers glial fibrillary acidic protein and ß-tubulin III, respectively. Taken together, these results indicate that Kir4.1 may control gain of stemness but not differentiation of stem cells.

8.
J Biomed Sci ; 28(1): 51, 2021 Jul 07.
Article En | MEDLINE | ID: mdl-34229656

BACKGROUND: Endoplasmic reticulum (ER) stress is a common feature of Parkinson's disease (PD), and several PD-related genes are responsible for ER dysfunction. Recent studies suggested LRRK2-G2019S, a pathogenic mutation in the PD-associated gene LRRK2, cause ER dysfunction, and could thereby contribute to the development of PD. It remains unclear, however, how mutant LRRK2 influence ER stress to control cellular outcome. In this study, we identified the mechanism by which LRRK2-G2019S accelerates ER stress and cell death in astrocytes. METHODS: To investigate changes in ER stress response genes, we treated LRRK2-wild type and LRRK2-G2019S astrocytes with tunicamycin, an ER stress-inducing agent, and performed gene expression profiling with microarrays. The XBP1 SUMOylation and PIAS1 ubiquitination were performed using immunoprecipitation assay. The effect of astrocyte to neuronal survival were assessed by astrocytes-neuron coculture and slice culture systems. To provide in vivo proof-of-concept of our approach, we measured ER stress response in mouse brain. RESULTS: Microarray gene expression profiling revealed that LRRK2-G2019S decreased signaling through XBP1, a key transcription factor of the ER stress response, while increasing the apoptotic ER stress response typified by PERK signaling. In LRRK2-G2019S astrocytes, the transcriptional activity of XBP1 was decreased by PIAS1-mediated SUMOylation. Intriguingly, LRRK2-GS stabilized PIAS1 by increasing the level of small heterodimer partner (SHP), a negative regulator of PIAS1 degradation, thereby promoting XBP1 SUMOylation. When SHP was depleted, XBP1 SUMOylation and cell death were reduced. In addition, we identified agents that can disrupt SHP-mediated XBP1 SUMOylation and may therefore have therapeutic activity in PD caused by the LRRK2-G2019S mutation. CONCLUSION: Our findings reveal a novel regulatory mechanism involving XBP1 in LRRK2-G2019S mutant astrocytes, and highlight the importance of the SHP/PIAS1/XBP1 axis in PD models. These findings provide important insight into the basis of the correlation between mutant LRRK2 and pathophysiological ER stress in PD, and suggest a plausible model that explains this connection.


Astrocytes/metabolism , Endoplasmic Reticulum Stress/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Receptors, Cytoplasmic and Nuclear/genetics , X-Box Binding Protein 1/genetics , Animals , Disease Models, Animal , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mice , Mutation , Parkinson Disease/physiopathology , Receptors, Cytoplasmic and Nuclear/metabolism , Sumoylation , X-Box Binding Protein 1/metabolism
9.
Glia ; 69(4): 1037-1052, 2021 04.
Article En | MEDLINE | ID: mdl-33300228

The brain has an intrinsic capacity to repair injury, but the specific mechanisms are largely unknown. In this study, we found that, despite their incipient death, damaged neurons play a key repair role with the help of monocytes infiltrated from blood. Monocytes phagocytosed damaged and/or dying neurons that expressed osteopontin (OPN), with possible subsequent activation of their inflammasome pathway, resulting in pyroptosis. During this process, monocytes released CD63-positive exosome-like vesicles containing OPN. Importantly, following the exosome-like vesicles, neuron and astrocyte processes elongated toward the injury core. In addition, exosomes prepared from the injured brain contained OPN, and enhanced neurite outgrowth of cultured neurons in an OPN-dependent manner. Thus, our results introduce the concept that neurons in the injured brain that are destined to die perceive the stressful condition and begin the regeneration processes through induction of OPN, ultimately executing the repair process with the help of monocytes recruited from the circulation.


Monocytes , Osteopontin , Brain/metabolism , Monocytes/metabolism , Neurons/metabolism , Osteopontin/metabolism , Phagocytosis
11.
Mol Brain ; 13(1): 103, 2020 07 22.
Article En | MEDLINE | ID: mdl-32698847

Brain injury causes astrocytes to become reactive (astrogliosis). In this study, we compared astrogliosis in acutely injured cortex and striatum of adult FVB/N mice induced by stereotaxic injection of ATP, a component of danger-associated molecular patterns (DAMPs). Interestingly, MR analysis showed that same amount of ATP induced smaller damage in the cortex than in the striatum. However, in histological analysis, thick and dense scar-like astrogliosis was found in the injured cortex near meninges within 2 wk., but not in other regions, including the striatum and even the cortex near the corpus callosum for up to 30 d. There was little regional difference in the number of Ki67(+)-proliferating astrocytes or mRNA expression of inflammatory cytokines. The most prominent difference between regions with and without scar-like astrogliosis was blood vessel formation. Blood vessels highly expressing collagen 1A1 formed densely near meninges, and astrocytes converged on them. In other regions, however, both blood vessels and astrocytes were relatively evenly distributed. Consistent with this, inhibition of blood vessel formation with the vascular endothelial growth factor (VEGF)-blocking antibody, Avastin, attenuated scar-like astrogliosis near meninges. These results indicate that region-specific astrogliosis occurs following brain injury, and that blood vessel formation plays a critical role in scar formation.


Blood Vessels/pathology , Cerebral Cortex/blood supply , Corpus Striatum/blood supply , Gliosis/pathology , Animals , Biomarkers/metabolism , Brain Injuries/pathology , Cell Proliferation , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Corpus Striatum/diagnostic imaging , Corpus Striatum/pathology , Inflammation/pathology , Leukocyte Common Antigens/metabolism , Magnetic Resonance Imaging , Male , Meninges/pathology , Mice , Organ Specificity , Time Factors
12.
EMBO Rep ; 21(7): e48950, 2020 07 03.
Article En | MEDLINE | ID: mdl-32372484

Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons located in the substantia nigra pars compacta and the presence of proteinaceous inclusions called Lewy bodies and Lewy neurites in numerous brain regions. Increasing evidence indicates that Lewy pathology progressively involves additional regions of the nervous system as the disease advances, and the prion-like propagation of α-synuclein (α-syn) pathology promotes PD progression. Accordingly, the modulation of α-syn transmission may be important for the development of disease-modifying therapies in patients with PD. Here, we demonstrate that α-syn fibrils induce c-src activation in neurons, which depends on the FcγRIIb-SHP-1/-2-c-src pathway and enhances signals for the uptake of α-syn into neurons. Blockade of c-src activation inhibits the uptake of α-syn and the formation of Lewy body-like inclusions. Furthermore, the blockade of c-src activation also inhibits the release of α-syn via activation of autophagy. The brain-permeable c-src inhibitor, saracatinib, efficiently reduces α-syn propagation into neighboring regions in an in vivo model system. These results suggest a new therapeutic target against progressive PD.


Parkinson Disease , alpha-Synuclein , Brain/metabolism , Dopaminergic Neurons/metabolism , Humans , Lewy Bodies/metabolism , Parkinson Disease/genetics , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
13.
Glia ; 68(10): 2086-2101, 2020 10.
Article En | MEDLINE | ID: mdl-32176388

Monocyte-derived macrophages play a role in the repair of the injured brain. We previously reported that a deficiency of the Parkinson's disease (PD)-associated gene DJ-1 delays repair of brain injury produced by stereotaxic injection of ATP, a component of damage-associated molecular patterns. Here, we show that a DJ-1 deficiency attenuates monocyte infiltration into the damaged brain owing to a decrease in C-C motif chemokine ligand 2 (CCL2) expression in astrocytes. Like DJ-1-knockout (KO) mice, CCL2 receptor (CCR2)-KO mice showed defects in monocyte infiltration and delayed recovery of brain injury, as determined by 9.4 T magnetic resonance imaging analysis and immunostaining for tyrosine hydroxylase and glial fibrillary acid protein. Notably, transcriptome analyses showed that genes related to regeneration and synapse formation were similarly downregulated in injured brains of DJ-1-KO and CCR2-KO mice compared with the injured wild-type brain. These results indicate that defective astrogliosis in DJ-1-KO mice is associated with decreased CCL2 expression and attenuated monocyte infiltration, resulting in delayed repair of brain injury. Thus, delayed repair of brain injury could contribute to the development of PD. MAIN POINTS: A DJ-1 deficiency attenuates infiltration of monocytes owing to a decrease in CCL2 expression in astrocytes, which in turn led to delay in repair of brain injury.


Astrocytes/metabolism , Brain Injuries/metabolism , Chemokine CCL2/biosynthesis , Monocytes/metabolism , Protein Deglycase DJ-1/deficiency , Animals , Astrocytes/pathology , Brain Injuries/genetics , Brain Injuries/pathology , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/pathology , Protein Deglycase DJ-1/genetics
14.
Exp Neurobiol ; 29(1): 70-79, 2020 Feb 29.
Article En | MEDLINE | ID: mdl-32122109

Many previous studies have shown reduced glucose uptake in the ischemic brain. In contrast, in a permanent unilateral common carotid artery occlusion (UCCAO) mouse model, our pilot experiments using 18F-fluorodeoxyglucose positron emission tomography (FDG PET) revealed that a subset of mice exhibited conspicuously high uptake of glucose in the ipsilateral hemisphere at 1 week post-occlusion (asymmetric group), whereas other mice showed symmetric uptake in both hemispheres (symmetric group). Thus, we aimed to understand the discrepancy between the two groups. Cerebral blood flow and histological/metabolic changes were analyzed using laser Doppler flowmetry and immunohistochemistry/Western blotting, respectively. Contrary to the increased glucose uptake observed in the ischemic cerebral hemisphere on FDG PET (p<0.001), cerebral blood flow tended to be lower in the asymmetric group than in the symmetric group (right to left ratio [%], 36.4±21.8 vs. 58.0±24.8, p=0.059). Neuronal death was observed only in the ischemic hemisphere of the asymmetric group. In contrast, astrocytes were more activated in the asymmetric group than in the symmetric group (p<0.05). Glucose transporter-1, and monocarboxylate transporter-1 were also upregulated in the asymmetric group, compared with the symmetric group (p<0.05, respectively). These results suggest that the increased FDG uptake was associated with relatively severe ischemia, and glucose transporter-1 upregulation and astrocyte activation. Glucose metabolism may thus be a compensatory mechanism in the moderately severe ischemic brain.

15.
Acta Neuropathol Commun ; 7(1): 149, 2019 09 17.
Article En | MEDLINE | ID: mdl-31530290

Multifunctional signal transducer and activator of transcription (STAT) proteins play important roles in cancer. Here, we have shown that STAT6 is epigenetically silenced in some cases of malignant glioblastoma, which facilitates cancer cell survival in a hypoxic microenvironment. This downregulation results from hypermethylation of CpG islands within the STAT6 promoter by DNA methyltransferases. STAT6 interacts with Rheb under hypoxia and inhibits mTOR/S6K/S6 signaling, in turn, inducing increased HIF-1α translation. STAT6 silencing and consequent tumor-promoting effects are additionally observed in glioma stem-like cells (GSC). Despite recent advances in cancer treatment, survival rates have shown little improvement. This is particularly true in the case of glioma, where multimodal treatment and precision medicine is needed. Our study supports the application of epigenetic restoration of STAT6 with the aid of DNA methyltransferase inhibitors, such as 5-aza-2-deoxycytidine, for treatment of STAT6-silenced gliomas.


Brain Neoplasms/metabolism , Brain/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , STAT6 Transcription Factor/metabolism , Cell Line, Tumor , Cell Survival , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , Down-Regulation , Humans , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Hypoxia
16.
Acta Neuropathol Commun ; 7(1): 68, 2019 05 02.
Article En | MEDLINE | ID: mdl-31046837

Accumulating evidence indicates that endoplasmic reticulum (ER) stress is a common feature of Parkinson's disease (PD) and further suggests that several PD-related genes are responsible for ER dysfunction. However, the underlying mechanisms are largely unknown. Here, we defined the mechanism by which LRRK2-G2019S (LRRK2-GS), a pathogenic mutation in the PD-associated gene LRRK2, accelerates ER stress and cell death. Treatment of cells with α-synuclein increased the expression of ER stress proteins and subsequent cell death in LRRK2-GS astrocytes. Intriguingly, we found that LRRK2-GS localizes to the ER membrane, where it interacts with sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) and suppress its activity by preventing displacement of phospholamban (PLN). LRRK2-GS-mediated SERCA malfunction leads to ER Ca2+ depletion, which induces the formation of mitochondria-ER contacts and subsequent Ca2+ overload in mitochondria, ultimately resulting in mitochondrial dysfunction. Collectively, our data suggest that, in astrocytes, LRRK2-GS impairs ER Ca2+ homeostasis, which determines cell survival, and as a result, could contribute to the development of PD.


Astrocytes/metabolism , Endoplasmic Reticulum Stress/physiology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Parkinson Disease/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Apoptosis , Cells, Cultured , Cerebral Cortex/metabolism , Disease Models, Animal , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mice, Transgenic , Mutation , Neurons/metabolism
17.
Neurobiol Dis ; 127: 482-491, 2019 07.
Article En | MEDLINE | ID: mdl-30954702

Dysfunctional regulation of inflammation may contribute to the progression of neurodegenerative diseases. The results of this study revealed that DJ-1, a Parkinson's disease (PD) gene, regulated expression of prostaglandin D2 synthase (PTGDS) and production of prostaglandin D2 (PGD2), by which DJ-1 enhanced anti-inflammatory function of astrocytes. In injured DJ-1 knockout (KO) brain, expression of tumor necrosis factor-alpha (TNF-α) was more increased, but that of anti-inflammatory heme oxygenase-1 (HO-1) was less increased compared with that in injured wild-type (WT) brain. Similarly, astrocyte-conditioned media (ACM) prepared from DJ-1-KO astrocytes less induced HO-1 expression and less inhibited expression of inflammatory mediators in microglia. With respect to the underlying mechanism, we found that PTGDS that induced expression of HO-1 was lower in DJ-1 KO astrocytes and brains compared with their WT counterparts. In addition, PTGDS levels increased in the injured brain of WT mice, but barely in that of KO mice. We also found that DJ-1 regulated PTGDS expression through Sox9. Thus, Sox9 siRNAs reduced PTGDS expression in WT astrocytes, and Sox9 overexpression rescued PTGDS expression in DJ-1 KO astrocytes. In agreement with these results, ACM from Sox9 siRNA-treated astrocytes and that from Sox9-overexpression astrocytes exerted opposite effects on HO-1 expression and anti-inflammation. These findings suggest that DJ-1 positively regulates anti-inflammatory functions of astrocytes, and that DJ-1 dysfunction contributes to the excessive inflammatory response in PD development.


Astrocytes/metabolism , Brain/metabolism , Gene Expression Regulation , Inflammation/genetics , Intramolecular Oxidoreductases/genetics , Lipocalins/genetics , Protein Deglycase DJ-1/genetics , Animals , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Inflammation/metabolism , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Deglycase DJ-1/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
Neurosci Lett ; 685: 144-149, 2018 10 15.
Article En | MEDLINE | ID: mdl-30171910

In the injured brain, astrocytes become activated and increase GFAP expression; a phenomenon termed as astrogliosis. Previously, we have reported that DJ-1, a Parkinson's disease gene, positively regulates astrogliosis in the injured brain. Moreover, STAT3 is known to play a key role in the regulation of astrogliosis. The purpose of this study was to evaluate whether DJ-1 regulates astrogliosis, via STAT3 activation. The results of this study reveal that DJ-1 is a positive regulator of STAT3 activation, the most important mediator of astrogliosis. In brain slice cultures obtained from wild-type (WT) mice, STAT3 was activated, as demonstrated by its phosphorylation to pSTAT3, within 3 h, and GFAP expression was increased within 2 days after slice preparation. Interestingly, levels of pSTAT3 and GFAP in DJ-1 knockout brain slice were attenuated compared with that of WT. STAT3 inhibitors, DPP (5 µM) and S3I-201 (50 µM), reduced expression of BDNF, GDNF, and GFAP. However, the STAT3 inhibitors did not affect mRNA levels of Sox9, another important regulator of astrogliosis. Additionally, Sox9 siRNAs did not affect STAT3 phosphorylation. These results indicate that STAT3 and Sox9 may be the independent targets of DJ-1.


Gliosis/genetics , Parkinson Disease/genetics , Protein Deglycase DJ-1/genetics , Protein Deglycase DJ-1/metabolism , STAT3 Transcription Factor/metabolism , Animals , Astrocytes/metabolism , Glial Fibrillary Acidic Protein/metabolism , Gliosis/metabolism , Mice, Knockout , Parkinson Disease/metabolism , Phosphorylation , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , Signal Transduction/physiology
19.
Exp Neurobiol ; 27(2): 77-87, 2018 Apr.
Article En | MEDLINE | ID: mdl-29731673

Astrocytes and microglia support well-being and well-function of the brain through diverse functions in both intact and injured brain. For example, astrocytes maintain homeostasis of microenvironment of the brain through up-taking ions and neurotransmitters, and provide growth factors and metabolites for neurons, etc. Microglia keep surveying surroundings, and remove abnormal synapses or respond to injury by isolating injury sites and expressing inflammatory cytokines. Therefore, their loss and/or functional alteration may be directly linked to brain diseases. Since Parkinson's disease (PD)-related genes are expressed in astrocytes and microglia, mutations of these genes may alter the functions of these cells, thereby contributing to disease onset and progression. Here, we review the roles of astrocytes and microglia in intact and injured brain, and discuss how PD genes regulate their functions.

20.
Proc Natl Acad Sci U S A ; 115(7): 1629-1634, 2018 02 13.
Article En | MEDLINE | ID: mdl-29386384

Mutations in DJ-1 (PARK7) are a known cause of early-onset autosomal recessive Parkinson's disease (PD). Accumulating evidence indicates that abnormalities of synaptic vesicle trafficking underlie the pathophysiological mechanism of PD. In the present study, we explored whether DJ-1 is involved in CNS synaptic function. DJ-1 deficiency impaired synaptic vesicle endocytosis and reavailability without inducing structural alterations in synapses. Familial mutants of DJ-1 (M26I, E64D, and L166P) were unable to rescue defective endocytosis of synaptic vesicles, whereas WT DJ-1 expression completely restored endocytic function in DJ-1 KO neurons. The defective synaptic endocytosis shown in DJ-1 KO neurons may be attributable to alterations in membrane cholesterol level. Thus, DJ-1 appears essential for synaptic vesicle endocytosis and reavailability, and impairment of this function by familial mutants of DJ-1 may be related to the pathogenesis of PD.


Endocytosis/physiology , Nerve Endings/pathology , Protein Deglycase DJ-1/physiology , Synapses/pathology , Synaptic Vesicles/pathology , Animals , Cells, Cultured , Mice , Mice, Knockout , Mutation , Nerve Endings/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism
...