Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters










Publication year range
1.
iScience ; 25(9): 104923, 2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36065189

ABSTRACT

Although it is reported that mitochondria-localized nuclear transcription factors (TFs) regulate mitochondrial processes such as apoptosis and mitochondrial transcription/respiration, the functions and mechanisms of mitochondrial dynamics regulated by mitochondria-localized nuclear TFs are yet to be fully characterized. Here, we identify STAT6 as a mitochondrial protein that is localized in the outer membrane of mitochondria (OMM). STAT6 in OMM inhibits mitochondrial fusion by blocking MFN2 dimerization. This implies that STAT6 has a critical role in mitochondrial dynamics. Moreover, mitochondrial accumulation of STAT6 in response to hypoxic conditions reveals that STAT6 is a regulator of mitochondrial processes including fusion/fission mechanisms.

2.
FEBS J ; 289(19): 5914-5932, 2022 10.
Article in English | MEDLINE | ID: mdl-35429212

ABSTRACT

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.


Subject(s)
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
3.
Free Radic Biol Med ; 179: 252-265, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34808332

ABSTRACT

Acute microglial activation plays an important role in neuroprotection. However, dysregulated, prolonged microgliosis exacerbates neurodegeneration through excessive release of pro-inflammatory cytokines and cytotoxic factors. Interferon-gamma (IFN-γ), an inflammatory cytokine, exacerbates the detrimental microglial response. Although various anti-inflammatory drugs have been evaluated as interventions for microglia-mediated neuroinflammation, no anti-inflammatories are in clinical use for microgliosis. The present study evaluated the anti-inflammatory mechanisms of oxysterols, blood brain barrier (BBB) penetrable bioactive lipids, revealing that this intervention suppresses neuroinflammation by disrupting membrane lipid raft formation and caveolae-mediated endosomal IFN-γ signaling. We find that 25-hydroxycholesterol (25-HC) rapidly repressed IFN-γ receptor trafficking to lipid rafts in microglia by disrupting raft formation, thereby suppressing microglial inflammatory response. IFN-γ treatment upregulated expression of Cav-1, a major component of caveolae, and IFN-γ signaling was sustained through Cav-1+ signaling endosomes. 25-HC repressed IFN-γ induction of Cav-1 expression in microglia, and subsequently suppressed the chronic inflammatory response. Taken together, these findings demonstrated that 25-HC effectively regulate the inflammatory status of microglia by mediating the formation of rafts and caveolae-dependent signaling endosomes. Given the important roles of IFN-γ and microglia in the pathology of neurodegenerative brain diseases, a novel anti-inflammatory mechanism of 25-HC that is not receptor-dependent, but rather is related to the regulation of membrane rafts and caveolae, suggests a new therapeutic target for inflammatory neurodegenerations.


Subject(s)
Hydroxycholesterols/pharmacology , Interferon-gamma , Membrane Microdomains , Microglia , Animals , Caveolins , Endosomes , Inflammation , Interferon-gamma/genetics , Mice, Inbred C57BL , Neuroinflammatory Diseases
4.
Korean J Physiol Pharmacol ; 25(6): 565-574, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34697267

ABSTRACT

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.

5.
J Biomed Sci ; 28(1): 51, 2021 Jul 07.
Article in English | MEDLINE | ID: mdl-34229656

ABSTRACT

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.


Subject(s)
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
6.
Glia ; 69(4): 1037-1052, 2021 04.
Article in English | MEDLINE | ID: mdl-33300228

ABSTRACT

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.


Subject(s)
Monocytes , Osteopontin , Brain/metabolism , Monocytes/metabolism , Neurons/metabolism , Osteopontin/metabolism , Phagocytosis
7.
Mol Brain ; 13(1): 103, 2020 07 22.
Article in English | MEDLINE | ID: mdl-32698847

ABSTRACT

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.


Subject(s)
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
9.
EMBO Rep ; 21(7): e48950, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32372484

ABSTRACT

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.


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

ABSTRACT

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.


Subject(s)
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
11.
Acta Neuropathol Commun ; 7(1): 149, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31530290

ABSTRACT

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.


Subject(s)
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
12.
Acta Neuropathol Commun ; 7(1): 68, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31046837

ABSTRACT

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.


Subject(s)
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
13.
Neurobiol Dis ; 127: 482-491, 2019 07.
Article in English | MEDLINE | ID: mdl-30954702

ABSTRACT

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.


Subject(s)
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
14.
J Cell Mol Med ; 22(9): 4117-4129, 2018 09.
Article in English | MEDLINE | ID: mdl-29851245

ABSTRACT

Phosphatidylinositol 4-phosphate 5-kinase (PIP5K) family members generate phosphatidylinositol 4,5-bisphosphate (PIP2), a critical lipid regulator of diverse physiological processes. The PIP5K-dependent PIP2 generation can also act upstream of the oncogenic phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Many studies have demonstrated various mechanisms of spatiotemporal regulation of PIP5K catalytic activity. However, there are few studies on regulation of PIP5K protein stability. Here, we examined potential regulation of PIP5Kα, a PIP5K isoform, via ubiquitin-proteasome system, and its implication for breast cancer. Our results showed that the ubiquitin ligase NEDD4 (neural precursor cell expressed, developmentally down-regulated gene 4) mediated ubiquitination and proteasomal degradation of PIP5Kα, consequently reducing plasma membrane PIP2 level. NEDD4 interacted with the C-terminal region and ubiquitinated the N-terminal lysine 88 in PIP5Kα. In addition, PIP5Kα gene disruption inhibited epidermal growth factor (EGF)-induced Akt activation and caused significant proliferation defect in breast cancer cells. Notably, PIP5Kα K88R mutant that was resistant to NEDD4-mediated ubiquitination and degradation showed more potentiating effects on Akt activation by EGF and cell proliferation than wild-type PIP5Kα. Collectively, these results suggest that PIP5Kα is a novel degradative substrate of NEDD4 and that the PIP5Kα-dependent PIP2 pool contributing to breast cancer cell proliferation through PI3K/Akt activation is negatively controlled by NEDD4.


Subject(s)
Cell Membrane/metabolism , Gene Expression Regulation, Neoplastic , Nedd4 Ubiquitin Protein Ligases/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proto-Oncogene Proteins c-akt/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CRISPR-Cas Systems , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Proliferation , Epidermal Growth Factor/pharmacology , Female , Gene Editing , Humans , Mutation , Nedd4 Ubiquitin Protein Ligases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Ubiquitination/drug effects
15.
Exp Neurobiol ; 27(2): 77-87, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29731673

ABSTRACT

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.

16.
Proc Natl Acad Sci U S A ; 115(7): 1629-1634, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29386384

ABSTRACT

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.


Subject(s)
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
17.
Cell Rep ; 22(1): 136-148, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29298416

ABSTRACT

Recent evidence of prion-like propagation of α-synuclein (α-syn) into neighboring neurons set up a paradigm to elucidate the mechanism of progression of Parkinson's disease (PD) and to develop therapeutic strategies. Here, we show that FcγRIIB expressed in neurons functions as a receptor for α-syn fibrils and mediates cell-to-cell transmission of α-syn. SHP-1 and 2 are activated downstream by α-syn fibrils through FcγRIIB and play an important role in cell-to-cell transmission of α-syn. Also, taking advantage of a co-culture system, we show that cell-to-cell transmission of α-syn induces intracellular Lewy body-like inclusion body formation and that the FcγRIIB/SHP-1/2 signaling pathway is involved in it. Therefore, the FcγRIIB-SHP-1/-2 signaling pathway may be a therapeutic target for the progression of PD. The in vitro system is an efficient tool for further high-throughput screening that can be used for developing a therapeutic intervention in PD.


Subject(s)
Neurons/metabolism , Parkinson Disease/metabolism , Prions , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, IgG/metabolism , Signal Transduction , alpha-Synuclein/metabolism , Cell Line , Humans , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Transport/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Receptors, IgG/genetics , alpha-Synuclein/genetics
18.
Glia ; 66(2): 445-458, 2018 02.
Article in English | MEDLINE | ID: mdl-29105838

ABSTRACT

Defects in repair of damaged brain accumulate injury and contribute to slow-developing neurodegeneration. Here, we report that a deficiency of DJ-1, a Parkinson's disease (PD) gene, delays repair of brain injury due to destabilization of Sox9, a positive regulator of astrogliosis. Stereotaxic injection of ATP into the brain striatum produces similar size of acute injury in wild-type and DJ-1-knockout (KO) mice. However, recovery of the injury is delayed in KO mice, which is confirmed by 9.4T magnetic resonance imaging and tyrosine hydroxylase immunostaining. DJ-1 regulates neurite outgrowth from damaged neurons in a non-cell autonomous manner. In DJ-1 KO brains and astrocytes, Sox9 protein levels are decreased due to enhanced ubiquitination, resulting in defects in astrogliosis and glial cell-derived neurotrophic factor/ brain-derived neurotrophic factor expression in injured brain and astrocytes. These results indicate that DJ-1 deficiency causes defects in astrocyte-mediated repair of brain damage, which may contribute to the development of PD.


Subject(s)
Astrocytes/metabolism , Brain Injuries/metabolism , Gliosis/metabolism , Protein Deglycase DJ-1/deficiency , SOX9 Transcription Factor/metabolism , Animals , Astrocytes/pathology , Brain Injuries/genetics , Brain Injuries/pathology , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Gliosis/genetics , Gliosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Deglycase DJ-1/genetics , Protein Stability , SOX9 Transcription Factor/genetics
19.
Cell Signal ; 38: 159-170, 2017 10.
Article in English | MEDLINE | ID: mdl-28711717

ABSTRACT

Phosphatidylinositol 4,5-bisphosphate (PIP2) is an important lipid regulator of membrane signaling and remodeling processes. Accumulating evidence indicates a link between PIP2 metabolism and Toll-like receptor (TLR) signaling, a key transducer of immune responses such as inflammation, phagocytosis, and autophagy. Microglia are immune effector cells that serve as macrophages in the brain. Here, we examined the potential role of phosphatidylinositol 4-phosphate 5-kinase α (PIP5Kα), a PIP2-producing enzyme, in TLR2 signaling in microglial cells. Treatment of BV2 microglial cells with lipoteichoic acid (LTA), a TLR2 agonist, increased PIP5Kα expression in BV2 and primary microglial cells, but not in primary cultures from TLR2-deficient mice. PIP5Kα knockdown of BV2 cells with shRNA significantly suppressed LTA-induced activation of TLR2 downstream signaling, including the production of proinflammatory cytokines and phosphorylation of NF-κB, JNK, and p38 MAP kinase. Such suppression was reversed by complementation of PIP5Kα. PIP5Kα knockdown lowered PIP2 levels and impaired LTA-induced plasma membrane targeting of TIRAP, a PIP2-dependent adaptor required for TLR2 activation. Besides, PIP5Kα knockdown inhibited phagocytic uptake of E. coli particles and autophagy-related vesicle formation triggered by LTA. Taken together, these results support that PIP5Kα can positively mediate TLR2-associated immune responses through PIP2 production in microglial cells.


Subject(s)
Immunity/drug effects , Microglia/enzymology , Microglia/immunology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Toll-Like Receptor 2/metabolism , Actins/metabolism , Animals , Autophagy/drug effects , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Down-Regulation/drug effects , Gene Knockdown Techniques , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides , Mice, Knockout , Microglia/drug effects , Microglia/metabolism , Phagocytosis/drug effects , Phosphatidylinositol Phosphates/metabolism , Polymerization/drug effects , Protein Transport/drug effects , Signal Transduction/drug effects , Teichoic Acids , Up-Regulation/drug effects
20.
Exp Neurobiol ; 25(5): 269-276, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27790061

ABSTRACT

Mutation of leucine-rich repeat kinase 2 (LRRK2) causes an autosomal dominant and late-onset familial Parkinson's disease (PD). Recently, we reported that LRRK2 directly binds to and phosphorylates the threonine 474 (T474)-containing Thr-X-Arg(Lys) (TXR) motif of focal adhesion kinase (FAK), thereby inhibiting the phosphorylation of FAK at tyrosine (Y) 397 residue (pY397-FAK), which is a marker of its activation. Mechanistically, however, it remained unclear how T474-FAK phosphorylation suppressed FAK activation. Here, we report that T474-FAK phosphorylation could inhibit FAK activation via at least two different mechanisms. First, T474 phosphorylation appears to induce a conformational change of FAK, enabling its N-terminal FERM domain to autoinhibit Y397 phosphorylation. This is supported by the observation that the levels of pY397-FAK were increased by deletion of the FERM domain and/or mutation of the FERM domain to prevent its interaction with the kinase domain of FAK. Second, pT474-FAK appears to recruit SHP-2, which is a phosphatase responsible for dephosphorylating pY397-FAK. We found that mutation of T474 into glutamate (T474E-FAK) to mimic phosphorylation induced more strong interaction with SHP-2 than WT-FAK, and that pharmacological inhibition of SHP-2 with NSC-87877 rescued the level of pY397 in HEK293T cells. These results collectively show that LRRK2 suppresses FAK activation through diverse mechanisms that include the promotion of autoinhibition and/or the recruitment of phosphatases, such as SHP-2.

SELECTION OF CITATIONS
SEARCH DETAIL
...