Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-37464816

ABSTRACT

Aims: Cystathionine ß-synthase (CBS) is essential for homocysteine (Hcy) transsulfuration, yielding cysteine as a common precursor of hydrogen sulfide (H2S), glutathione (GSH), and other sulfur molecules, which produce neuroprotective effects in neurological conditions. We previously reported a disruption of microglial CBS/H2S signaling in a Parkinson's disease (PD) mouse model. Yet, it remains unclear whether CBS affects nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing 3 (NLRP3) inflammasome activity and other pathologies in PD. Results: Microglial CBS expression decreased after lipopolysaccharide (LPS) stimulation. Elevated GSSG (the oxidized GSH) content and decreased H2S generation were found in the brains of microglial cbs conditional-knockout (cbscKO) mice, whereas serum and brain Hcy levels remained unaltered. Moreover, microglial cbscKO mice were susceptible to NLRP3 inflammasome activation and dopaminergic neuron losses caused by LPS injection into the substantia nigra, whereas cbs overexpression or activation produced opposite effects. In vitro studies showed that cbs overexpression or activation suppressed microglial NLRP3 inflammasome activation and interleukin (IL)-1ß secretion by reducing mitochondrial reactive oxygen species (mitoROS) level. Conversely, ablation of cbs enhanced NLRP3 expression and mitoROS generation and augmented microglial NLRP3 inflammasome activity in response to adenosine triphosphate challenge, which was blocked by the mitoROS scavenger. Innovation and Conclusion: The study demonstrated an elevated GSSG level and reduced H2S generation, which correlated with a susceptible status of microglia in the brain of cbscKO mice. Our findings reveal a critical role of CBS in restraining the microglial NLRP3 inflammasome by controlling redox homeostasis and highlight that activation or upregulation of CBS may become a potential strategy for PD treatment.

2.
Front Aging Neurosci ; 14: 979869, 2022.
Article in English | MEDLINE | ID: mdl-36034136

ABSTRACT

Microglia are the resident innate immune cells in the central nervous system (CNS) that serve as the first line innate immunity in response to pathogen invasion, ischemia and other pathological stimuli. Once activated, they rapidly release a variety of inflammatory cytokines and phagocytose pathogens or cell debris (termed neuroinflammation), which is beneficial for maintaining brain homeostasis if appropriately activated. However, excessive or uncontrolled neuroinflammation may damage neurons and exacerbate the pathologies in neurological disorders. Microglia are highly dynamic cells, dependent on energy supply from mitochondria. Moreover, dysfunctional mitochondria can serve as a signaling platform to facilitate innate immune responses in microglia. Mitophagy is a means of clearing damaged or redundant mitochondria, playing a critical role in the quality control of mitochondrial homeostasis and turnover. Mounting evidence has shown that mitophagy not only limits the inflammatory response in microglia but also affects their phagocytosis, whereas mitochondria dysfunction and mitophagy defects are associated with aging and neurological disorders. Therefore, targeting microglial mitophagy is a promising therapeutic strategy for neurological disorders. This article reviews and highlights the role and regulation of mitophagy in microglia in neurological conditions, and the research progress in manipulating microglial mitophagy and future directions in this field are also discussed.

3.
Aging Cell ; 20(12): e13522, 2021 12.
Article in English | MEDLINE | ID: mdl-34811872

ABSTRACT

The cell-to-cell transfer of α-synuclein (α-Syn) greatly contributes to Parkinson's disease (PD) pathogenesis and underlies the spread of α-Syn pathology. During this process, extracellular α-Syn can activate microglia and neuroinflammation, which plays an important role in PD. However, the effect of extracellular α-Syn on microglia autophagy is poorly understood. In the present study, we reported that extracellular α-Syn inhibited the autophagy initiation, as indicated by LC3-II reduction and p62 protein elevation in BV2 and cultured primary microglia. The in vitro findings were verified in microglia-enriched population isolated from α-Syn-overexpressing mice induced by adeno-associated virus (AAV2/9)-encoded wildtype human α-Syn injection into the substantia nigra (SN). Mechanistically, α-Syn led to microglial autophagic impairment through activating toll-like receptor 4 (Tlr4) and its downstream p38 and Akt-mTOR signaling because Tlr4 knockout and inhibition of p38, Akt as well as mTOR prevented α-Syn-induced autophagy inhibition. Moreover, inhibition of Akt reversed the mTOR activation but failed to affect p38 phosphorylation triggered by α-Syn. Functionally, the in vivo evidence showed that lysozyme 2 Cre (Lyz2cre )-mediated depletion of autophagy-related gene 5 (Atg5) in microglia aggravated the neuroinflammation and dopaminergic neuron losses in the SN and exacerbated the locomotor deficit in α-Syn-overexpressing mice. Taken together, the results suggest that extracellular α-Syn, via Tlr4-dependent p38 and Akt-mTOR signaling cascades, disrupts microglial autophagy activity which synergistically contributes to neuroinflammation and PD development.


Subject(s)
Autophagy/genetics , Neuroinflammatory Diseases/genetics , Parkinson Disease/genetics , alpha-Synuclein/metabolism , Animals , Disease Models, Animal , Mice
4.
Neurochem Int ; 150: 105187, 2021 11.
Article in English | MEDLINE | ID: mdl-34534609

ABSTRACT

Hydrogen sulfide (H2S) serves as a neuromodulator and regulator of neuroinflammation. It is reported to be therapeutic for Parkinson's disease (PD) animal and cellular models. However, whether it affects α-synuclein accumulation in dopaminergic cells, the key pathological feature in PD, is poorly understood. In this study we reported that exogenous H2S donors NaHS and GYY4137 (GYY) enhanced the autophagy activity, as indicated by the increases of autophagy marker LC3-II expression and LC3 dots formation even during lysosome inhibition in dopaminergic cell lines and HEK293 cells. The enhancement of H2S donors on autophagic flux was mediated by adenosine 5'-monophosphate-activated protein kinase (AMPK)-dependent mammalian target of rapamycin (mTOR) inhibition, as H2S donors activated AMPK but reduced the mTOR activity and H2S donors-induced LC3-II increase was diminished by mTOR activator. Moreover, point mutation of Cys302 into alanine (C302A) in AMPKα2 subunit abolished the AMPK activation and mTOR inhibition, as well as autophagic flux increase elicited by NaHS. Interestingly, NaHS triggered AMPK S-sulfuration, which was not observed in AMPK C302A-transfected cells. Further, NaHS was able to attenuate α-synuclein accumulation in a cellular model induced by dopamine oxidized metabolite 3, 4-dihydroxyphenylacetaldehyde (DOPAL), and this effect was interfered by autophagy inhibitor wortmannin and also eliminated in AMPK Cys302A-transfected cells. In sum, the findings identified a role of Cys302 S-sulfuration in AMPK activation induced by exogenous H2S and demonstrated that H2S donors could enhance the autophagic flux via AMPK-mTOR signaling and thus reduce α-synuclein accumulation in vitro.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/physiology , Dopaminergic Neurons/metabolism , Hydrogen Sulfide/metabolism , Morpholines/pharmacology , Organothiophosphorus Compounds/pharmacology , Sulfides/pharmacology , Animals , Autophagy/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hydrogen Sulfide/agonists , PC12 Cells , Phosphorylation/drug effects , Phosphorylation/physiology , Rats
5.
Brain Behav Immun ; 67: 77-90, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28774789

ABSTRACT

Hydrogen sulfide (H2S), a novel neuromodulator, is linked to the pathogenesis of several neurodegenerative disorders. Exogenous application of H2S exerts neuroprotection via anti-inflammation and anti-oxidative stress in animal and cellular models of Parkinson's disease (PD). However, the role of endogenous H2S and the contribution of its various synthases in PD remain unclear. In the present study, we found a decline of plasma and striatal sulfide level in 1-methy-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced PD mouse model. Interestingly, among the three H2S generating enzymes, only cystathionine ß-synthase (CBS) expression was largely reduced in the striatum of MPTP-treated mice. The in vitro study confirmed a significant decrease of CBS expression in 1-methyl-4-phenylpyridinium (MPP+)-stimulated astrocytes and microglia, but not in neurons or SH-SY5Y dopaminergic cells. Striatal CBS overexpression, elicited by stereotaxic delivery with Cbs gene using recombinant adeno-associated-virus (rAAV-Cbs), successfully enhanced the sulfide level in the striatum and partially rescued the MPTP-induced dopaminergic neurotoxicity in the midbrain. Specifically, striatal CBS overexpression alleviated the motor deficits and dopaminergic neuron losses in the nigro-striatal pathway, with a concomitant inhibition of glial activation in MPTP-treated mice. Furthermore, compared to rAAV-Vector, rAAV-Cbs injection reduced the aberrant accumulation of nitric oxide and 3-nitrotyrosine (an indicator of protein nitration) in the striatum of MPTP-treated mice. Notably, it also attenuated the increase of nitrated α-synuclein level in MPTP mice. The in vitro study demonstrated that lentivirus-mediated CBS overexpression elevated the sulfide generation in glial cells. Moreover, glial CBS overexpression offered protection to midbrain dopaminergic neurons through repressing nitric oxide overproduction in both glial and neuronal cells induced by MPP+. Taken together, our data suggest that impaired CBS-H2S axis may contribute to the pathogenesis of PD, and that modulation of this axis may become a novel therapeutic approach for PD.


Subject(s)
Corpus Striatum/enzymology , Cystathionine beta-Synthase/metabolism , Hydrogen Sulfide/metabolism , Parkinson Disease/enzymology , Animals , Astrocytes/enzymology , Cell Line, Tumor , Cells, Cultured , Disease Models, Animal , Dopaminergic Neurons/enzymology , Humans , Male , Mice, Inbred C57BL , Microglia/enzymology , Parkinsonian Disorders/enzymology , Signal Transduction
6.
Exp Neurol ; 297: 138-147, 2017 11.
Article in English | MEDLINE | ID: mdl-28821398

ABSTRACT

Serum urate levels are reported to be significantly lowered in patients with Parkinson's disease (PD) and inversely correlated to the risk and progression of PD. However, the mechanism by which urate affects PD is poorly understood. Here we showed that treatment with uric acid (UA) resulted in an autophagy activity enhancement in PC12 cells in dose- and time-dependent manners, as indicated by LC3-II increase and P62 decrease. Moreover, UA was still able to increase the LC3-II level and the number of LC3 puncta in the presence of Bafilomycin A1, a lysosomal inhibitor. These changes of autophagic markers were preceded by mTOR inhibition and ULK1 activation. Co-treatment with 3-benzyl-5-((2-nitrophenoxy) methyl)-dihydrofuran-2(3H)-one (3BDO), an mTOR activator, abolished the UA-induced LC3-II increase. More importantly, UA reduced SNCA/α-synuclein accumulation in PC12 cells that overexpress wildtype or A53T mutant SNCA, and this was blocked by Bafilomycin A1 co-treatment. The in vivo study showed that UA administration was able to modulate the levels of autophagy markers, increase the autophagosome/autolysosome formation, and reduce SNCA accumulation in the midbrain of SNCAA53T transgenic mice. Taken together, our findings suggest that UA could induce autophagy activation via an mTOR-dependent signaling and ameliorate SNCA accumulation. This implicates that urate-elevating agent may become a potential strategy for PD therapy.


Subject(s)
Autophagy/physiology , TOR Serine-Threonine Kinases/physiology , Uric Acid/pharmacology , alpha-Synuclein/metabolism , Animals , Autophagy/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Humans , Male , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , PC12 Cells , Pilot Projects , Rats
7.
Acta Pharmacol Sin ; 36(11): 1300-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26364802

ABSTRACT

AIM: Parkin has been shown to exert protective effects against 6-hydroxydopamine (6-OHDA)-induced neurotoxicity in different models of Parkinson disease. In the present study we investigated the molecular mechanisms underlying the neuroprotective action of parkin in vitro. METHODS: HEK293, HeLa and PC12 cells were transfected with parkin, parkin mutants, p62 or si-p62. Protein expression and ubiquitination were assessed using immunoblot analysis. Immunoprecipitation assay was performed to identify the interaction between parkin and scaffold protein p62. PC12 and SH-SY5Y cells were treated with 6-OHDA (200 µmol/L), and cell apoptosis was detected using PI and Hoechst staining. RESULTS: In HEK293 cells co-transfected with parkin and p62, parkin was co-immunoprecipitated with p62, and parkin overexpression increased p62 protein levels. In parkin-deficient HeLa cells, transfection with wild-type pakin, but not with ligase activity-deficient pakin mutants, significantly increased p62 levels, suggesting that parkin stabilized p62 through its E3 ligase activity. Transfection with parkin or p62 significantly repressed ERK1/2 phosphorylation in HeLa cells, but transfection with parkin did not repress ERK1/2 phosphorylation in p62-knockdown HeLa cells, suggesting that p62 was involved in parkin-induced inhibition on ERK1/2 phosphorylation. Overexpression of parkin or p62 significantly repressed 6-OHDA-induced ERK1/2 phosphorylation in PC12 cells, and parkin overexpression inhibited 6-OHDA-induced apoptosis in PC12 and SH-SY5Y cells. CONCLUSION: Parkin protects PC12 cells against 6-OHDA-induced apoptosis via ubiquitinating and stabilizing scaffold protein p62, and repressing ERK1/2 activation.


Subject(s)
Apoptosis , Heat-Shock Proteins/metabolism , Oxidopamine/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , HEK293 Cells , HeLa Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , PC12 Cells , Rats , Sequestosome-1 Protein , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...