Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(2): eadi8287, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38198537

ABSTRACT

Parkinson's disease (PD) is characterized pathologically by the loss of dopaminergic (DA) neurons in the substantia nigra (SN). Whether cell types beyond DA neurons in the SN show vulnerability in PD remains unclear. Through transcriptomic profiling of 315,867 high-quality single nuclei in the SN from individuals with and without PD, we identified cell clusters representing various neuron types, glia, endothelial cells, pericytes, fibroblasts, and T cells and investigated cell type-dependent alterations in gene expression in PD. Notably, a unique neuron cluster marked by the expression of RIT2, a PD risk gene, also displayed vulnerability in PD. We validated RIT2-enriched neurons in midbrain organoids and the mouse SN. Our results demonstrated distinct transcriptomic signatures of the RIT2-enriched neurons in the human SN and implicated reduced RIT2 expression in the pathogenesis of PD. Our study sheds light on the diversity of cell types, including DA neurons, in the SN and the complexity of molecular and cellular changes associated with PD pathogenesis.


Subject(s)
Endothelial Cells , Parkinson Disease , Humans , Animals , Mice , Parkinson Disease/genetics , Substantia Nigra , Dopaminergic Neurons , Neuroglia
2.
Nat Cell Biol ; 25(7): 963-974, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37231161

ABSTRACT

Dysfunctional autophagy has been implicated in the pathogenesis of Alzheimer's disease (AD). Previous evidence suggested disruptions of multiple stages of the autophagy-lysosomal pathway in affected neurons. However, whether and how deregulated autophagy in microglia, a cell type with an important link to AD, contributes to AD progression remains elusive. Here we report that autophagy is activated in microglia, particularly of disease-associated microglia surrounding amyloid plaques in AD mouse models. Inhibition of microglial autophagy causes disengagement of microglia from amyloid plaques, suppression of disease-associated microglia, and aggravation of neuropathology in AD mice. Mechanistically, autophagy deficiency promotes senescence-associated microglia as evidenced by reduced proliferation, increased Cdkn1a/p21Cip1, dystrophic morphologies and senescence-associated secretory phenotype. Pharmacological treatment removes autophagy-deficient senescent microglia and alleviates neuropathology in AD mice. Our study demonstrates the protective role of microglial autophagy in regulating the homeostasis of amyloid plaques and preventing senescence; removal of senescent microglia is a promising therapeutic strategy.


Subject(s)
Alzheimer Disease , Microglia , Mice , Animals , Microglia/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Autophagy/physiology , Neurons/metabolism , Mice, Transgenic , Disease Models, Animal
3.
Sci Adv ; 8(43): eabn1298, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36288297

ABSTRACT

Autophagy clears protein aggregates, damaged cellular organelles, and pathogens through the lysosome. Although autophagy is highly conserved across all cell types, its activity in each cell is specifically adapted to carry out distinct physiological functions. The role of autophagy in neurons has been well characterized; however, in glial cells, its function remains largely unknown. Microglia are brain-resident macrophages that survey the brain to remove injured neurons, excessive synapses, protein aggregates, and infectious agents. Current studies have demonstrated that dysfunctional microglia contribute to neurodegenerative diseases. In Alzheimer's disease animal models, microglia play a critical role in regulating amyloid plaque formation and neurotoxicity. However, how microglia are involved in Parkinson's disease (PD) remains poorly understood. Propagation of aggregated α-synuclein via cell-to-cell transmission and neuroinflammation have emerged as important mechanisms underlying neuropathologies in PD. Here, we review converging evidence that microglial autophagy maintains α-synuclein homeostasis, regulates neuroinflammation, and confers neuroprotection in PD experimental models.


Subject(s)
Parkinson Disease , alpha-Synuclein , Animals , Microglia/metabolism , Inflammasomes/metabolism , Protein Aggregates , Parkinson Disease/metabolism , Autophagy , Homeostasis
4.
Proc Natl Acad Sci U S A ; 118(14)2021 04 06.
Article in English | MEDLINE | ID: mdl-33785595

ABSTRACT

Autophagy is a catabolic pathway that provides self-nourishment and maintenance of cellular homeostasis. Autophagy is a fundamental cell protection pathway through metabolic recycling of various intracellular cargos and supplying the breakdown products. Here, we report an autophagy function in governing cell protection during cellular response to energy crisis through cell metabolic rewiring. We observe a role of selective type of autophagy in direct activation of cyclic AMP protein kinase A (PKA) and rejuvenation of mitochondrial function. Mechanistically, autophagy selectively degrades the inhibitory subunit RI of PKA holoenzyme through A-kinase-anchoring protein (AKAP) 11. AKAP11 acts as an autophagy receptor that recruits RI to autophagosomes via LC3. Glucose starvation induces AKAP11-dependent degradation of RI, resulting in PKA activation that potentiates PKA-cAMP response element-binding signaling, mitochondria respiration, and ATP production in accordance with mitochondrial elongation. AKAP11 deficiency inhibits PKA activation and impairs cell survival upon glucose starvation. Our results thus expand the view of autophagy cytoprotection mechanism by demonstrating selective autophagy in RI degradation and PKA activation that fuels the mitochondrial metabolism and confers cell resistance to glucose deprivation implicated in tumor growth.


Subject(s)
A Kinase Anchor Proteins/metabolism , Autophagy , Mitochondria/metabolism , Neoplasms/metabolism , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Mice
5.
Front Neurosci ; 14: 674, 2020.
Article in English | MEDLINE | ID: mdl-32765209

ABSTRACT

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most frequent cause of familial Parkinson's disease (PD). Several genetic manipulations of the LRRK2 gene have been developed in animal models such as rodents, Drosophila, Caenorhabditis elegans, and zebrafish. These models can help us further understand the biological function and derive potential pathological mechanisms for LRRK2. Here we discuss common phenotypic themes found in LRRK2-associated PD animal models, highlight several issues that should be addressed in future models, and discuss emerging areas to guide their future development.

6.
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
7.
Hum Mol Genet ; 29(14): 2300-2312, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32356558

ABSTRACT

Synaptojanin1 (synj1) is a phosphoinositide phosphatase with dual SAC1 and 5'-phosphatase enzymatic activities in regulating phospholipid signaling. The brain-enriched isoform has been shown to participate in synaptic vesicle (SV) recycling. More recently, recessive human mutations were identified in the two phosphatase domains of SYNJ1, including R258Q, R459P and R839C, which are linked to rare forms of early-onset Parkinsonism. We now demonstrate that Synj1 heterozygous deletion (Synj1+/-), which is associated with an impaired 5'-phosphatase activity, also leads to Parkinson's disease (PD)-like pathologies in mice. We report that male Synj1+/- mice display age-dependent motor function abnormalities as well as alpha-synuclein accumulation, impaired autophagy and dopaminergic terminal degeneration. Synj1+/- mice contain elevated 5'-phosphatase substrate, PI(4,5)P2, particularly in the midbrain neurons. Moreover, pharmacological elevation of membrane PI(4,5)P2 in cultured neurons impairs SV endocytosis, specifically in midbrain neurons, and further exacerbates SV trafficking defects in Synj1+/- midbrain neurons. We demonstrate down-regulation of SYNJ1 transcript in a subset of sporadic PD brains, implicating a potential role of Synj1 deficiency in the decline of dopaminergic function during aging.


Subject(s)
Nerve Tissue Proteins/genetics , Parkinson Disease/genetics , Phosphoric Monoester Hydrolases/genetics , alpha-Synuclein/genetics , Animals , Autophagy/genetics , Disease Models, Animal , Dopamine/genetics , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Endocytosis/genetics , Haploinsufficiency/genetics , Humans , Mesencephalon/metabolism , Mesencephalon/pathology , Mice , Parkinson Disease/pathology , Sequence Deletion/genetics
8.
Autophagy ; 16(9): 1718-1720, 2020 09.
Article in English | MEDLINE | ID: mdl-32453651

ABSTRACT

SNCA/α-synuclein is a major component in the Lewy body (LB), a pathological hallmark of Parkinson disease (PD) and dementia with Lewy body (DLB), collectively known as synucleinopathies. SNCA/α-synuclein can be secreted from neurons and transmitted to neighboring cells including neurons and glia, which underlie the spreading of LB pathology as described by Braak and colleagues. We recently have investigated the mechanism and significance for microglia, a prototypic phagocyte in the brain, in engulfing and controlling SNCA/α-synuclein homeostasis in the brain. Using microglia-specific autophagy-deficient mice, we demonstrated that microglia ingest and degrade neuron-released SNCA/α-synuclein through SQSTM1/p62-mediated selective autophagy both in vivo and in vitro. This process requires the presence of TLR4 (toll like receptor 4), which interacts with SNCA/α-synuclein to induce the transcriptional upregulation of Sqstm1/p62 through the NFKB/NF-κB pathway. We term the selective autophagy of SNCA/α-synuclein as "synucleinphagy". We showed that the disruption of microglial autophagy causes accumulation of misfolded SNCA/α-synuclein and loss of dopaminergic neurons, two hallmarks of PD. Hence, our study reveals a neuroprotective role of microglia through an autophagy-mediated "community cleanup program".


Subject(s)
Autophagy , Neuroprotection , alpha-Synuclein/metabolism , Animals , Humans , Mice, Transgenic , Microglia/metabolism , Models, Biological , Proteolysis , Sequestosome-1 Protein/metabolism
9.
Nat Commun ; 11(1): 1386, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32170061

ABSTRACT

Microglia maintain brain homeostasis by removing neuron-derived components such as myelin and cell debris. The evidence linking microglia to neurodegenerative diseases is growing; however, the precise mechanisms remain poorly understood. Herein, we report a neuroprotective role for microglia in the clearance of neuron-released α-synuclein. Neuronal α-synuclein activates microglia, which in turn engulf α-synuclein into autophagosomes for degradation via selective autophagy (termed synucleinphagy). Synucleinphagy requires the presence of microglial Toll-like receptor 4 (TLR4), which induces transcriptional upregulation of p62/SQSTM1 through the NF-κB signaling pathway. Induction of p62, an autophagy receptor, is necessary for the formation of α-synuclein/ubiquitin-positive puncta that are degraded by autophagy. Finally, disruption of microglial autophagy in mice expressing human α-synuclein promotes the accumulation of misfolded α-synuclein and causes midbrain dopaminergic neuron degeneration. Our study thus identifies a neuroprotective function of microglia in the clearance of α-synuclein via TLR4-NF-κB-p62 mediated synucleinphagy.


Subject(s)
Autophagy/physiology , Microglia/metabolism , Neurodegenerative Diseases/metabolism , Toll-Like Receptor 4/metabolism , alpha-Synuclein/metabolism , Animals , Autoantigens/metabolism , Brain/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Female , HEK293 Cells , Humans , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , NF-kappa B/metabolism , Signal Transduction
10.
Mol Neurodegener ; 14(1): 43, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31775806

ABSTRACT

BACKGROUND: Dysfunctional autophagy is implicated in Alzheimer's Disease (AD) pathogenesis. The alterations in the expression of many autophagy related genes (ATGs) have been reported in AD brains; however, the disparity of the changes confounds the role of autophagy in AD. METHODS: To further understand the autophagy alteration in AD brains, we analyzed transcriptomic (RNAseq) datasets of several brain regions (BA10, BA22, BA36 and BA44 in 223 patients compared to 59 healthy controls) and measured the expression of 130 ATGs. We used autophagy-deficient mouse models to assess the impact of the identified ATGs depletion on memory, autophagic activity and amyloid-ß (Aß) production. RESULTS: We observed significant downregulation of multiple components of two autophagy kinase complexes BECN1-PIK3C3 and ULK1/2-FIP200 specifically in the parahippocampal gyrus (BA36). Most importantly, we demonstrated that deletion of NRBF2, a component of the BECN1-PIK3C3 complex, which also associates with ULK1/2-FIP200 complex, impairs memory in mice, alters long-term potentiation (LTP), reduces autophagy in mouse hippocampus, and promotes Aß accumulation. Furthermore, AAV-mediated NRBF2 overexpression in the hippocampus not only rescues the impaired autophagy and memory deficits in NRBF2-depleted mice, but also reduces ß-amyloid levels and improves memory in an AD mouse model. CONCLUSIONS: Our data not only implicates NRBF2 deficiency as a risk factor for cognitive impairment associated with AD, but also support the idea of NRBF2 as a potential therapeutic target for AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Autophagy-Related Proteins/genetics , Autophagy/physiology , Memory/physiology , Trans-Activators/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Cognitive Dysfunction/pathology , Disease Models, Animal , Memory Disorders/metabolism , Mice , Mice, Transgenic , Neurons/metabolism
11.
Nat Commun ; 10(1): 5234, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31748532

ABSTRACT

Genetic and genomic studies have advanced our knowledge of inherited Parkinson's disease (PD), however, the etiology and pathophysiology of idiopathic PD remain unclear. Herein, we perform a meta-analysis of 8 PD postmortem brain transcriptome studies by employing a multiscale network biology approach to delineate the gene-gene regulatory structures in the substantia nigra and determine key regulators of the PD transcriptomic networks. We identify STMN2, which encodes a stathmin family protein and is down-regulated in PD brains, as a key regulator functionally connected to known PD risk genes. Our network analysis predicts a function of human STMN2 in synaptic trafficking, which is validated in Stmn2-knockdown mouse dopaminergic neurons. Stmn2 reduction in the mouse midbrain causes dopaminergic neuron degeneration, phosphorylated α-synuclein elevation, and locomotor deficits. Our integrative analysis not only begins to elucidate the global landscape of PD transcriptomic networks but also pinpoints potential key regulators of PD pathogenic pathways.


Subject(s)
Gene Regulatory Networks/genetics , Parkinson Disease/genetics , Stathmin/genetics , Substantia Nigra/metabolism , Animals , Dopaminergic Neurons , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Locomotion , Mice , Phosphorylation , Transcriptome , alpha-Synuclein/metabolism
12.
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.

13.
PLoS One ; 11(9): e0163029, 2016.
Article in English | MEDLINE | ID: mdl-27631370

ABSTRACT

The deposit of polyubiquitinated aggregates has been implicated in the pathophysiology of Parkinson's disease (PD), and growing evidence indicates that selective autophagy plays a critical role in the clearance of ubiquitin-positive protein aggregates by autophagosomes. The selective autophagic receptor p62/SQSTM-1, which associates directly with both ubiquitin and LC3, transports ubiquitin conjugates to autophagosomes for degradation. Leucine-rich repeat kinase 2 (LRRK2), a PD-associated protein kinase, is tightly controlled by autophagy-lysosome degradation as well as by the ubiquitin-proteasome pathway. However, little is known about the degradation of ubiquitinated LRRK2 via selective autophagy. In the present study, we found that p62/SQSTM-1 physically interacts with LRRK2 as a selective autophagic receptor. The overexpression of p62 leads to the robust degradation of LRRK2 through the autophagy-lysosome pathway. In addition, LRRK2 indirectly regulates Ser351 and Ser403 phosphorylation of p62. Of particular interest, the interaction between phosphorylated p62 and Keap1 is reduced by LRRK2 overexpression. Therefore, we propose that the interplay between LRRK2 and p62 may contribute to the pathophysiological function and homeostasis of LRRK2 protein.


Subject(s)
Autophagy , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Sequestosome-1 Protein/metabolism , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Lysosomes/metabolism , Neurons/metabolism , Phosphorylation , Pregnancy , Proteasome Endopeptidase Complex/metabolism , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
14.
Exp Neurobiol ; 25(1): 14-23, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26924929

ABSTRACT

PTEN-induced putative kinase 1 (PINK1) is a Parkinson's disease (PD) gene. We examined miRNAs regulated by PINK1 during brain development and neural stem cell (NSC) differentiation, and found that lvels of miRNAs related to tumors and inflammation were different between 1-day-old-wild type (WT) and PINK1-knockout (KO) mouse brains. Notably, levels of miR-326, miR-330 and miR-3099, which are related to astroglioma, increased during brain development and NSC differentiation, and were significantly reduced in the absence of PINK1. Interestingly, in the presence of ciliary neurotrophic factor (CNTF), which pushes differentiation of NSCs into astrocytes, miR-326, miR-330, and miR-3099 levels in KO NSCs were also lower than those in WT NSCs. Furthermore, mimics of all three miRNAs increased expression of the astrocytic marker glial fibrillary acidic protein (GFAP) during differentiation of KO NSCs, but inhibitors of these miRNAs decreased GFAP expression in WT NSCs. Moreover, these miRNAs increased the translational efficacy of GFAP through the 3'-UTR of GFAP mRNA. Taken together, these results suggest that PINK1 deficiency reduce expression levels of miR-326, miR-330 and miR-3099, which may regulate GFAP expression during NSC differentiation and brain development.

16.
Mol Brain ; 9: 5, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26746235

ABSTRACT

BACKGROUND: Mutation of PTEN-induced putative kinase 1 (PINK1) causes autosomal recessive early-onset Parkinson's disease (PD). Despite of its ubiquitous expression in brain, its roles in non-neuronal cells such as neural stem cells (NSCs) and astrocytes were poorly unknown. RESULTS: We show that PINK1 expression increases from embryonic day 12 to postnatal day 1 in mice, which represents the main period of brain development. PINK1 expression also increases during neural stem cell (NSC) differentiation. Interestingly, expression of GFAP (a marker of astrocytes) was lower in PINK1 knockout (KO) mouse brain lysates compared to wild-type (WT) lysates at postnatal days 1-8, whereas there was little difference in the expression of markers for other brain cell types (e.g., neurons and oligodendrocytes). Further experiments showed that PINK1-KO NSCs were defective in their differentiation to astrocytes, producing fewer GFAP-positive cells compared to WT NSCs. However, the KO and WT NSCs did not differ in their self-renewal capabilities or ability to differentiate to neurons and oligodendrocytes. Interestingly, during differentiation of KO NSCs there were no defects in mitochondrial function, and there were not changes in signaling molecules such as SMAD1/5/8, STAT3, and HES1 involved in differentiation of NSCs into astrocytes. In brain sections, GFAP-positive astrocytes were more sparsely distributed in the corpus callosum and substantia nigra of KO animals compared with WT. CONCLUSION: Our study suggests that PINK1 deficiency causes defects in GFAP-positive astrogliogenesis during brain development and NSC differentiation, which may be a factor to increase risk for PD.


Subject(s)
Astrocytes/cytology , Brain/embryology , Brain/metabolism , Cell Differentiation , Glial Fibrillary Acidic Protein/metabolism , Neural Stem Cells/cytology , Protein Kinases/metabolism , Animals , Astrocytes/metabolism , Brain/cytology , Cell Proliferation , Cell Self Renewal/genetics , Cerebral Ventricles/metabolism , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein/genetics , Mice, Knockout , Mitochondria/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Protein Kinases/deficiency , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Substantia Nigra/metabolism
17.
Nat Commun ; 6: 8255, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26365310

ABSTRACT

In response to brain injury, microglia rapidly extend processes that isolate lesion sites and protect the brain from further injury. Here we report that microglia carrying a pathogenic mutation in the Parkinson's disease (PD)-associated gene, G2019S-LRRK2 (GS-Tg microglia), show retarded ADP-induced motility and delayed isolation of injury, compared with non-Tg microglia. Conversely, LRRK2 knockdown microglia are highly motile compared with control cells. In our functional assays, LRRK2 binds to focal adhesion kinase (FAK) and phosphorylates its Thr-X-Arg/Lys (TXR/K) motif(s), eventually attenuating FAK activity marked by decreased pY397 phosphorylation (pY397). GS-LRRK2 decreases the levels of pY397 in the brain, microglia and HEK cells. In addition, treatment with an inhibitor of LRRK2 kinase restores pY397 levels, decreased pTXR levels and rescued motility of GS-Tg microglia. These results collectively suggest that G2019S mutation of LRRK2 may contribute to the development of PD by inhibiting microglial response to brain injury.


Subject(s)
Brain Injuries , Cell Movement/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Microglia/metabolism , Protein Serine-Threonine Kinases/genetics , Wound Healing/genetics , Animals , Blotting, Western , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Gene Knockdown Techniques , HEK293 Cells , Humans , Immunoprecipitation , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Mice, Transgenic , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Rats
18.
Hum Mol Genet ; 24(4): 1127-41, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25305081

ABSTRACT

Deciphering the molecular basis of neuronal cell death is a central issue in the etiology of neurodegenerative diseases, such as Parkinson's and Alzheimer's. Dysregulation of p53 levels has been implicated in neuronal apoptosis. The role of histone deacetylase 3 (HDAC3) in suppressing p53-dependent apoptosis has been recently emphasized; however, the molecular basis of modulation of p53 function by HDAC3 remains unclear. Here we show that PTEN-induced putative kinase 1 (PINK1), which is linked to autosomal recessive early-onset familial Parkinson's disease, phosphorylates HDAC3 at Ser-424 to enhance its HDAC activity in a neural cell-specific manner. PINK1 prevents H2O2-induced C-terminal cleavage of HDAC3 via phosphorylation of HDAC3 at Ser-424, which is reversed by protein phosphatase 4c. PINK1-mediated phosphorylation of HDAC3 enhances its direct association with p53 and causes subsequent hypoacetylation of p53. Genetic deletion of PINK1 partly impaired the suppressive role of HDAC3 in regulating p53 acetylation and transcriptional activity. However, depletion of HDAC3 fully abolished the PINK1-mediated p53 inhibitory loop. Finally, ectopic expression of phosphomometic-HDAC3(S424E) substantially overcomes the defective action of PINK1 against oxidative stress in dopaminergic neuronal cells. Together, our results uncovered a mechanism by which PINK1-HDAC3 network mediates p53 inhibitory loop in response to oxidative stress-induced damage.


Subject(s)
Dopaminergic Neurons/metabolism , Histone Deacetylases/metabolism , Protein Kinases/metabolism , Acetylation/drug effects , Animals , Caspase 7/metabolism , Cell Death/genetics , Cell Line , Cytoplasm/metabolism , Dopaminergic Neurons/pathology , Enzyme Activation , Histone Deacetylases/genetics , Humans , Hydrogen Peroxide/pharmacology , Mice , Organ Specificity , Phosphorylation , Protein Kinases/genetics , Proteolysis , Tumor Suppressor Protein p53/metabolism
19.
Mol Cells ; 37(4): 345-55, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24802057

ABSTRACT

Mitigating secondary delayed neuronal injury has been a therapeutic strategy for minimizing neurological symptoms after several types of brain injury. Interestingly, secondary neuronal loss appeared to be closely related to functional loss and/or death of astrocytes. In the brain damage induced by agonists of two glutamate receptors, N-ethyl-D-aspartic acid (NMDA) and kainic acid (KA), NMDA induced neuronal death within 3 h, but did not increase further thereafter. However, in the KA-injected brain, neuronal death was not obviously detectable even at injection sites at 3 h, but extensively increased to encompass the entire hemisphere at 7 days. Brain inflammation, a possible cause of secondary neuronal damage, showed little differences between the two models. Importantly, however, astrocyte behavior was completely different. In the NMDA-injected cortex, the loss of glial fibrillary acidic protein-expressing (GFAP+) astrocytes was confined to the injection site until 7 days after the injection, and astrocytes around the damage sites showed extensive gliosis and appeared to isolate the damage sites. In contrast, in the KA-injected brain, GFAP+ astrocytes, like neurons, slowly, but progressively, disappeared across the entire hemisphere. Other markers of astrocytes, including S100ß, glutamate transporter EAAT2, the potassium channel Kir4.1 and glutamine synthase, showed patterns similar to that of GFAP in both NMDA- and KA-injected cortexes. More importantly, astrocyte disappearance and/or functional loss preceded neuronal death in the KA-injected brain. Taken together, these results suggest that loss of astrocyte support to neurons may be a critical cause of delayed neuronal death in the injured brain.


Subject(s)
Astrocytes/drug effects , Brain Injuries/drug therapy , Cell Death , Cerebral Cortex/drug effects , Gliosis/drug therapy , Kainic Acid/administration & dosage , N-Methylaspartate/administration & dosage , Animals , Astrocytes/physiology , Biomarkers/metabolism , Cell Communication/drug effects , Cell Death/drug effects , Excitatory Amino Acid Transporter 2/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glutamate-Ammonia Ligase/metabolism , Male , Neurons/drug effects , Neurons/physiology , Potassium Channels, Inwardly Rectifying/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism
20.
Exp Neurobiol ; 22(1): 38-44, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23585721

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative motor disease caused by degeneration of dopaminergic neurons in the substantia nigra. Because brain inflammation has been considered a risk factor for PD, we analyzed whether PTEN induced putative kinase 1 (PINK1), an autosomal recessive familial PD gene, regulates brain inflammation during injury states. Using acutely prepared cortical slices to mimic injury, we analyzed expression of the pro-inflammatory cytokines tumor necrosis factor-α, interleukin (IL)-1ß, and IL-6 at the mRNA and protein levels. Both mRNA and protein expression of these cytokines was higher at 6-24 h after slicing in PINK1 knockout (KO) slices compared to that in wild-type (WT) slices. In serial experiments to understand the signaling pathways that increase inflammatory responses in KO slices, we found that IκB degradation was enhanced but Akt phosphorylation decreased in KO slices compared to those in WT slices. In further experiments, an inhibitor of PI3K (LY294002) upstream of Akt increased expression of pro-inflammatory cytokines. Taken together, these results suggest that PINK1 deficiency enhance brain inflammation through reduced Akt activation and enhanced IκB degradation in response to brain injury.

SELECTION OF CITATIONS
SEARCH DETAIL
...