Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters











Publication year range
1.
J Neuroinflammation ; 21(1): 216, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39218899

ABSTRACT

Parkinson's disease (PD) is characterized by neuroinflammation, progressive loss of dopaminergic neurons, and accumulation of α-synuclein (α-Syn) into insoluble aggregates called Lewy pathology. The Line 61 α-Syn mouse is an established preclinical model of PD; Thy-1 is used to promote human α-Syn expression, and features of sporadic PD develop at 9-18 months of age. To accelerate the PD phenotypes, we injected sonicated human α-Syn preformed fibrils (PFFs) into the striatum, which produced phospho-Syn (p-α-Syn) inclusions in the substantia nigra pars compacta and significantly increased MHC Class II-positive immune cells. Additionally, there was enhanced infiltration and activation of innate and adaptive immune cells in the midbrain. We then used this new model, Line 61-PFF, to investigate the effect of inhibiting the JAK/STAT signaling pathway, which is critical for regulation of innate and adaptive immune responses. After administration of the JAK1/2 inhibitor AZD1480, immunofluorescence staining showed a significant decrease in p-α-Syn inclusions and MHC Class II expression. Flow cytometry showed reduced infiltration of CD4+ T-cells, CD8+ T-cells, CD19+ B-cells, dendritic cells, macrophages, and endogenous microglia into the midbrain. Importantly, single-cell RNA-Sequencing analysis of CD45+ cells from the midbrain identified 9 microglia clusters, 5 monocyte/macrophage (MM) clusters, and 5 T-cell (T) clusters, in which potentially pathogenic MM4 and T3 clusters were associated with neuroinflammatory responses in Line 61-PFF mice. AZD1480 treatment reduced cell numbers and cluster-specific expression of the antigen-presentation genes H2-Eb1, H2-Aa, H2-Ab1, and Cd74 in the MM4 cluster and proinflammatory genes such as Tnf, Il1b, C1qa, and C1qc in the T3 cluster. Together, these results indicate that inhibiting the JAK/STAT pathway suppresses the activation and infiltration of innate and adaptive cells, reducing neuroinflammation in the Line 61-PFF mouse model.


Subject(s)
Disease Models, Animal , Neuroinflammatory Diseases , Parkinson Disease , STAT Transcription Factors , Signal Transduction , alpha-Synuclein , Animals , Mice , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , STAT Transcription Factors/metabolism , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/immunology , Humans , Mice, Transgenic , Mice, Inbred C57BL , Janus Kinases/metabolism , Janus Kinases/antagonists & inhibitors , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Parkinsonian Disorders/immunology , Pyrimidines/pharmacology
3.
Acta Neuropathol Commun ; 9(1): 139, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34412704

ABSTRACT

BACKGROUND: The etiology of sporadic Parkinson's disease (PD) remains uncertain, but genetic, epidemiological, and physiological overlap between PD and inflammatory bowel disease suggests that gut inflammation could promote dysfunction of dopamine-producing neurons in the brain. Mechanisms behind this pathological gut-brain effect and their interactions with sex and with environmental factors are not well understood but may represent targets for therapeutic intervention. METHODS: We sought to identify active inflammatory mechanisms which could potentially contribute to neuroinflammation and neurological disease in colon biopsies and peripheral blood immune cells from PD patients. Then, in mouse models, we assessed whether dextran sodium sulfate-mediated colitis could exert lingering effects on dopaminergic pathways in the brain and whether colitis increased vulnerability to a subsequent exposure to the dopaminergic neurotoxicant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). We assessed the involvement of inflammatory mechanisms identified in the PD patients in colitis-related neurological dysfunction in male and female mice, utilizing mice lacking the Regulator of G-Protein Signaling 10 (RGS10)-an inhibitor of nuclear factor kappa B (NFκB)-to model enhanced NFκB activity, and mice in which CD8+ T-cells were depleted. RESULTS: High levels of inflammatory markers including CD8B and NFκB p65 were found in colon biopsies from PD patients, and reduced levels of RGS10 were found in immune cells in the blood. Male mice that experienced colitis exhibited sustained reductions in tyrosine hydroxylase but not in dopamine as well as sustained CD8+ T-cell infiltration and elevated Ifng expression in the brain. CD8+ T-cell depletion prevented colitis-associated reductions in dopaminergic markers in males. In both sexes, colitis potentiated the effects of MPTP. RGS10 deficiency increased baseline intestinal inflammation, colitis severity, and neuropathology. CONCLUSIONS: This study identifies peripheral inflammatory mechanisms in PD patients and explores their potential to impact central dopaminergic pathways in mice. Our findings implicate a sex-specific interaction between gastrointestinal inflammation and neurologic vulnerability that could contribute to PD pathogenesis, and they establish the importance of CD8+ T-cells in this process in male mice.


Subject(s)
Brain/metabolism , CD8-Positive T-Lymphocytes/immunology , Colitis/immunology , Neuroinflammatory Diseases/immunology , Parkinson Disease/immunology , Parkinsonian Disorders/immunology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Brain/pathology , CD8 Antigens/metabolism , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Colon/metabolism , Dextran Sulfate , Dopamine/metabolism , Dopamine Agents , Female , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interferon-gamma/metabolism , Male , Mice , Mice, Knockout , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/metabolism , Parkinson Disease/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , RGS Proteins/genetics , RGS Proteins/metabolism , Sex Factors , Transcription Factor RelA/metabolism , Tyrosine 3-Monooxygenase/metabolism
4.
Aging (Albany NY) ; 13(8): 11455-11469, 2021 04 04.
Article in English | MEDLINE | ID: mdl-33839699

ABSTRACT

BACKGROUND: Studies have revealed that lncRNA HOXA11-AS contributes to regulating inflammation, while the role of HOXA11-AS in Parkinson's disease (PD) remains unclear. METHODS: Both in vivo and in vitro PD models were induced. Gain- or loss-assays of HOXA11-AS and miR-124-3p were conducted. The neurological functions, dopaminergic neurons damage, microglia activation of PD mice were measured. Afterwards, the expressions of inflammatory factors were examined with RT-PCR. Western blot was employed to detect the level of FSTL1, NF-κB and NLRP3 inflammasome. Meanwhile, bioinformatics analysis and dual-luciferase reporter assay were utilized to confirm the targeting relationships among miR-124-3p, HOXA11-AS and FSTL1. RESULTS: HOXA11-AS promoted MPTP-mediated SH-SY5Y neuronal injury and LPS-induced microglia activation, while miR-124-3p had the opposite effects. Additionally, miR-124-3p was the target of HOXA11-AS and FSTL1. HOXA11-AS overexpression enhanced the expression of inflammatory factors and FSTL1, NF-κB and NLRP3 inflammasome, while inhibiting NF-κB weakened HOXA11-AS-mediated neuronal damage and microglia activation. Moreover, HOXA11-AS1 downregulation ameliorated MPTP-induced neurological damages and neuroinflammation in mice. CONCLUSION: Inhibition of HOXA11-AS protects mice against PD through repressing neuroinflammation and neuronal apoptosis through miR-124-3p-FSTL1-NF-κB axis.


Subject(s)
Dopaminergic Neurons/pathology , MicroRNAs/metabolism , Parkinsonian Disorders/immunology , RNA, Long Noncoding/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Animals , Apoptosis/genetics , Apoptosis/immunology , Disease Models, Animal , Dopaminergic Neurons/immunology , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Male , Mice , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Signal Transduction/genetics , Signal Transduction/immunology
5.
J Neuroinflammation ; 18(1): 20, 2021 Jan 09.
Article in English | MEDLINE | ID: mdl-33422110

ABSTRACT

BACKGROUND: Recent work has established that Parkinson's disease (PD) patients have an altered gut microbiome, along with signs of intestinal inflammation. This could help explain the high degree of gastric disturbances in PD patients, as well as potentially be linked to the migration of peripheral inflammatory factors into the brain. To our knowledge, this is the first study to examine microbiome alteration prior to the induction of a PD murine model. METHODS: We presently assessed whether pre-treatment with the probiotic, VSL #3, or the inflammatory inducer, dextran sodium sulphate (DSS), would influence the PD-like pathology provoked by a dual hit toxin model using lipopolysaccharide (LPS) and paraquat exposure. RESULTS: While VSL #3 has been reported to have anti-inflammatory effects, DSS is often used as a model of colitis because of the gut inflammation and the breach of the intestinal barrier that it induces. We found that VSL#3 did not have any significant effects (beyond a blunting of LPS paraquat-induced weight loss). However, the DSS treatment caused marked changes in the gut microbiome and was also associated with augmented behavioral and inflammatory outcomes. In fact, DSS markedly increased taxa belonging to the Bacteroidaceae and Porphyromonadaceae families but reduced those from Rikencellaceae and S24-7, as well as provoking colonic pro-inflammatory cytokine expression, consistent with an inflamed gut. The DSS also increased the impact of LPS plus paraquat upon microglial morphology, along with circulating lipocalin-2 (neutrophil marker) and IL-6. Yet, neither DSS nor VSL#3 influenced the loss of substantia nigra dopamine neurons or the astrocytic and cytoskeleton remodeling protein changes that were provoked by the LPS followed by paraquat treatment. CONCLUSIONS: These data suggest that disruption of the intestinal integrity and the associated microbiome can interact with systemic inflammatory events to promote widespread brain-gut changes that could be relevant for PD and at the very least, suggestive of novel neuro-immune communication.


Subject(s)
Dextran Sulfate/administration & dosage , Disease Models, Animal , Gastrointestinal Microbiome/physiology , Inflammation Mediators/metabolism , Parkinsonian Disorders/metabolism , Probiotics/administration & dosage , Animals , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/immunology , Dopaminergic Neurons/metabolism , Gastrointestinal Microbiome/drug effects , Inflammation Mediators/immunology , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/immunology
6.
J Neuroimmune Pharmacol ; 16(2): 470-482, 2021 06.
Article in English | MEDLINE | ID: mdl-32671618

ABSTRACT

MP-10 (PF-2545920) is a selective inhibitor of phosphodiesterase 10A (PDE10A), an enzyme highly enriched in the striatum, nucleus accumbens, olfactory tubercle, and substantia nigra. The therapeutic effect of MP-10 has been reported in psychiatric and neurodegenerative disorders such as schizophrenia, depression, and Huntington's disease. However, the effect of MP-10 in Parkinson's disease (PD) has not been reported to date. In this study, we examined the effect of MP-10 in neuroinflammation and PD mouse models. MP-10 inhibited nitric oxide, tumor necrosis factor alpha, and interleukin (IL)-6 production, while it promoted IL-10 production in lipopolysaccharide (LPS)-stimulated BV2 microglial cells. Subsequent western blot and reverse transcription polymerase chain reaction analyses showed that MP-10 reduced the mRNA and protein levels of inducible nitric oxide synthase, cyclooxygenase-2, proinflammatory cytokines, and matrix metalloproteinase-3, -8, and - 9 in LPS-stimulated BV2 cells. Further mechanistic studies revealed that MP-10 exerts anti-inflammatory effects by inhibiting the phosphorylation of c-Jun N-terminal kinase and Akt, reducing the activity of nuclear factor-kappa B/activator protein-1, and upregulating the nuclear factor erythroid 2-related factor 2/antioxidant response element and protein kinase A/cAMP response element-binding protein signaling pathways. The anti-inflammatory effect of MP-10 was confirmed in vivo. Specifically, MP-10 inhibited microglial activation and proinflammatory gene expression in the brains of LPS-injected mice. Moreover, MP-10 rescued behavioral deficits and recovered dopaminergic neuronal cell death in the brains of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced PD mice. MP-10 also reduced microglial activation in this PD mouse model. These data collectively suggest that MP-10 may have therapeutic potential in PD and other neuroinflammatory disorders. Graphical Abstract.


Subject(s)
Brain/drug effects , Inflammation/immunology , Microglia/drug effects , Parkinsonian Disorders/immunology , Pyrazoles/pharmacology , Quinolines/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Brain/metabolism , Inflammation/chemically induced , Lipopolysaccharides/immunology , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases
7.
J Neuroimmune Pharmacol ; 16(2): 454-469, 2021 06.
Article in English | MEDLINE | ID: mdl-32648088

ABSTRACT

Deep brain stimulation of the subthalamic nucleus (DBS-STN) is an effective treatment for advanced motor symptoms of Parkinson's disease (PD). Recently, a connection between the limbic part of the STN and side effects of DBS-STN has been increasingly recognized. Animal studies have shown that DBS-STN influences behavior and provokes neurochemical changes in regions of the limbic system. Some of these regions, which are activated during DBS-STN, are involved in neuroimmunomodulation. The therapeutic effects of DBS-STN in PD treatment are clear, but the influence of DBS-STN on peripheral immunity has not been reported so far. In this study, we examined the effects of unilateral DBS-STN applied in male Wistar rats with 6-hydroxydopamine PD model (DBS-6OHDA) and rats without nigral dopamine depletion (DBS) on corticosterone (CORT) plasma concentration, blood natural killer cell cytotoxicity (NKCC), leukocyte numbers, lymphocyte population and apoptosis numbers, plasma interferon gamma (IFN-γ), interleukin 6 (IL-6), and tumor necrosis factor (TNF-α) concentration. The same peripheral immune parameters we measured also in non-stimulated rats with PD model (6OHDA). We observed peripheral immunity changes related to PD model. The NKCC and percentage of T cytotoxic lymphocytes were enhanced, while the level of lymphocyte apoptosis was down regulated in 6OHDA and DBS-6OHDA groups. After DBS-STN (DBS-6OHDA and DBS groups), the plasma CORT and TNF-α were elevated, the number of NK cells and percentage of apoptosis were increased, while the number of B lymphocytes was decreased. We also found, changes in plasma IFN-γ and IL-6 levels in all the groups. These results suggest potential peripheral immunomodulative effects of DBS-STN in the rat model of PD. However, further studies are necessary to explain these findings and their clinical implication. Graphical Abstract Influence of deep brain stimulation of the subthalamic nucleus on peripheral immunity in rat model of Parkinson's disease.


Subject(s)
Corticosterone/blood , Neuroimmunomodulation/physiology , Parkinsonian Disorders/immunology , Subthalamic Nucleus/physiology , Animals , Deep Brain Stimulation , Male , Rats , Rats, Wistar
8.
Int Immunopharmacol ; 91: 107269, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33340781

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease with complicated pathogenesis. A novel bibenzyl compound 2-[4-hydroxy-3-(4-hydroxyphenyl)benzyl]-4-(4-hydroxyphenyl)phenol (20C) has been shown to have some neuroprotective effects, and its mechanism still needs further research. In this study, we used a 6-hydroxydopamine (6-OHDA)-induced PD rat model to evaluate the protective effect of 20C. Our study found that 20C could improve behavioral defects in 6-OHDA-lesion rats, decrease neuroinflammation and protect their DA neurons. It could inhibit the activity of inducible nitric oxide synthase (iNOS) induced by 6-OHDA, and lead to a decrease in the expression of nitrated-α-synuclein. When exposed to AMT-an inhibitor of iNOS, the nitrated-α-synuclein in PC12 decreased, and 20C demonstrated the same function on nitrated-α-synuclein as AMT. Besides, we also found that nitrated-α-synuclein was displayed in microglia. And 20C could decrease the expression of antigen-presenting molecule major histocompatibility complex I (MHC I) in dopamine (DA) neurons and MHC II in microglia induced by 6-OHDA. So, these imply that nitrated-α-synuclein might act as an endogenous antigen activating adaptive immunity, and the neuroprotection of 20C might be associated with inhibiting the activity of iNOS, decreasing the expression of the antigen molecule nitrated-α-synuclein and the antigen presenting molecule MHC. Our results indicated that inhibiting iNOS might be an effective strategy to protect neurons from oxidative stress.


Subject(s)
Bibenzyls/pharmacology , Brain/drug effects , Dopaminergic Neurons/drug effects , Microglia/drug effects , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Animals , Antioxidants/pharmacology , Brain/immunology , Brain/metabolism , Brain/pathology , Cytokines/metabolism , Disease Models, Animal , Dopaminergic Neurons/immunology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Inflammation Mediators/metabolism , Male , Microglia/immunology , Microglia/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Oxidopamine , PC12 Cells , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/immunology , Parkinsonian Disorders/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , alpha-Synuclein/metabolism
10.
Bull Exp Biol Med ; 169(4): 431-434, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32889565

ABSTRACT

We studied the effect of single and repeated intranasal administration of antibodies to glutamate in experimental parkinsonian syndrome induced by injections of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to C57BL/6J mice. Intranasal administration of anti-glutamate antibodies to mice in parallel with administration of MPTP over 10 days alleviated parkinsonian symptoms (oligokinesia and rigidity). In the serum of mice injected with antibodies to glutamate and/or MPTP, the titers of autoantibodies to glutamate and dopamine were higher than in control animals receiving saline. Single intranasal administration of anti-glutamate antibodies to mice with established parkinsonian syndrome did not affect the severity of parkinsonian symptoms.


Subject(s)
Antibodies/pharmacology , Antiparkinson Agents/pharmacology , Dopamine/immunology , Glutamic Acid/immunology , Hypokinesia/drug therapy , Parkinsonian Disorders/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Administration, Intranasal , Animals , Antibodies/chemistry , Antibodies/isolation & purification , Antiparkinson Agents/chemistry , Antiparkinson Agents/isolation & purification , Autoantibodies/biosynthesis , Dopamine/chemistry , Glutamic Acid/chemistry , Horses , Hypokinesia/chemically induced , Hypokinesia/immunology , Hypokinesia/physiopathology , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Male , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/immunology , Parkinsonian Disorders/physiopathology , Rabbits , gamma-Globulins/chemistry , gamma-Globulins/immunology
11.
Article in English | MEDLINE | ID: mdl-32817412

ABSTRACT

OBJECTIVE: To develop a diagnostic model based on plasma-derived extracellular vesicle (EV) subpopulations in Parkinson disease (PD) and atypical parkinsonism (AP), we applied an innovative flow cytometric multiplex bead-based platform. METHODS: Plasma-derived EVs were isolated from PD, matched healthy controls, multiple system atrophy (MSA), and AP with tauopathies (AP-Tau). The expression levels of 37 EV surface markers were measured by flow cytometry and correlated with clinical scales. A diagnostic model based on EV surface markers expression was built via supervised machine learning algorithms and validated in an external cohort. RESULTS: Distinctive pools of EV surface markers related to inflammatory and immune cells stratified patients according to the clinical diagnosis. PD and MSA displayed a greater pool of overexpressed immune markers, suggesting a different immune dysregulation in PD and MSA vs AP-Tau. The receiver operating characteristic curve analysis of a compound EV marker showed optimal diagnostic performance for PD (area under the curve [AUC] 0.908; sensitivity 96.3%, specificity 78.9%) and MSA (AUC 0.974; sensitivity 100%, specificity 94.7%) and good accuracy for AP-Tau (AUC 0.718; sensitivity 77.8%, specificity 89.5%). A diagnostic model based on EV marker expression correctly classified 88.9% of patients with reliable diagnostic performance after internal and external validations. CONCLUSIONS: Immune profiling of plasmatic EVs represents a crucial step toward the identification of biomarkers of disease for PD and AP.


Subject(s)
Extracellular Vesicles/immunology , Parkinsonian Disorders/diagnosis , Parkinsonian Disorders/immunology , Tauopathies/diagnosis , Tauopathies/immunology , Aged , Aged, 80 and over , Antigens, Surface , Biomarkers/blood , Case-Control Studies , Cross-Sectional Studies , Female , Flow Cytometry , Humans , Male , Middle Aged , Multiple System Atrophy/blood , Multiple System Atrophy/classification , Multiple System Atrophy/diagnosis , Multiple System Atrophy/immunology , Parkinson Disease/blood , Parkinson Disease/classification , Parkinson Disease/diagnosis , Parkinson Disease/immunology , Parkinsonian Disorders/blood , Parkinsonian Disorders/classification , Protein Interaction Maps , Sensitivity and Specificity , Supervised Machine Learning , Tauopathies/blood , Tauopathies/classification
12.
J Med Case Rep ; 14(1): 84, 2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32600450

ABSTRACT

BACKGROUND: Immunological causes of parkinsonism are very rare and usually characterized by early presentation, poor response to levodopa, and additional clinical features. CASE PRESENTATION: We describe a 58-year-old white man who presented with a 1-year history of gait disturbance with disequilibrium leading to falls. We report an association between parkinsonism and presence of anti-glutamic acid decarboxylase antibodies in his cerebrospinal fluid, discussing clinical presentation and follow-up. CONCLUSIONS: Besides the possibility of a casual association, this case allows us to hypothesize an alternative pathophysiological mechanism of parkinsonism implying interference with glutamic acid decarboxylase and gamma-aminobutyric acid functions, eventually resulting in basal ganglia circuit dysregulation.


Subject(s)
Autoantibodies/cerebrospinal fluid , Glutamate Decarboxylase/immunology , Parkinsonian Disorders/immunology , Humans , Male , Middle Aged
13.
Exp Neurol ; 330: 113315, 2020 08.
Article in English | MEDLINE | ID: mdl-32302678

ABSTRACT

Parkinson's disease (PD), a debilitating progressive degenerative movement disorder associated with loss of dopaminergic (DA) neurons in the substantia nigra (SN), afflicts approximately one million people in the U.S., including a significant number of Veterans. Disease characteristics include tremor, rigidity, postural instability, bradykinesia, and at a cellular level, glial cell activation and Lewy body inclusions in DA neurons. The most potent medical/surgical treatments do not ultimately prevent disease progression. Therefore, new therapies must be developed to halt progression of the disease. While the mechanisms of the degenerative process in PD remain elusive, chronic inflammation, a common factor in many neurodegenerative diseases, has been implicated with associated accumulation of toxic aggregated α-synuclein in neurons. Calpain, a calcium-activated cysteine neutral protease, plays a pivotal role in SN and spinal cord degeneration in PD via its role in α-synuclein aggregation, activation/migration of microglia and T cells, and upregulation of inflammatory processes. Here we report an increased expression of a subset of CD4+ T cells in rodent models of PD, including MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) mice and DSP-4 [N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride]/6-hydroxydopamine rats, which produced higher levels of perforin and granzyme B - typically found in cytotoxic T cells. Importantly, the CD4+ cytotoxic subtype was attenuated following calpain inhibition in MPTP mice, suggesting that calpain and this distinct CD4+ T cell subset may have critical roles in the inflammatory process, disease progression, and neurodegeneration in PD.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Calpain/immunology , Parkinsonian Disorders/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Calpain/metabolism , Disease Models, Animal , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/pathology , Rats , Rats, Long-Evans , T-Lymphocyte Subsets/immunology
14.
J Neuroimmune Pharmacol ; 15(2): 224-237, 2020 06.
Article in English | MEDLINE | ID: mdl-31802419

ABSTRACT

Regulatory T cells (Tregs), which secrete transforming growth factor (TGF)-ß and interleukin (IL)-10, have essential role in anti-inflammatory and neurotrophic functions. Herein, we explore the neuroprotection of Tregs in Parkinson's disease (PD) by adoptive transfer of Tregs. Tregs, isolated by magnetic sorting, were activated in vitro and then were adoptively transferred to 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP)-treated mice. Neuroinflammation, dopaminergic neuronal loss and behavioral changes of PD mice were evaluated. Live cell imaging system detected a dynamic contact of Tregs with MN9D cells that were stained with CD45 and galectin-1, respectively. Tregs prevented MPTP-induced dopaminergic neuronal loss, behavioral changes, and attenuated the inflammatory reaction in the brain. When blockade the LFA-1 activity in Tregs or the ICAM-1 activity in endothelial cells, the percentage of Tregs in substantia nigra (SN) decreased. CD45 and galectin-1 were expressed by Tregs and MN9D cells, respectively. CD45-labeled Tregs dynamically contacted with galectin-1-labeled MN9D cells. Inhibiting CD45 in Tregs impaired the ability of Tregs to protect dopaminergic neurons against MPP+ toxicity. Similarly, galectin-1 knockdown in MN9D cells reduced the ability of Tregs neuroprotection. Adoptive transfer of Tregs protects dopaminergic neurons in PD mice by a cell-to-cell contact mechanism underlying CD45-galectin-1 interaction. Graphical Abstract.


Subject(s)
Adoptive Transfer/methods , Dopaminergic Neurons/immunology , Inflammation Mediators/immunology , Parkinsonian Disorders/immunology , Parkinsonian Disorders/therapy , T-Lymphocytes, Regulatory/immunology , Animals , Corpus Striatum/immunology , Corpus Striatum/pathology , Inflammation Mediators/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/pathology , T-Lymphocytes, Regulatory/transplantation
15.
Brain Res ; 1725: 146460, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31525350

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide. It is typically associated with motor symptoms originating from the degeneration of nigrostriatal dopamine (DA) neurons. Early stages of PD have been associated with an alteration in DA production in intestinal DAergic neurons along with inflammation. Interestingly, decreased serum concentrations of ethanolamine plasmalogens (PlsEtn) have been reported in PD patients. Ethanolamine plasmalogens play a role in vesicular fusion and release during neurotransmission, and store neuroprotective polyunsaturated fatty acids, such as docosahexaenoic acid (DHA) and are strong anti-oxidants, highlighting areas of potential therapeutic interest. Docosahexaenoic acid is known to play important roles in both the central nervous and peripheral systems, in addition to acting as a precursor of several molecules that regulate the resolution of inflammation. The present study investigated the neuroprotective and anti-inflammatory properties of the DHA-containing PlsEtn precursor, PPI-1011, in the intestine of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. Treatment with PPI-1011 prevented the MPTP-induced decrease in PlsEtn levels. In addition it prevented the loss of tyrosine hydroxylase (TH) expression and reduced the infiltration of macrophages in the myenteric plexus of MPTP-treated mice. The protective effects of PPI-1011 were observed regardless of whether it was administered pre- or post- MPTP treatment. These results suggest that PPI-1011 has neuroprotective and anti-inflammatory properties in the gut and indicate its potential utility as a treatment for both early and more advanced stages of PD.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Ileum/drug effects , Myenteric Plexus/drug effects , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Parkinsonian Disorders/immunology , Plasmalogens/administration & dosage , Animals , Ileum/immunology , Macrophages/drug effects , Macrophages/immunology , Male , Mice, Inbred C57BL , Myenteric Plexus/immunology , Neurons/immunology , Plasmalogens/blood , Tyrosine 3-Monooxygenase/metabolism
16.
Neurobiol Dis ; 132: 104575, 2019 12.
Article in English | MEDLINE | ID: mdl-31445159

ABSTRACT

Although Parkinson's disease (PD) is a progressive neurodegenerative disease, the disease does not progress or persist in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model, the most common animal model of PD. Recently, we have described that supplementation of regulated on activation, normal T cell expressed and secreted (RANTES), a chemokine known to drive infiltration of T cells, induces persistent nigrostriatal pathology in MPTP mouse model. However, which particular T cell subsets are recruited to the substantia nigra (SN) by RANTES is not known. Here, by adoptive transfer of different subset of T cells from tomato red transgenic mice to MPTP-intoxicated immunodeficient Rag1-/- mice, we describe that invasion of Th17 cells into the SN is stimulated by exogenous RANTES administration. On the other hand, RANTES supplementation remained unable to influence the infiltration of Th1 and Tregs into the SN of MPTP-insulted Rag1-/- mice. Accordingly, RANTES supplementation increased MPTP-induced TH cell loss in Rag1-/-mice receiving Th17, but neither Th1 nor Tregs. RANTES-mediated aggravation of nigral TH neurons also paralleled with significant DA loss in striatum and locomotor deficits in MPTP-intoxicated Rag1-/- mice receiving Th17 cells. Finally, we demonstrate that levels of IL-17 (a Th17-specific cytokine) and RANTES are higher in serum of PD patients than age-matched controls and that RANTES positively correlated with IL-17 in serum of PD patients. Together, these results highlight the importance of RANTES-Th17 pathway in progressive dopaminergic neuronal loss and associated PD pathology.


Subject(s)
Chemokine CCL5/immunology , Dopaminergic Neurons/pathology , Parkinsonian Disorders/immunology , Substantia Nigra/immunology , Th17 Cells/immunology , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Parkinson Disease/immunology , Parkinson Disease/pathology , Parkinsonian Disorders/pathology , Substantia Nigra/pathology
17.
J Neuroimmune Pharmacol ; 14(3): 478-492, 2019 09.
Article in English | MEDLINE | ID: mdl-31069623

ABSTRACT

Parkinson's disease (PD) is characterized by the degeneration of dopaminergic neurons and excessive microglial activation in the substantia nigra pars compacta (SNpc). In the present study, we aimed to demonstrate the therapeutic effectiveness of the potent sphingosine-1-phosphate receptor antagonist fingolimod (FTY720) in an animal model of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and to identify the potential mechanisms underlying these therapeutic effects. C57BL/6J mice were orally administered FTY720 before subcutaneous injection of MPTP. Open-field and rotarod tests were performed to determine the therapeutic effect of FTY720. The damage to dopaminergic neurons and the production of monoamine neurotransmitters were assessed using immunohistochemistry, high-performance liquid chromatography, and flow cytometry. Immunofluorescence (CD68- positive) and enzyme-linked immunosorbent assay were used to analyze the activation of microglia, and the levels of activated signaling molecules were measured using Western blotting. Our findings indicated that FTY720 significantly attenuated MPTP-induced behavioral deficits, reduced the loss of dopaminergic neurons, and increased dopamine release. FTY720 directly inhibited MPTP-induced microglial activation in the SNpc, suppressed the production of interleukin (IL)-6, IL-1ß, and tumor necrosis factor-α in BV-2 microglial cells treated with 1-methyl-4-phenylpyridinium (MPP+), and subsequently decreased apoptosis in SH-SY5Y neuroblastoma cells. Moreover, in MPP+-treated BV-2 cells and primary microglia, FTY720 treatment significantly attenuated the increases in the phosphorylation of PI3K/AKT/GSK-3ß, reduced ROS generation and p65 activation, and also inhibited the activation of NLRP3 inflammasome and caspase-1. In conclusion, FTY720 may reduce PD progression by inhibiting NLRP3 inflammasome activation via its effects on ROS generation and p65 activation in microglia. These findings provide novel insights into the mechanisms underlying the therapeutic effects of FTY720, suggesting its potential as a novel therapeutic strategy against PD. Graphical Abstract FTY720 may reduce ROS production by inhibiting the PI3K/AKT/GSK-3ß signaling pathway, while at the same time reducing p65 phosphorylation, thus decreasing NLRP3 inflammasome activation through these two pathways, ultimately reducing microglia activation-induced neuronal damage.


Subject(s)
Antiparkinson Agents/pharmacology , Fingolimod Hydrochloride/pharmacology , Inflammasomes/drug effects , Microglia/drug effects , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Animals , Apoptosis/drug effects , Cell Line , Corpus Striatum/chemistry , Corpus Striatum/drug effects , Cytokines/biosynthesis , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Drug Evaluation, Preclinical , Exploratory Behavior/drug effects , Inflammasomes/metabolism , MPTP Poisoning/drug therapy , MPTP Poisoning/immunology , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Mitochondria/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Parkinsonian Disorders/immunology , Pars Compacta/chemistry , Pars Compacta/drug effects , Pars Compacta/pathology , Reactive Oxygen Species , Rotarod Performance Test , Signal Transduction/drug effects
18.
Neurotoxicology ; 73: 112-119, 2019 07.
Article in English | MEDLINE | ID: mdl-30904435

ABSTRACT

This study investigated the expression and role of immunoproteasome (i-proteasome) in a cell model of Parkinson's disease (PD). The cytotoxicity of rotenone was measured by CCK-8 assay. The i-proteasome ß1i subunit PSMB9 was suppressed by a specific shRNA or transfected with an overexpression plasmid in the SH-SY5Y cells. Under the exposure to rotenone or not, the expression of constitutive proteasome ß subunits, i-proteasome ßi subunits, antigen presentation related proteins, α-syn and TH were detected by Western blot in PSMB9-silenced or -overexpressed cells, and the proteasomal activities were detected by fluorogenic peptide substrates. The location of i-proteasome ßi subunits and α-syn were detected by immunofluorescence staining. The levels of ROS, GSH and MDA were measured by commercial kits. Cell apoptosis was detected by flow cytometry. Besides impairing the constitutive proteasomes, rotenone induced the expression of ßi subunits of i-proteasome and antigen presentation related proteins such as TAP1, TAP2 and MHC-I. Silencing or overexpressing PSMB9 had no obvious effect on the levels of other subunits, but could regulate the chymotrypsin-like activity of 20S proteasome and the expression of TAP1, TAP2 and MHC-I. Three ßi subunits (PSMB9, PSMB10, PSMB8) of i-proteasome were all co-localized with α-syn. PSMB9 knockdown aggravated accumulation of α-syn, degradation of TH, release of ROS, increased level of MDA, decreased level of GSH and eventually promoted apoptosis in SH-SY5Y cells after rotenone treatment, while over-expression of PSMB9 could attenuate these toxic effects of rotenone. I-proteasome is activated in SH-SY5Y cells treated with rotenone and may play a neuroprotective role.


Subject(s)
Apoptosis/drug effects , Cysteine Endopeptidases/metabolism , Neurons/drug effects , Oxidative Stress/drug effects , Parkinsonian Disorders/prevention & control , Proteasome Endopeptidase Complex/metabolism , Rotenone/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 2/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 3/metabolism , Cell Line, Tumor , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/immunology , Gene Expression Regulation , Histocompatibility Antigens Class I/metabolism , Humans , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/immunology , Parkinsonian Disorders/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology , Signal Transduction , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/metabolism
19.
Life Sci ; 223: 158-165, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30880023

ABSTRACT

Inflammation contributes to the pathological processes in patients and animal models of PD. Rosmarinic acid (RA) has been demonstrated to protect neurons in PD models. The present study aimed to evaluate the anti-inflammatory effect of RA on PD and reveal possible pharmacological mechanisms. 1-Methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) was injected to mice to establish PD model in vivo. BV-2 cells were exposed to 1-methyl-4-phenylpyridinium (MPP+) and α-synuclein to establish PD model in vitro. Results showed that treatment with RA dose-dependently improved motor function of PD mice, increased the number of tyrosine hydroxylase-positive cells, reduced production of pro-inflammatory cytokines, and inhibited microglia activation in ventral midbrain. In cell study, RA also decreased MPP+ or α-synuclein-induced secretion of pro-inflammatory cytokines. Furthermore, RA treatment downregulated the expression levels of HMGB1, TLR4 and Myd88 and inhibited NF-κB nuclear expression both in PD animal and cell models. These findings indicated that RA could attenuate inflammatory responses through suppressing HMGB1/TLR4/NF-κB signaling pathway, which may contribute to its anti-PD activity.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cinnamates/therapeutic use , Depsides/therapeutic use , HMGB1 Protein/metabolism , NF-kappa B/metabolism , Parkinsonian Disorders/drug therapy , Toll-Like Receptor 4/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Anti-Inflammatory Agents/administration & dosage , Behavior, Animal/drug effects , Cinnamates/administration & dosage , Cytokines/metabolism , Depsides/administration & dosage , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Motor Activity/drug effects , Parkinsonian Disorders/immunology , Parkinsonian Disorders/metabolism , Signal Transduction/drug effects , Rosmarinic Acid
20.
Neurobiol Dis ; 124: 276-288, 2019 04.
Article in English | MEDLINE | ID: mdl-30381260

ABSTRACT

Aggregation of α-synuclein (α-syn) is neuropathologically and genetically linked to Parkinson's disease (PD). Since stereotypic cell-to-cell spreading of α-syn pathology is believed to contribute to disease progression, immunotherapy with antibodies directed against α-syn is considered a promising therapeutic approach for slowing disease progression. Here we report the identification, binding characteristics, and efficacy in PD mouse models of the human-derived α-syn antibody BIIB054, which is currently under investigation in a Phase 2 clinical trial for PD. BIIB054 was generated by screening human memory B-cell libraries from healthy elderly individuals. Epitope mapping studies conducted using peptide scanning, X-ray crystallography, and mutagenesis show that BIIB054 binds to α-syn residues 1-10. BIIB054 is highly selective for aggregated forms of α-syn with at least an 800-fold higher apparent affinity for fibrillar versus monomeric recombinant α-syn and a strong preference for human PD brain tissue. BIIB054 discriminates between monomers and oligomeric/fibrillar forms of α-syn based on high avidity for aggregates, driven by weak monovalent affinity and fast binding kinetics. In efficacy studies in three different mouse models with intracerebrally inoculated preformed α-syn fibrils, BIIB054 treatment attenuated the spreading of α-syn pathology, rescued motor impairments, and reduced the loss of dopamine transporter density in dopaminergic terminals in striatum. The preclinical data reported here provide a compelling rationale for clinical development of BIIB054 for the treatment and prevention of PD.


Subject(s)
Antibodies, Monoclonal/pharmacology , Parkinsonian Disorders/immunology , Parkinsonian Disorders/pathology , alpha-Synuclein/antagonists & inhibitors , Animals , Humans , Mice , Phenotype , Protein Aggregates
SELECTION OF CITATIONS
SEARCH DETAIL