Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
J Neuroinflammation ; 17(1): 64, 2020 Feb 18.
Article in English | MEDLINE | ID: mdl-32070376

ABSTRACT

BACKGROUND: Sepsis-associated acute brain inflammation, if unresolved, may cause chronic neuroinflammation and resultant neurodegenerative diseases. However, little is known how the transition from acute to chronic neuroinflammation, which is critical for the following progressive neurodegeneration, occurs in sepsis. The goal of this study was to investigate potential immune factors regulating the transition process using a widely used endotoxemia LPS mouse model. This model shows distinct acute and chronic phases of neuroinflammation and recapitulates many cardinal features of Parkinson's disease, thus, providing a unique opportunity for studying phase transition of neuroinflammation. METHODS: C57BL/6 J, NLRP3-/-, and IL-1R1-/- mice were employed. Mild and severe endotoxemia were produced by LPS ip injection at 1 or 5 mg/kg. Neuroinflammation in vitro and in vivo was assessed with proinflammatory cytokine expression by qPCR or ELISA and microglial activation by immunohistochemical analysis. Neurodegeneration was measured by manual and stereological counts of nigral dopaminergic neurons and immunohistochemical analysis of protein nitrosylation and α-synuclein phosphorylation. RESULTS: LPS-elicited initial increases in mouse brain mRNA levels of TNFα, IL-6, IL-1ß, and MCP-1, and nigral microglial activation were not dose-related. By contrast, the delayed increase in brain mature IL-1ß levels was dependent on LPS doses and protracted nigral microglial activation was only observed in high dose of LPS-treated mice. LPS-elicited increase in brain mature IL-1ß but not IL-1α level was NLRP3-dependent. After high dose LPS treatment, deficiency of NLRP3 or IL-1R1 did not prevent the initiation of acute neuroinflammation but abolished chronic neuroinflammation. Genetic or pharmacological inhibition of the NLRP3-IL-1ß axis repressed LPS-stimulated upregulation of chronic neuroinflammatory mediators including MHC-II, NOX2, and Mac1, and protected dopaminergic neurons. Ten months after LPS-elicited severe endotoxemia, nigral persisted microglial activation, elevated nitrosylated proteins and phosphorylated α-synuclein, and significant neuronal degeneration developed in wild-type mice but not in NLRP3-/- or IL-1R1-/- mice. CONCLUSIONS: This study uncovers a novel role of the NLRP3-IL-1ß signaling pathway in gauging the severity of sepsis-associated inflammation and determining whether acute neuroinflammation will resolve or transition to low grade chronic neuroinflammation. These findings also provide novel targets for developing therapy for severe systemic infection-related neurodegeneration.


Subject(s)
Disease Progression , Inflammation Mediators/metabolism , Interleukin-1beta/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Neurodegenerative Diseases/metabolism , Sepsis/metabolism , Acute Disease , Animals , Cells, Cultured , Chronic Disease , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurodegenerative Diseases/chemically induced , Sepsis/chemically induced
2.
Brain Behav Immun ; 87: 359-368, 2020 07.
Article in English | MEDLINE | ID: mdl-31923552

ABSTRACT

Parkinson's disease (PD) develops over decades through spatiotemporal stages that ascend from the brainstem to the forebrain. The mechanism behind this caudo-rostral neurodegeneration remains largely undefined. In unraveling this phenomenon, we recently developed a lipopolysaccharide (LPS)-elicited chronic neuroinflammatory mouse model that displays sequential losses of neurons in brainstem, substantia nigra, hippocampus and cortex. In this study, we aimed to investigate the mechanisms of caudo-rostral neurodegeneration and focused our efforts on the earliest neurodegeneration of vulnerable noradrenergic locus coeruleus (NE-LC) neurons in the brainstem. We found that compared with neurons in other brain regions, NE-LC neurons in untreated mice displayed high levels of mitochondrial oxidative stress that was severely exacerbated in the presence of LPS-elicited chronic neuroinflammation. In agreement, NE-LC neurons in LPS-treated mice displayed early reduction of complex IV expression and mitochondrial swelling and loss of cristae. Mechanistically, the activation of the superoxide-generating enzyme NADPH oxidase (NOX2) on NE-LC neurons was essential for their heightened vulnerability during chronic neuroinflammation. LPS induced early and high expressions of NOX2 in NE-LC neurons. Genetic or pharmacological inactivation of NOX2 markedly reduced mitochondrial oxidative stress and dysfunction in LPS-treated mice. Furthermore, inhibition of NOX2 significantly ameliorated LPS-induced NE-LC neurodegeneration. More importantly, post-treatment with NOX2 inhibitor diphenyleneiodonium when NE-LC neurodegeneration had already begun, still showed high efficacy in protecting NE-LC neurons from degeneration in LPS-treated mice. This study strongly supports that chronic neuroinflammation and NOX2 expression among vulnerable neuronal populations contribute to caudo-rostral degeneration in PD.


Subject(s)
Adrenergic Neurons , Locus Coeruleus , Animals , Dopaminergic Neurons , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Microglia
3.
Brain Behav Immun ; 81: 374-387, 2019 10.
Article in English | MEDLINE | ID: mdl-31247288

ABSTRACT

The loss of central norepinephrine (NE) released by neurons of the locus coeruleus (LC) occurs with aging, and is thought to be an important factor in producing the many of the nonmotor symptoms and exacerbating the degenerative process in animal models of Parkinson's disease (PD). We hypothesize that selectively depleting noradrenergic LC neurons prior to the induction of chronic neuroinflammation may not only accelerate the rate of progressive neurodegeneration throughout the brain, but may exacerbate nonmotor and motor behavioral phenotypes that recapitulate symptoms of PD. For this reason, we used a "two-hit" mouse model whereby brain NE were initially depleted by DSP-4 one week prior to exposing mice to LPS. We found that pretreatment with DSP-4 potentiated LPS-induced sequential neurodegeneration in SNpc, hippocampus, and motor cortex, but not in VTA and caudate/putamen. Mechanistic study revealed that DSP-4 enhanced LPS-induced microglial activation and subsequently elevated neuronal oxidative stress in affected brain regions in a time-dependent pattern. To further characterize the effects of DSP-4 on non-motor and motor symptoms in the LPS model, physiological and behavioral tests were performed at different time points following injection. Consistent with the enhanced neurodegeneration, DSP-4 accelerated the progressive deficits of non-motor symptoms including hyposmia, constipation, anxiety, sociability, exaggerated startle response and impaired learning. Furthermore, notable decreases of motor functions, including decreased rotarod activity, grip strength, and gait disturbance, were observed in treated mice. In summary, our studies provided not only an accelerated "two-hit" PD model that recapitulates the features of sequential neuron loss and the progression of motor/non-motor symptoms of PD, but also revealed the critical role of early LC noradrenergic neuron damage in the pathogenesis of PD-like symptoms.


Subject(s)
Nerve Degeneration/pathology , Neurodegenerative Diseases/physiopathology , Parkinson Disease/physiopathology , Adrenergic Neurons/pathology , Aging , Animals , Benzylamines/pharmacology , Brain/drug effects , Disease Models, Animal , Dopaminergic Neurons/drug effects , Hippocampus/pathology , Inflammation/pathology , Lipopolysaccharides/pharmacology , Locus Coeruleus/pathology , Male , Mice , Mice, Inbred C57BL , Microglia/pathology , Motor Activity/drug effects , Norepinephrine/pharmacology , Oxidative Stress
4.
Proc Natl Acad Sci U S A ; 112(15): E1926-35, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25825709

ABSTRACT

Malformed α-Synuclein (α-syn) aggregates in neurons are released into the extracellular space, activating microglia to induce chronic neuroinflammation that further enhances neuronal damage in α-synucleinopathies, such as Parkinson's disease. The mechanisms by which α-syn aggregates activate and recruit microglia remain unclear, however. Here we show that α-syn aggregates act as chemoattractants to direct microglia toward damaged neurons. In addition, we describe a mechanism underlying this directional migration of microglia. Specifically, chemotaxis occurs when α-syn binds to integrin CD11b, leading to H2O2 production by NADPH oxidase. H2O2 directly attracts microglia via a process in which extracellularly generated H2O2 diffuses into the cytoplasm and tyrosine protein kinase Lyn, phosphorylates the F-actin-associated protein cortactin after sensing changes in the microglial intracellular concentration of H2O2. Finally, phosphorylated cortactin mediates actin cytoskeleton rearrangement and facilitates directional cell migration. These findings have significant implications, given that α-syn-mediated microglial migration reaches beyond Parkinson's disease.


Subject(s)
Cell Movement , Hydrogen Peroxide/metabolism , Microglia/metabolism , alpha-Synuclein/metabolism , src-Family Kinases/metabolism , Animals , Animals, Newborn , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cells, Cultured , Chemotactic Factors/metabolism , Cortactin/metabolism , Hydrogen Peroxide/pharmacology , Immunoblotting , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Microglia/drug effects , Microscopy, Confocal , NADPH Oxidase 2 , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Neurons/cytology , Neurons/metabolism , Oxidants/metabolism , Oxidants/pharmacology , Phosphorylation , RNA Interference , Rats , Signal Transduction , alpha-Synuclein/genetics , alpha-Synuclein/pharmacology , src-Family Kinases/genetics
5.
J Neuroinflammation ; 13(1): 158, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27329107

ABSTRACT

BACKGROUND: Misfolded α-synuclein (α-Syn) aggregates participate in the pathogenesis of synucleinopathies, such as Parkinson's disease. Whereas much is known about how the various domains within full-length α-Syn (FL-α-Syn) contribute to the formation of α-Syn aggregates and therefore to their neurotoxicity, little is known about whether the individual peptides that can be generated from α-syn, possibly as intermediate metabolites during degradation of misfolded α-Syn aggregates, are neurotoxic themselves. METHODS: A series of synthesized α-Syn peptides, corresponding to the locus in FL-α-Syn containing alanine 30, substitution of which with a proline causes a familial form of Parkinson's disease, were examined for their capacity of inducing release of microglial superoxide. The neurotoxicity of these peptides was measured according to their influence on the ability of neuroglial cultures deficient in gp91 (phox) , the catalytic unit of NADPH oxidase (Nox2), or wild-type cultures to take up (3)H-labeled dopamine and on the number of tyrosine hydroxylase-staining-positive neurons. Western blots and confocal images were utilized to analyze membrane translocation of p47 (phox) and p67 (phox) , phosphorylation of p47 (phox) and Erk1/2 kinase, and binding of α-Syn peptides to gp91 (phox) . Activation of brain microglia in mice injected with α-Syn peptides was demonstrated by immunostaining for major histocompatibility complex (MHC)-II along with qPCR for Iba-1 and MHC-II. RESULTS: We report α-Syn (29-40) as a specific peptide capable of activating microglial Nox2 to produce superoxide and cause dopaminergic neuronal damage. Administered to mice, this peptide also activated brain microglia to increase expression of MHC-II and Iba-1 and stimulated oxidation reaction. Exploring the underlying mechanisms showed that α-Syn (29-40) peptide triggered Nox2 to generate extracellular superoxide and its metabolite H2O2 by binding to the catalytic unit gp91 (phox) of Nox2; diffusing into cytosol, H2O2 activated Erk1/2 kinase to phosphorylate p47 (phox) and p67 (phox) and further activated Nox2, establishing a positive feedback loop to amplify the Nox2-mediated response. CONCLUSIONS: Collectively, our study suggests novel information regarding how α-Syn causes neuronal injury, possibly including mechanisms involving abnormal metabolites of α-Syn aggregates.


Subject(s)
Dopaminergic Neurons/drug effects , Neuroglia/drug effects , Superoxides/metabolism , Superoxides/toxicity , alpha-Synuclein/pharmacology , Animals , Animals, Newborn , Calcium-Binding Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian , Histocompatibility Antigens Class II/metabolism , MAP Kinase Signaling System/drug effects , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Neuroglia/metabolism , Peptide Fragments/pharmacology , Protein Transport/drug effects , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/genetics
6.
J Neuroinflammation ; 13(1): 110, 2016 05 16.
Article in English | MEDLINE | ID: mdl-27184631

ABSTRACT

BACKGROUND: Clozapine, an atypical antipsychotic medication, has been effectively used to treat refractory schizophrenia. However, the clinical usage of clozapine is limited due to a high incidence of neutropenia or agranulocytosis. We previously reported that clozapine protected dopaminergic neurons through inhibition of microglial activation. The purpose of this study was to explore the neuroprotective effects of clozapine metabolites clozapine N-oxide (CNO) and N-desmethylclozapine (NDC), as well as their propensity to cause neutropenia. METHODS: The primary midbrain neuron-glia culture was applied to detect the neuroprotective and anti-inflammatory effect of clozapine and its metabolites in lipopolysaccharide (LPS) and MPP(+)-induced toxicity. And the subsequent mechanism was demonstrated by gp91 (phox) mutant cell cultures as well as microgliosis cell lines. In vivo, to confirm the neuroprotective effect of clozapine and CNO, we measured the dopaminergic neuronal loss and rotarod motor deficits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-generated mouse Parkinson's disease (PD) model. The neutropenia or agranulocytosis of clozapine and its metabolites was illustrated by white blood cell count of the treated mice. RESULTS: We found that, in midbrain neuron-glia cultures, CNO and NDC were more potent than clozapine in protecting dopaminergic neurons against LPS and MPP(+)-induced toxicity. CNO and NDC-afforded neuroprotection was linked to inhibition of microglia-mediated neuroinflammation, as demonstrated by abolished neuroprotection in microglia-depleted cultures and their capacity of inhibiting LPS-induced release of proinflammatory factors from activated microglia. NADPH oxidase (NOX2) was subsequently recognized as the main target of CNO and NDC since genetic ablation of gp91 (phox) , the catalytic subunit of NOX2, abolished their neuroprotective effects. CNO and NDC inhibited NOX2 activation through interfering with the membrane translocation of the NOX2 cytosolic subunit, p47 (phox) . The neuroprotective effects of CNO were further verified in vivo as shown by attenuation of dopaminergic neurodegeneration, motor deficits, and reactive microgliosis in MPTP-generated mouse PD model. More importantly, unlike clozapine, CNO did not lower the white blood cell count. CONCLUSIONS: Altogether, our results show that clozapine metabolites elicited neuroprotection through inactivation of microglia by inhibiting NOX2. The robust neuroprotective effects and lack of neutropenia suggest that clozapine metabolites may be promising candidates for potential therapy for neurodegenerative diseases.


Subject(s)
Clozapine/metabolism , Dopaminergic Neurons/enzymology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Microglia/enzymology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neuroprotective Agents/metabolism , Animals , Antipsychotic Agents/metabolism , Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Coculture Techniques , Dopaminergic Neurons/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Female , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , NADPH Oxidase 2 , Neuroprotective Agents/pharmacology , Pregnancy , Rats , Rats, Inbred F344
7.
Brain ; 138(Pt 5): 1247-62, 2015 May.
Article in English | MEDLINE | ID: mdl-25716193

ABSTRACT

Nicotinamide adenine dinucleotide phosphate oxidase, a key superoxide-producing enzyme, plays a critical role in microglia-mediated chronic neuroinflammation and subsequent progressive dopaminergic neurodegeneration in Parkinson's disease. Although nicotinamide adenine dinucleotide phosphate oxidase-targeting anti-inflammatory therapy for Parkinson's disease has been proposed, its application in translational research remains limited. The aim of this study was to obtain preclinical evidence supporting this therapeutic strategy by testing the efficacy of an ultra-low dose of the nicotinamide adenine dinucleotide phosphate oxidase inhibitor diphenyleneiodonium in both endotoxin (lipopolysaccharide)- and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice using post-treatment regimens. Our data revealed that post-treatment with diphenyleneiodonium significantly attenuated progressive dopaminergic degeneration and improved rotarod activity. Remarkably, post-treatment with diphenyleneiodonium 10 months after lipopolysaccharide injection when mice had 30% loss of nigral dopaminergic neurons, showed high efficacy in protecting the remaining neuronal population and restoring motor function. Diphenyleneiodonium-elicited neuroprotection was associated with the inhibition of microglial activation, a reduction in the expression of proinflammatory factors and an attenuation of α-synuclein aggregation. A pathophysiological evaluation of diphenyleneiodonium-treated mice, including assessment of body weight, organs health, and neuronal counts, revealed no overt signs of toxicity. In summary, infusion of ultra-low dose diphenyleneiodonium potently reduced microglia-mediated chronic neuroinflammation by selectively inhibiting nicotinamide adenine dinucleotide phosphate oxidase and halted the progression of neurodegeneration in mouse models of Parkinson's disease. The robust neuroprotective effects and lack of apparent toxic side effects suggest that diphenyleneiodonium at ultra-low dose may be a promising candidate for future clinical trials in Parkinson's disease patients.


Subject(s)
Dopaminergic Neurons/drug effects , Enzyme Inhibitors/pharmacology , Microglia/drug effects , NADPH Oxidases/antagonists & inhibitors , Nerve Degeneration/drug therapy , Onium Compounds/pharmacology , Parkinson Disease/drug therapy , Animals , Disease Models, Animal , Disease Progression , Mice, Inbred C57BL , NADPH Oxidases/metabolism , Nerve Degeneration/pathology , Neuroprotective Agents/therapeutic use , Parkinson Disease/pathology , Substantia Nigra/metabolism
8.
J Neurosci ; 34(37): 12490-503, 2014 Sep 10.
Article in English | MEDLINE | ID: mdl-25209287

ABSTRACT

Although dysregulated substance P (SP) has been implicated in the pathophysiology of Parkinson's disease (PD), how SP affects the survival of dopaminergic neurons remains unclear. Here, we found that mice lacking endogenous SP (TAC1(-/-)), but not those deficient in the SP receptor (neurokinin-1 receptor, NK1R), were more resistant to lipopolysaccharide (LPS)- and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced nigral dopaminergic neurodegeneration than wild-type controls, suggesting a NK1R-independent toxic action of SP. In vitro dose-response studies revealed that exogenous SP enhanced LPS- and 1-methyl-4-phenylpyridinium (MPP(+))-induced dopaminergic neurodegeneration in a bimodal manner, peaking at submicromolar and subpicomolar concentrations, but was substantially less effective at intermediate concentrations. Mechanistically, the actions of submicromolar levels of SP were NK1R-dependent, whereas subpicomolar SP-elicited actions required microglial NADPH oxidase (NOX2), the key superoxide-producing enzyme, but not NK1R. Subpicomolar concentrations of SP activated NOX2 by binding to the catalytic subunit gp91(phox) and inducing membrane translocation of the cytosolic subunits p47(phox) and p67(phox). The importance of NOX2 was further corroborated by showing that inhibition or disruption of NOX2 blocked subpicomolar SP-exacerbated neurotoxicity. Together, our findings revealed a critical role of microglial NOX2 in mediating the neuroinflammatory and dopaminergic neurodegenerative effects of SP, which may provide new insights into the pathogenesis of PD.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/metabolism , Microglia/metabolism , NADPH Oxidases/metabolism , Parkinsonian Disorders/metabolism , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Animals , Enzyme Activation , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology
9.
Glia ; 63(6): 1057-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25740080

ABSTRACT

Although the peripheral anti-inflammatory effect of norepinephrine (NE) is well documented, the mechanism by which this neurotransmitter functions as an anti-inflammatory/neuroprotective agent in the central nervous system (CNS) is unclear. This article aimed to determine the anti-inflammatory/neuroprotective effects and underlying mechanisms of NE in inflammation-based dopaminergic neurotoxicity models. In mice, NE-depleting toxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) was injected at 6 months of lipopolysaccharide (LPS)-induced neuroinflammation. It was found that NE depletion enhanced LPS-induced dopaminergic neuron loss in the substantia nigra. This piece of in vivo data prompted us to conduct a series of studies in an effort to elucidate the mechanism as to how NE affects dopamine neuron survival by using primary midbrain neuron/glia cultures. Results showed that submicromolar concentrations of NE dose-dependently protected dopaminergic neurons from LPS-induced neurotoxicity by inhibiting microglia activation and subsequent release of pro-inflammatory factors. However, NE-elicited neuroprotection was not totally abolished in cultures from ß2-adrenergic receptor (ß2-AR)-deficient mice, suggesting that novel pathways other than ß2-AR are involved. To this end, It was found that submicromolar NE dose-dependently inhibited NADPH oxidase (NOX2)-generated superoxide, which contributes to the anti-inflammatory and neuroprotective effects of NE. This novel mechanism was indeed adrenergic receptors independent since both (+) and (-) optic isomers of NE displayed the same potency. We further demonstrated that NE inhibited LPS-induced NOX2 activation by blocking the translocation of its cytosolic subunit to plasma membranes. In summary, we revealed a potential physiological role of NE in maintaining brain immune homeostasis and protecting neurons via a novel mechanism.


Subject(s)
Brain/immunology , Dopaminergic Neurons/immunology , Microglia/enzymology , NADPH Oxidases/metabolism , Norepinephrine/metabolism , Animals , Benzylamines/pharmacology , Brain/drug effects , Brain/pathology , COS Cells , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Chlorocebus aethiops , Coculture Techniques , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Homeostasis/physiology , Lipopolysaccharides/toxicity , Male , Mice, Inbred BALB C , Mice, Knockout , Microglia/drug effects , Microglia/pathology , Neurotransmitter Uptake Inhibitors/pharmacology , Rats, Inbred F344 , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism
10.
Clin Sci (Lond) ; 129(8): 757-67, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26223840

ABSTRACT

The distribution of microglia varies greatly throughout the brain. The substantia nigra (SN) contains the highest density of microglia among different brain regions. However, the mechanism underlying this uneven distribution remains unclear. Substance P (SP) is a potent proinflammatory neuropeptide with high concentrations in the SN. We recently demonstrated that SP can regulate nigral microglial activity. In the present study, we further investigated the involvement of SP in modulating nigral microglial density in postnatal developing mice. Nigral microglial density was quantified in wild-type (WT) and SP-deficient mice from postnatal day 1 (P1) to P30. SP was detected at high levels in the SN as early as P1 and microglial density did not peak until around P30 in WT mice. SP-deficient mice (TAC1(-/-)) had a significant reduction in nigral microglial density. No differences in the ability of microglia to proliferate were observed between TAC1(-/-) and WT mice, suggesting that SP may alter microglial density through chemotaxic recruitment. SP was confirmed to dose-dependently attract microglia using a trans-well culture system. Mechanistic studies revealed that both the SP receptor neurokinin-1 receptor (NK1R) and the superoxide-producing enzyme NADPH oxidase (NOX2) were necessary for SP-mediated chemotaxis in microglia. Furthermore, genetic ablation and pharmacological inhibition of NK1R or NOX2 attenuated SP-induced microglial migration. Finally, protein kinase Cδ (PKCδ) was recognized to couple SP/NK1R-mediated NOX2 activation. Altogether, we found that SP partly accounts for the increased density of microglia in the SN through chemotaxic recruitment via a novel NK1R-NOX2 axis-mediated pathway.


Subject(s)
Membrane Glycoproteins/metabolism , Microglia/physiology , NADPH Oxidases/metabolism , Receptors, Neurokinin-1/metabolism , Substance P/physiology , Substantia Nigra/cytology , Animals , Cell Movement , Cell Proliferation , Chemotaxis , Enzyme Activation , Mice, Inbred C57BL , Mice, Transgenic , NADPH Oxidase 2 , Protein Kinase C-delta/metabolism , Substantia Nigra/physiology , Superoxides/metabolism
11.
J Immunol ; 190(1): 115-25, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23209319

ABSTRACT

During viral infection, extracellular dsRNA is a potent signaling molecule that activates many innate immune cells, including macrophages. TLR3 is a well-known receptor for extracellular dsRNA, and internalization of extracellular dsRNA is required for endosomal TLR3 activation. Preserved inflammatory responses of TLR3-deficient macrophages to extracellular dsRNA strongly support a TLR3-independent mechanism in dsRNA-mediated immune responses. The present study demonstrated that CD11b/CD18 (Mac-1 [macrophage-1 Ag]), a surface integrin receptor, recognized extracellular dsRNA and induced macrophage immune responses. CD11b deficiency reduced inflammatory cytokine induction elicited by polyinosinic:polycytidylic acid (poly I:C; a synthetic dsRNA) in mouse sera and livers, as well as in cultured peritoneal macrophages. dsRNA-binding assay and confocal immunofluorescence showed that Mac-1, especially the CD11b subunit, interacted and colocalized with poly I:C on the surface of macrophages. Further mechanistic studies revealed two distinct signaling events following dsRNA recognition by Mac-1. First, Mac-1 facilitated poly I:C internalization through the activation of PI3K signaling and enhanced TLR3-dependent activation of IRF3 in macrophages. Second, poly I:C induced activation of phagocyte NADPH oxidase in a TLR3-independent, but Mac-1-dependent, manner. Subsequently, phagocyte NADPH oxidase-derived intracellular reactive oxygen species activated MAPK and NF-κB pathways. Our results indicate that extracellular dsRNA activates Mac-1 to enhance TLR3-dependent signaling and to trigger TLR3-independent, but Mac-1-dependent, inflammatory oxidative signaling, identifying a novel mechanistic basis for macrophages to recognize extracellular dsRNA to regulate innate immune responses. This study identifies Mac-1 as a novel surface receptor for extracellular dsRNA and implicates it as a potential therapeutic target for virus-related inflammatory diseases.


Subject(s)
CD11b Antigen/metabolism , CD18 Antigens/metabolism , Extracellular Space/genetics , Inflammation Mediators/physiology , Macrophage-1 Antigen/metabolism , RNA, Double-Stranded/physiology , Animals , CD11b Antigen/genetics , CD18 Antigens/genetics , Cell Line , Extracellular Space/immunology , Extracellular Space/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Macrophage-1 Antigen/genetics , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Knockout , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 3
12.
Glia ; 62(12): 2034-43, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25043383

ABSTRACT

Activation of microglial NADPH oxidase (NOX2) plays a critical role in mediating neuroinflammation, which is closely linked with the pathogenesis of a variety of neurodegenerative diseases, including Parkinson's disease (PD). The inhibition of NOX2-generated superoxide has become an effective strategy for developing disease-modifying therapies for PD. However, the lack of specific and potent NOX2 inhibitors has hampered the progress of this approach. Diphenyleneiodonium (DPI) is a widely used, long-acting NOX2 inhibitor. However, due to its non-specificity for NOX2 and high cytotoxicity at standard doses (µM), DPI has been precluded from human studies. In this study, using ultra-low doses of DPI, we aimed to: (1) investigate whether these problems could be circumvented and (2) determine whether ultra-low doses of DPI were able to preserve its utility as a potent NOX2 inhibitor. We found that DPI at subpicomolar concentrations (10(-14) and 10(-13) M) displays no toxicity in primary midbrain neuron-glia cultures. More importantly, we observed that subpicomolar DPI inhibited phorbol myristate acetate (PMA)-induced activation of NOX2. The same concentrations of DPI did not inhibit the activities of a series of flavoprotein-containing enzymes. Furthermore, potent neuroprotective efficacy was demonstrated in a post-treatment study. When subpicomolar DPI was added to neuron-glia cultures pretreated with lipopolysaccharide, 1-methyl-4-phenylpyridinium or rotenone, it potently protected the dopaminergic neurons. In summary, DPI's unique combination of high specificity toward NOX2, low cytotoxicity and potent neuroprotective efficacy in post-treatment regimens suggests that subpicomolar DPI may be an ideal candidate for further animal studies and potential clinical trials.


Subject(s)
Enzyme Inhibitors/pharmacology , Microglia/drug effects , NADPH Oxidases/metabolism , Onium Compounds/pharmacology , Animals , Calcium-Binding Proteins/metabolism , Cell Count , Cells, Cultured , Dopamine/metabolism , Electron Transport Complex I/metabolism , Glial Fibrillary Acidic Protein/metabolism , Mesencephalon/cytology , Microfilament Proteins/metabolism , NADPH-Ferrihemoprotein Reductase/metabolism , Neurons/drug effects , Neurons/physiology , Nitric Oxide Synthase Type II/metabolism , Nitrites/metabolism , Superoxides/metabolism , Thioredoxin-Disulfide Reductase/metabolism , Tyrosine 3-Monooxygenase/metabolism , Xanthine Oxidase/metabolism
13.
Antiviral Res ; 228: 105934, 2024 08.
Article in English | MEDLINE | ID: mdl-38880195

ABSTRACT

Herpes simplex virus type 1 (HSV-1), a neurotropic DNA virus, establishes latency in neural tissues, with reactivation causing severe consequences like encephalitis. Emerging evidence links HSV-1 infection to chronic neuroinflammation and neurodegenerative diseases. Microglia, the central nervous system's (CNS) immune sentinels, express diverse receptors, including α7 nicotinic acetylcholine receptors (α7 nAChRs), critical for immune regulation. Recent studies suggest α7 nAChR activation protects against viral infections. Here, we show that α7 nAChR agonists, choline and PNU-282987, significantly inhibit HSV-1 replication in microglial BV2 cells. Notably, this inhibition is independent of the traditional ionotropic nAChR signaling pathway. mRNA profiling revealed that choline stimulates the expression of antiviral factors, IL-1ß and Nos2, and down-regulates the apoptosis genes and type A Lamins in BV2 cells. These findings suggest a novel mechanism by which microglial α7 nAChRs restrict viral infections by regulating innate immune responses.


Subject(s)
Choline , Herpesvirus 1, Human , Microglia , Virus Replication , alpha7 Nicotinic Acetylcholine Receptor , alpha7 Nicotinic Acetylcholine Receptor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics , Microglia/virology , Microglia/drug effects , Microglia/metabolism , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/drug effects , Animals , Cell Line , Mice , Virus Replication/drug effects , Choline/pharmacology , Choline/metabolism , Bridged Bicyclo Compounds/pharmacology , Benzamides/pharmacology , Immunity, Innate , Herpes Simplex/virology , Herpes Simplex/metabolism , Interleukin-1beta/metabolism , Signal Transduction/drug effects , Apoptosis/drug effects , Antiviral Agents/pharmacology , Nicotinic Agonists/pharmacology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type II/genetics
14.
J Immunol ; 186(7): 4443-54, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21335487

ABSTRACT

The role of the ß2 adrenergic receptor (ß2AR) in the regulation of chronic neurodegenerative inflammation within the CNS is poorly understood. The purpose of this study was to determine neuroprotective effects of long-acting ß2AR agonists such as salmeterol in rodent models of Parkinson's disease. Results showed salmeterol exerted potent neuroprotection against both LPS and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine/1-methyl-4-phenylpyridinium-induced dopaminergic neurotoxicity both in primary neuron-glia cultures (at subnanomolar concentrations) and in mice (1-10 µg/kg/day doses). Further studies demonstrated that salmeterol-mediated neuroprotection is not a direct effect on neurons; instead, it is mediated through the inhibition of LPS-induced microglial activation. Salmeterol significantly inhibited LPS-induced production of microglial proinflammatory neurotoxic mediators, such as TNF-α, superoxide, and NO, as well as the inhibition of TAK1-mediated phosphorylation of MAPK and p65 NF-κB. The anti-inflammatory effects of salmeterol required ß2AR expression in microglia but were not mediated through the conventional G protein-coupled receptor/cAMP pathway. Rather, salmeterol failed to induce microglial cAMP production, could not be reversed by either protein kinase A inhibitors or an exchange protein directly activated by cAMP agonist, and was dependent on ß-arrestin2 expression. Taken together, our results demonstrate that administration of extremely low doses of salmeterol exhibit potent neuroprotective effects by inhibiting microglial cell activation through a ß2AR/ß-arrestin2-dependent but cAMP/protein kinase A-independent pathway.


Subject(s)
Adrenergic beta-2 Receptor Agonists/therapeutic use , Dopamine/toxicity , Microglia/immunology , Neural Inhibition/immunology , Neuroprotective Agents/therapeutic use , Signal Transduction/immunology , Adrenergic beta-2 Receptor Agonists/metabolism , Animals , Cells, Cultured , Coculture Techniques , Dopamine/biosynthesis , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/therapeutic use , Inflammation Mediators/toxicity , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/pathology , Neural Inhibition/drug effects , Neural Inhibition/genetics , Neuroprotective Agents/metabolism , Parkinson Disease/immunology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Rats , Rats, Inbred F344 , Signal Transduction/drug effects , Signal Transduction/genetics
15.
Front Immunol ; 14: 1083513, 2023.
Article in English | MEDLINE | ID: mdl-36845109

ABSTRACT

Clinical and pathological evidence revealed that α-synuclein (α-syn) pathology seen in PD patients starts in the gut and spreads via anatomically connected structures from the gut to the brain. Our previous study demonstrated that depletion of central norepinephrine (NE) disrupted brain immune homeostasis, producing a spatiotemporal order of neurodegeneration in the mouse brain. The purpose of this study was 1) to determine the role of peripheral noradrenergic system in the maintenance of gut immune homeostasis and in the pathogenesis of PD and 2) to investigate whether NE-depletion induced PD-like α-syn pathological changes starts from the gut. For these purposes, we investigated time-dependent changes of α-synucleinopathy and neuronal loss in the gut following a single injection of DSP-4 (a selective noradrenergic neurotoxin) to A53T-SNCA (human mutant α-syn) over-expression mice. We found DPS-4 significantly reduced the tissue level of NE and increased immune activities in gut, characterized by increased number of phagocytes and proinflammatory gene expression. Furthermore, a rapid-onset of α-syn pathology was observed in enteric neurons after 2 weeks and delayed dopaminergic neurodegeneration in the substantia nigra was detected after 3-5 months, associated with the appearance of constipation and impaired motor function, respectively. The increased α-syn pathology was only observed in large, but not in the small, intestine, which is similar to what was observed in PD patients. Mechanistic studies reveal that DSP-4-elicited upregulation of NADPH oxidase (NOX2) initially occurred only in immune cells during the acute intestinal inflammation stage, and then spread to enteric neurons and mucosal epithelial cells during the chronic inflammation stage. The upregulation of neuronal NOX2 correlated well with the extent of α-syn aggregation and subsequent enteric neuronal loss, suggesting that NOX2-generated reactive oxygen species play a key role in α-synucleinopathy. Moreover, inhibiting NOX2 by diphenyleneiodonium or restoring NE function by salmeterol (a ß2-receptor agonist) significantly attenuated colon inflammation, α-syn aggregation/propagation, and enteric neurodegeneration in the colon and ameliorated subsequent behavioral deficits. Taken together, our model of PD shows a progressive pattern of pathological changes from the gut to the brain and suggests a potential role of the noradrenergic dysfunction in the pathogenesis of PD.


Subject(s)
Synucleinopathies , Humans , Animals , Mice , Inflammation/pathology , Norepinephrine/metabolism , Colon/pathology
16.
J Neurosci ; 31(3): 1081-92, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21248133

ABSTRACT

What drives the gradual degeneration of dopamine neurons in Parkinson's disease (PD), the second most common neurodegenerative disease, remains elusive. Here, we demonstrated, for the first time, that persistent neuroinflammation was indispensible for such a neurodegenerative process. 1-Methyl-4-phenylpyridinium, lipopolysaccharide (LPS), and rotenone, three toxins often used to create PD models, produced acute but nonprogressive neurotoxicity in neuron-enriched cultures. In the presence of microglia (brain immune cells), these toxins induced progressive dopaminergic neurodegeneration. More importantly, such neurodegeneration was prevented by removing activated microglia. Collectively, chronic neuroinflammation may be a driving force of progressive dopaminergic neurodegeneration. Conversely, ongoing neurodegeneration sustained microglial activation. Microglial activation persisted only in the presence of neuronal damage in LPS-treated neuron-glia cultures but not in LPS-treated mixed-glia cultures. Thus, activated microglia and damaged neurons formed a vicious cycle mediating chronic, progressive neurodegeneration. Mechanistic studies indicated that HMGB1 (high-mobility group box 1), released from inflamed microglia and/or degenerating neurons, bound to microglial Mac1 (macrophage antigen complex 1) and activated nuclear factor-κB pathway and NADPH oxidase to stimulate production of multiple inflammatory and neurotoxic factors. The treatment of microglia with HMGB1 led to membrane translocation of p47(phox) (a cytosolic subunit of NADPH oxidase) and consequent superoxide release, which required the presence of Mac1. Neutralization of HMGB1 and genetic ablation of Mac1 and gp91(phox) (the catalytic submit of NADPH oxidase) blocked the progressive neurodegeneration. Our findings indicated that HMGB1-Mac1-NADPH oxidase signaling axis bridged chronic neuroinflammation and progressive dopaminergic neurodegeneration, thus identifying a mechanistic basis for chronic PD progression.


Subject(s)
HMGB1 Protein/metabolism , Inflammation/metabolism , Macrophage-1 Antigen/metabolism , Microglia/metabolism , Nerve Degeneration/metabolism , Analysis of Variance , Animals , Blotting, Western , Cell Count , Cell Fractionation , Cells, Cultured , Coculture Techniques , Dopamine/metabolism , Inflammation/chemically induced , Inflammation/pathology , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Mesencephalon/cytology , Mesencephalon/metabolism , Microglia/cytology , Microglia/pathology , NADPH Oxidases/metabolism , Nerve Degeneration/pathology , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Rats , Rats, Inbred F344 , Signal Transduction
17.
Biochem Biophys Res Commun ; 427(3): 518-24, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23022187

ABSTRACT

Ethyl acetate (EA) is an ordinary organic compound in fruits, wine and cosmetics, and used as a solvent frequently. With the recent observation in our experiment, we suspected that EA could affect immune function, in particular macrophage activity. In this paper, we tested EA's protect effect against death in Lipopolysaccharide/D-galactosamine (LPS/D-GalN)-induced endotoxic shock model in mice. And also found EA decreased the LPS-induced mRNA expression of mediators of inflammation including cyclooxygenase 2 (COX2), inducible NOS (iNOS), and tumor necrosis factor α (TNF α) in RAW264.7 cells. Consequently, EA decreased the production of, TNF α and the inflammatory agent nitric oxide (NO) in RAW264.7 cells treated with LPS. Other pro-inflammatory cytokines such as IL-1h and IL-6 were similarly decreased by EA treatment of RAW264.7 cells. The potential mechanism may associate with NF-κB activity as we shown. Taken together, these results suggest that EA has anti-inflammatory properties.


Subject(s)
Acetates/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Shock, Septic/drug therapy , Animals , Cell Line , Disease Models, Animal , Galactosamine/immunology , Gene Expression/drug effects , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred Strains , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Protein Transport , Shock, Septic/immunology , Tumor Necrosis Factor-alpha/biosynthesis
18.
J Neuroinflammation ; 9: 32, 2012 Feb 16.
Article in English | MEDLINE | ID: mdl-22340895

ABSTRACT

BACKGROUND: Both (-) and (+)-naloxone attenuate inflammation-mediated neurodegeneration by inhibition of microglial activation through superoxide reduction in an opioid receptor-independent manner. Multiple lines of evidence have documented a pivotal role of overactivated NADPH oxidase (NOX2) in inflammation-mediated neurodegeneration. We hypothesized that NOX2 might be a novel action site of naloxone to mediate its anti-inflammatory actions. METHODS: Inhibition of NOX-2-derived superoxide by (-) and (+)-naloxone was measured in lipopolysaccharide (LPS)-treated midbrain neuron-glia cultures and phorbol myristate acetate (PMA)-stimulated neutrophil membranes by measuring the superoxide dismutase (SOD)-inhibitable reduction of tetrazolium salt (WST-1) or ferricytochrome c. Further, various ligand (3H-naloxone) binding assays were performed in wild type and gp91phox-/- neutrophils and transfected COS-7 and HEK293 cells. The translocation of cytosolic subunit p47phox to plasma membrane was assessed by western blot. RESULTS: Both (-) and (+)-naloxone equally inhibited LPS- and PMA-induced superoxide production with an IC50 of 1.96 and 2.52 µM, respectively. Competitive binding of 3H-naloxone with cold (-) and (+)-naloxone in microglia showed equal potency with an IC50 of 2.73 and 1.57 µM, respectively. 3H-Naloxone binding was elevated in COS-7 and HEK293 cells transfected with gp91phox; in contrast, reduced 3H-naloxone binding was found in neutrophils deficient in gp91phox or in the presence of a NOX2 inhibitor. The specificity and an increase in binding capacity of 3H-naloxone were further demonstrated by 1) an immunoprecipitation study using gp91phox antibody, and 2) activation of NOX2 by PMA. Finally, western blot studies showed that naloxone suppressed translocation of the cytosolic subunit p47phox to the membrane, leading to NOX2 inactivation. CONCLUSIONS: Strong evidence is provided indicating that NOX2 is a non-opioid novel binding site for naloxone, which is critical in mediating its inhibitory effect on microglia overactivation and superoxide production.


Subject(s)
Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Naloxone/pharmacology , Neuroglia/metabolism , Neurons/metabolism , Superoxides/metabolism , Analysis of Variance , Animals , Biphenyl Compounds/pharmacology , Cell Membrane/drug effects , Cell Membrane/genetics , Cell Membrane/metabolism , Cells, Cultured , Chlorocebus aethiops , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Humans , Immunoprecipitation , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/deficiency , Mesencephalon/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/deficiency , Neuroglia/drug effects , Neurons/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Onium Compounds/pharmacology , Superoxide Dismutase/metabolism , Tetradecanoylphorbol Acetate , Tetrazolium Salts/pharmacology , Thyroid Function Tests/methods , Transfection , Tritium/pharmacokinetics , Xanthine Oxidase/pharmacology
19.
J Neuroinflammation ; 8(1): 3, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21232086

ABSTRACT

BACKGROUND: ß-Amyloid peptide (Aß) is a major protein in the brain associated with Alzheimer's and Parkinson's diseases. The purpose of this study was to investigate the role of macrophage antigen-1 (MAC1) receptor, an integrin scavenger receptor in microglia, and subsequent signaling events in mediating Aß-induced neurotoxicity. We have previously reported that NADPH oxidase (PHOX) on microglia and superoxide produced by PHOX are critical for Aß-induced loss of dopaminergic neurons. However, the upstream signaling pathway of superoxide production remains unclear. METHODS: For the in vitro study, mesencephalic neuron-glia cultures and microglia-enriched cultures from mice deficient in the MAC1 receptor (MAC1-/-) and wild type controls were used to investigate the role of MAC1 receptor in Aß-induced neurotoxicity and the role of phosphoinositide-3 kinase (PI3K) in the signal pathway between MAC1 receptor and PHOX. For the in vivo study, Aß was injected into the substantia nigra of MAC1-/- mice and wild type mice to confirm the role of MAC1 receptor. RESULTS: We found that Aß-induced activation of microglia, activation of PHOX, generation of superoxide and other reactive oxygen species, and loss of dopaminergic neurons were decreased in MAC1-/- cultures compared to MAC1+/+ cultures. In MAC1-/- mice, dopaminergic neuron loss in response to Aß injection into the substantia nigra was reduced relative to MAC1+/+ mice. Thus, MAC1 receptor-mediated PHOX activation and increased superoxide production are associated with Aß-induced neurotoxicity. PI3K activation was one downstream step in MAC1 signaling to PHOX and played an important role in Aß-induced neurotoxicity. In microglia-enriched cultures from MAC1-/- mice, Aß-induced activation of PI3K (phosphorylation of target proteins and PIP3 production) was reduced relative to MAC1+/+ cultures. CONCLUSIONS: Taken together, our data demonstrate that Aß activates MAC1 receptor to increase the activity of PI3K, which in turn phosphorylates p47phox, triggers the translocation of cytosolic subunits of PHOX to microglia membrane, increases PHOX activation and the subsequent production of superoxide and causes neurotoxicity.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Macrophage-1 Antigen/metabolism , Microglia/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cells, Cultured , Dopamine/metabolism , Enzyme Activation , Macrophage-1 Antigen/genetics , Male , Mesencephalon/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neurons/cytology , Neurons/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , gamma-Aminobutyric Acid/metabolism
20.
Brain ; 133(Pt 3): 808-21, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20123724

ABSTRACT

Microglia, the innate immune cells in the brain, can become chronically activated in response to dopaminergic neuron death, fuelling a self-renewing cycle of microglial activation followed by further neuron damage (reactive microgliosis), which is implicated in the progressive nature of Parkinson's disease. Here, we use an in vitro approach to separate neuron injury factors from the cellular actors of reactive microgliosis and discover molecular signals responsible for chronic and toxic microglial activation. Upon injury with the dopaminergic neurotoxin 1-methyl-4-phenylpyridinium, N27 cells (dopaminergic neuron cell line) released soluble neuron injury factors that activated microglia and were selectively toxic to dopaminergic neurons in mixed mesencephalic neuron-glia cultures through nicotinamide adenine dinucleotide phosphate oxidase. mu-Calpain was identified as a key signal released from damaged neurons, causing selective dopaminergic neuron death through activation of microglial nicotinamide adenine dinucleotide phosphate oxidase and superoxide production. These findings suggest that dopaminergic neurons may be inherently susceptible to the pro-inflammatory effects of neuron damage, i.e. reactive microgliosis, providing much needed insight into the chronic nature of Parkinson's disease.


Subject(s)
Calpain/metabolism , Dopamine/metabolism , Gliosis/physiopathology , Microglia/physiology , Neurons/physiology , Animals , Cell Death/physiology , Cell Line , Cells, Cultured , Coculture Techniques , Female , Male , Mesencephalon/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Rats , Rats, Inbred F344 , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL