Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters

Publication year range
1.
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
2.
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
3.
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
4.
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
5.
Neurol Sci ; 39(3): 455-460, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29264691

ABSTRACT

Contactin-associated protein 2 (CASPR2) antibodies are originally associated with Morvan's syndrome and peripheral nerve hyper excitability. Our objective was to study retrospectively the clinical spectrum of CASPR2 antibody-positive patients in our hospital. This is a retrospective observational study. Patients treated at the Amrita Institute of Medical Sciences from May 2013 to April 2016, who were tested positive for CASPR2 antibodies, were included. A total of 1584 samples were tested in the neuroimmunology laboratory during the study period for voltage-gated potassium channel (VGKC) complex antibodies-leucine-rich glioma-inactivated protein 1 (LGI1) and CASPR2 antibodies. Thirty-four were positive for LGI1, 13 were positive for CASPR2, and 7 were for both (total 54-3.4% positivity). Of these 54 cases, 11 were treated in our hospital. Seven were positive for LGI1, three for CASPR2, and one for both. The patient who had both CASPR2 and LGI1 antibody positive had Morvan's syndrome. One patient with CASPR2 had neuromyotonia. The other patient was admitted with status epilepticus with a syndrome of parkinsonism and ataxia. The third patient had encephalopathy and myoclonus with a syndrome of parkinsonism and ataxia. Two of them underwent siddha treatment for other ailments prior to the onset of the disease for other ailments. Our short series shows the expanding spectrum of CASPR2 autoimmunity. Syndrome of parkinsonism and ataxia is an important manifestation of CASPR2 autoimmunity where we can offer a definitive treatment.


Subject(s)
Ataxia/immunology , Autoantibodies/metabolism , Autoimmune Diseases of the Nervous System/immunology , Membrane Proteins/immunology , Nerve Tissue Proteins/immunology , Parkinsonian Disorders/immunology , Adult , Aged , Ataxia/therapy , Autoimmune Diseases of the Nervous System/therapy , Female , Humans , Intracellular Signaling Peptides and Proteins , Male , Middle Aged , Parkinsonian Disorders/therapy , Potassium Channels, Voltage-Gated/immunology , Proteins/immunology , Retrospective Studies , Syndrome
6.
J Biol Chem ; 291(29): 15267-81, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27226559

ABSTRACT

Parkinson disease (PD) is second only to Alzheimer disease as the most common human neurodegenerative disorder. Despite intense investigation, no interdictive therapy is available for PD. Recent studies indicate that both innate and adaptive immune processes are active in PD. Accordingly, we found a rapid increase in RANTES (regulated on activation normal T cell expressed and secreted) and eotaxin, chemokines that are involved in T cell trafficking, in vivo in the substantia nigra pars compacta and the serum of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mice. RANTES and eotaxin were also up-regulated in the substantia nigra pars compacta of post-mortem PD brains as compared with age-matched controls. Therefore, we investigated whether neutralization of RANTES and eotaxin could protect against nigrostriatal degeneration in MPTP-intoxicated mice. Interestingly, after peripheral administration, functional blocking antibodies against RANTES and eotaxin reduced the infiltration of CD4(+) and CD8(+) T cells into the nigra, attenuated nigral expression of proinflammatory molecules, and suppressed nigral activation of glial cells. These findings paralleled dopaminergic neuronal protection, normalized striatal neurotransmitters, and improved motor functions in MPTP-intoxicated mice. Therefore, we conclude that attenuation of the chemokine-dependent adaptive immune response may be of therapeutic benefit for PD patients.


Subject(s)
Chemokine CCL11/antagonists & inhibitors , Chemokine CCL5/antagonists & inhibitors , Dopaminergic Neurons/immunology , Parkinsonian Disorders/therapy , Adaptive Immunity , Aged , Aged, 80 and over , Animals , Antibodies, Blocking/administration & dosage , Brain/immunology , Brain/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Chemokine CCL11/biosynthesis , Chemokine CCL11/immunology , Chemokine CCL5/biosynthesis , Chemokine CCL5/immunology , Disease Models, Animal , Dopaminergic Neurons/pathology , Humans , Immunosuppression Therapy , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/immunology , Microglia/pathology , Middle Aged , Parkinsonian Disorders/immunology , Parkinsonian Disorders/pathology , Substantia Nigra/immunology , Substantia Nigra/pathology
7.
Neurobiol Dis ; 104: 85-96, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28476636

ABSTRACT

Disorders with progressive accumulation of α-synuclein (α-syn) are a common cause of dementia and parkinsonism in the aging population. Accumulation and propagation of α-syn play a role in the pathogenesis of these disorders. Previous studies have shown that immunization with antibodies that recognize C-terminus of α-syn reduces the intra-neuronal accumulation of α-syn and related deficits in transgenic models of synucleinopathy. These studies employed antibodies that recognize epitopes within monomeric and aggregated α-syn that were generated through active immunization or administered via passive immunization. However, it is possible that more specific effects might be achieved with antibodies recognizing selective species of the α-syn aggregates. In this respect we recently developed antibodies that differentially recognized various oligomers (Syn-O1, -O2, and -O4) and fibrilar (Syn-F1 and -F2) forms of α-syn. For this purpose wild-type α-syn transgenic (line 61) mice were immunized with these 5 different antibodies and neuropathologically and biochemically analyzed to determine which was most effective at reducing α-syn accumulation and related deficits. We found that Syn-O1, -O4 and -F1 antibodies were most effective at reducing accumulation of α-syn oligomers in multiple brain regions and at preventing neurodegeneration. Together this study supports the notion that selective antibodies against α-syn might be suitable for development new treatments for synucleinopathies such as PD and DLB.


Subject(s)
Dementia/therapy , Immunotherapy/methods , Parkinsonian Disorders/therapy , alpha-Synuclein/immunology , alpha-Synuclein/metabolism , Analysis of Variance , Animals , Antibodies/therapeutic use , Calcium-Binding Proteins/metabolism , Cell Cycle/genetics , Cell Line, Tumor , Dementia/genetics , Dementia/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Exploratory Behavior/physiology , Female , Glial Fibrillary Acidic Protein/metabolism , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Microscopy, Confocal , Neuroblastoma/pathology , Parkinsonian Disorders/genetics , Parkinsonian Disorders/immunology , Synaptophysin/metabolism , alpha-Synuclein/genetics
8.
J Neural Transm (Vienna) ; 124(2): 209-216, 2017 02.
Article in English | MEDLINE | ID: mdl-27766424

ABSTRACT

There is emerging evidence that glial cells are involved in the neuropathological process in Parkinson's disease (PD) in addition to degeneration of neuronal structures. Recently, we confirmed the presence of an adaptive immune response against different glial-derived antigens in PD, with a possible role of anti-MAG, anti-MBP and anti-PLP antibodies in the disease progression. The aim of the present study was to assess humoral response against myelin-associated glycoprotein (MAG) in patients with parkinsonism (both idiopathic and atypical) to check whether these antibodies could serve as biomarkers of PD, its severity and progression. Anti-MAG autoantibodies were measured by an ELISA system in 99 PD patients, 33 atypical parkinsonism patients, and 36 control subjects. In PD patients, anti-MAG IgM autoantibodies were significantly higher in comparison to healthy control subjects (p = 0.038). IgM anti-MAG autoantibodies titers were also significantly higher in the whole group of patients with parkinsonism (either idiopathic or atypical) in comparison to healthy control subjects (1.88 ± 0.84 vs 1.70 ± 1.19, p = 0.017). This difference was mainly driven by the PD group, as the atypical parkinsonism group did not differ significantly from the control group in anti-MAG antibody levels (p = 0.51). A negative correlation between anti-MAG levels and disease duration was found in PD patients. Our study provides evidence for an increased production of autoantibodies against a protein of glial origin in PD. The negative correlation between anti-MAG antibodies and disease duration may suggest possible involvement of the immune system in disease progression. Increasing evidence that glia are involved in the neurodegenerative process to a greater extent than previously thought may turn out be useful in the search for biomarkers of the neurodegenerative process in PD.


Subject(s)
Autoantibodies/blood , Myelin-Associated Glycoprotein/immunology , Parkinsonian Disorders/blood , Parkinsonian Disorders/immunology , Adult , Aged , Aged, 80 and over , Analysis of Variance , Biomarkers/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin G/blood , Male , Middle Aged , ROC Curve
9.
J Neuroinflammation ; 13(1): 74, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27055651

ABSTRACT

BACKGROUND: Passive and active immunization with α-synuclein has been shown to be neuroprotective in animal models of Parkinson's disease. We have previously shown that vaccination with α-synuclein, long before α-synuclein-induced brain pathology, prevents striatal degeneration by inducing regulatory T cell infiltration in parenchyma and antibody deposition on α-synuclein overexpressing neurons. However, the effect of peripheral α-synuclein on the immune system is unknown, as are the mechanistic changes induced in the CD4 T cell population during successful neuroprotective animal studies. We have studied the changes induced by vaccination with α-synuclein in the CD4 T cell pool and its impact on brain microglia to understand the immune mechanisms behind successful vaccination strategies in Parkinson's disease animal models. METHODS: Mice were immunized with WT or nitrated α-synuclein at a dose equivalent to the one used in our previous successful vaccination strategy and at a higher dose to determine potential dose-dependent effects. Animals were re-vaccinated 4 weeks after and sacrificed 5 days later. These studies were conducted in naive animals in the absence of human α-synuclein expression. RESULTS: The CD4 T cell response was modulated by α-synuclein in a dose-dependent manner, in particular the regulatory T cell population. Low-dose α-synuclein induced expansion of naive (Foxp3 + CCR6-CD127lo/neg) and dopamine receptor type D3+ regulatory T cells, as well as an increase in Stat5 protein levels. On the other hand, high dose promoted activation of regulatory T cells (Foxp3CCR6 + CD127lo/neg), which were dopamine receptor D2+D3-, and induced up-regulation of Stat5 and production of anti-α-synuclein antibodies. These effects were specific to the variant of α-synuclein used as the pathology-associated nitrated form induced distinct effects at both doses. The changes observed in the periphery after vaccination with low-dose α-synuclein correlated with an increase in CD154+, CD103+, and CD54+ microglia and the reduction of CD200R+ microglia. This resulted in the induction of a polarized tolerogenic microglia population that was CD200R-CD54CD103CD172a+ (82 % of total microglia). CONCLUSIONS: We have shown for the first time the mechanisms behind α-synuclein vaccination and, importantly, how we can modulate microglia's phenotype by regulating the CD4 T cell pool, thus shedding invaluable light on the design of neuroimmunoregulatory therapies for Parkinson's disease.


Subject(s)
Lymphocyte Activation , Microglia/immunology , Parkinsonian Disorders/immunology , T-Lymphocytes, Regulatory/immunology , alpha-Synuclein/immunology , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunohistochemistry , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/prevention & control , Recombinant Proteins/immunology , Vaccination , alpha-Synuclein/administration & dosage
10.
J Neurosci ; 34(28): 9441-54, 2014 Jul 09.
Article in English | MEDLINE | ID: mdl-25009275

ABSTRACT

Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are common neurodegenerative disorders of the aging population, characterized by progressive and abnormal accumulation of α-synuclein (α-syn). Recent studies have shown that C-terminus (CT) truncation and propagation of α-syn play a role in the pathogenesis of PD/DLB. Therefore, we explored the effect of passive immunization against the CT of α-syn in the mThy1-α-syn transgenic (tg) mouse model, which resembles the striato-nigral and motor deficits of PD. Mice were immunized with the new monoclonal antibodies 1H7, 5C1, or 5D12, all directed against the CT of α-syn. CT α-syn antibodies attenuated synaptic and axonal pathology, reduced the accumulation of CT-truncated α-syn (CT-α-syn) in axons, rescued the loss of tyrosine hydroxylase fibers in striatum, and improved motor and memory deficits. Among them, 1H7 and 5C1 were most effective at decreasing levels of CT-α-syn and higher-molecular-weight aggregates. Furthermore, in vitro studies showed that preincubation of recombinant α-syn with 1H7 and 5C1 prevented CT cleavage of α-syn. In a cell-based system, CT antibodies reduced cell-to-cell propagation of full-length α-syn, but not of the CT-α-syn that lacked the 118-126 aa recognition site needed for antibody binding. Furthermore, the results obtained after lentiviral expression of α-syn suggest that antibodies might be blocking the extracellular truncation of α-syn by calpain-1. Together, these results demonstrate that antibodies against the CT of α-syn reduce levels of CT-truncated fragments of the protein and its propagation, thus ameliorating PD-like pathology and improving behavioral and motor functions in a mouse model of this disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Brain/immunology , Movement Disorders/immunology , Movement Disorders/therapy , Parkinsonian Disorders/immunology , Parkinsonian Disorders/therapy , alpha-Synuclein/immunology , Animals , Brain/drug effects , Disease Models, Animal , Humans , Immunotherapy/methods , Mice , Mice, Transgenic , Tissue Distribution , Treatment Outcome
11.
Neurobiol Dis ; 82: 99-113, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26051538

ABSTRACT

Lewy pathology affects the gastrointestinal tract in Parkinson's disease (PD) and recent reports suggest a link between the disorder and gut inflammation. In this study, we investigated enteric neuroprotection and macrophage immunomodulation by 17ß-estradiol (E2) and the G protein-coupled estrogen receptor 1 (GPER1) in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse PD model. We found that both E2 and the GPER1 agonist G1 are protective against the loss of dopamine myenteric neurons and inhibited enteric macrophage infiltration in MPTP-treated mice. Coadministration of GPER1 antagonist G15, while completely blocking the neuroprotective and anti-inflammatory effects of G1 also partially prevented those of E2. Interestingly, we found that E2 and G1 treatments could directly alter MPTP-mediated immune responses independently from neurodegenerative processes. Analyses of monocyte/macrophage NF-κB and iNOS activation and FACs immunophenotype indicated that 1-methyl-4-phenylpyridinium (MPP(+)) treatment induces a strong immune response in monocytes, comparable to that of canonical challenge by lipopolysaccharide. In these cells, G1 and E2 treatment are equally potent in promoting a shift toward an anti-inflammatory "M2" immunophenotype reducing MPP(+)-induced NF-κB and iNOS activation. Moreover, G15 also antagonized the immunomodulatory effects of G1 in MPP(+)-treated macrophages. Together these data provide the first evidence for the role of GPER1 in enteric immunomodulation and neuroprotection. Considering increasing recognition for myenteric pathology as an early biomarker for PD, these findings provide a valuable contribution for better understanding and targeting of future therapeutic strategies.


Subject(s)
Immunomodulation/genetics , Myenteric Plexus/metabolism , Neuroprotection/genetics , Parkinsonian Disorders/metabolism , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Animals , Benzodioxoles/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/immunology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Estradiol/pharmacology , Estradiol/therapeutic use , Immunomodulation/drug effects , Mice , Myenteric Plexus/drug effects , Myenteric Plexus/immunology , Myenteric Plexus/pathology , NF-kappa B/metabolism , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nitric Oxide Synthase Type II/metabolism , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/immunology , Parkinsonian Disorders/pathology , Quinolines/pharmacology , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism
12.
Immunopharmacol Immunotoxicol ; 37(4): 393-9, 2015.
Article in English | MEDLINE | ID: mdl-26211726

ABSTRACT

CONTEXT: Neuroimmunological response is associated with neurodegeneration in the human substantia nigra (SN) in Parkinson's disease (PD). OBJECTIVE: To explore the possibility that the neurotoxin, 6-hydroxydopamine (6-OHDA), could be used as a tool in mice to understand the immune response in PD. MATERIALS AND METHODS: We employed unilateral administration of 6-OHDA into the mouse SN. At 1 week, 2 weeks and 4 weeks post-injection, we used immunohistochemistry for the markers Iba-1 and gp91PHOX to investigate activated microglia in the SN. To examine the adaptive immune response, we used immunohistochemistry for CD3-positive T-lymphocytes, CD45R-positive B-lymphocytes and anti-mouse immunoglobulin-G (IgG). Dopamine neuron loss was examined using immunohistochemistry for the dopamine neuron marker, tyrosine hydroxylase. RESULTS: Compared to vehicle, 6-OHDA administration induced an intense IgG deposition in the SN as well as increased infiltration of both T- and B- lymphocytes into the injected side of the midbrain. The adaptive immune response was associated with extensive destruction of dopamine neurons and extensive microglial activation at every time point in the 6-OHDA groups. CONCLUSION: Our results suggest that 6-OHDA administration in mice can a potential tool for understanding mechanisms underlying adaptive immune activation-induced neurodegeneration in PD.


Subject(s)
Adaptive Immunity/drug effects , Nerve Degeneration/drug therapy , Oxidopamine/pharmacology , Parkinsonian Disorders/immunology , Substantia Nigra/drug effects , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CD3 Complex/immunology , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/immunology , Dopaminergic Neurons/pathology , Immunoglobulin G/immunology , Immunohistochemistry , Leukocyte Common Antigens/immunology , Male , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Parkinsonian Disorders/pathology , Substantia Nigra/immunology , Substantia Nigra/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
13.
Neurobiol Dis ; 69: 134-43, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24851801

ABSTRACT

Several lines of evidence suggest that accumulation of aggregated alpha-synuclein (α-synuclein) in the central nervous system (CNS) is an early pathogenic event in Parkinson's disease and other Lewy body disorders. In recent years, animal studies have indicated immunotherapy with antibodies directed against α-synuclein as a promising novel treatment strategy. Since large α-synuclein oligomers, or protofibrils, have been demonstrated to possess pronounced cytotoxic properties, such species should be particularly attractive as therapeutic targets. In support of this, (Thy-1)-h[A30P] α-synuclein transgenic mice with motor dysfunction symptoms were found to display increased levels of α-synuclein protofibrils in the CNS. An α-synuclein protofibril-selective monoclonal antibody (mAb47) was evaluated in this α-synuclein transgenic mouse model. As measured by ELISA, 14month old mice treated for 14weeks with weekly intraperitoneal injections of mAb47 displayed significantly lower levels of both soluble and membrane-associated protofibrils in the spinal cord. Besides the lower levels of pathogenic α-synuclein demonstrated, a reduction of motor dysfunction in transgenic mice upon peripheral administration of mAb47 was indicated. Thus, immunotherapy with antibodies targeting toxic α-synuclein species holds promise as a future disease-modifying treatment in Parkinson's disease and related disorders.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunization, Passive , Parkinsonian Disorders/pathology , Parkinsonian Disorders/therapy , alpha-Synuclein/immunology , Animals , Antibodies, Monoclonal/metabolism , Brain/immunology , Brain/pathology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Injections, Intraperitoneal , Male , Mice, Transgenic , Motor Activity/physiology , Mutation , Parkinsonian Disorders/immunology , Severity of Illness Index , Spinal Cord/immunology , Spinal Cord/pathology , alpha-Synuclein/genetics
14.
J Neurosci Res ; 91(10): 1292-302, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23907992

ABSTRACT

We previously showed that, in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease (PD), vaccination with bacillus Calmette-Guerin (BCG) prior to MPTP exposure limited the loss of striatal dopamine (DA) and dopamine transporter (DAT) and prevented the activation of nigral microglia. Here, we conducted BCG dose studies and investigated the mechanisms underlying BCG vaccination's neuroprotective effects in this model. We found that a dose of 1 × 10(6) cfu BCG led to higher levels of striatal DA and DAT ligand binding (28% and 42%, respectively) in BCG-vaccinated vs. unvaccinated MPTP-treated mice, but without a significant increase in substantia nigra tyrosine hydroxylase-staining neurons. Previous studies showed that BCG can induce regulatory T cells (Tregs) and that Tregs are neuroprotective in models of neurodegenerative diseases. However, MPTP is lymphotoxic, so it was unclear whether Tregs were maintained after MPTP treatment and whether a relationship existed between Tregs and the preservation of striatal DA system integrity. We found that, 21 days post-MPTP treatment, Treg levels in mice that had received BCG prior to MPTP were threefold greater than those in MPTP-only-treated mice and elevated above those in saline-only-treated mice, suggesting that the persistent BCG infection continually promoted Treg responses. Notably, the magnitude of the Treg response correlated positively with both striatal DA levels and DAT ligand binding. Therefore, BCG vaccine-mediated neuroprotection is associated with Treg levels in this mouse model. Our results suggest that BCG-induced Tregs could provide a new adjunctive therapeutic approach to ameliorating pathology associated with PD and other neurodegenerative diseases.


Subject(s)
BCG Vaccine/immunology , Neuroprotective Agents/immunology , Parkinsonian Disorders/immunology , Parkinsonian Disorders/prevention & control , T-Lymphocytes, Regulatory/immunology , Animals , BCG Vaccine/administration & dosage , Brain/immunology , Brain/pathology , Chromatography, High Pressure Liquid , Disease Models, Animal , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/administration & dosage
15.
J Immunol ; 187(12): 6508-17, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22079984

ABSTRACT

This study examined whether the cannabinoid receptor type 1 (CB(1)) receptor contributes to the survival of nigrostriatal dopaminergic (DA) neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. MPTP induced significant loss of nigrostriatal DA neurons and microglial activation in the substantia nigra (SN), visualized with tyrosine hydroxylase or macrophage Ag complex-1 immunohistochemistry. Real-time PCR, ELISA, Western blotting, and immunohistochemistry disclosed upregulation of proinflammatory cytokines, activation of microglial NADPH oxidase, and subsequent reactive oxygen species production and oxidative damage of DNA and proteins in MPTP-treated SN, resulting in degeneration of DA neurons. Conversely, treatment with nonselective cannabinoid receptor agonists (WIN55,212-2 and HU210) led to increased survival of DA neurons in the SN, their fibers and dopamine levels in the striatum, and improved motor function. This neuroprotection by cannabinoids was accompanied by suppression of NADPH oxidase reactive oxygen species production and reduced expression of proinflammatory cytokines from activated microglia. Interestingly, cannabinoids protected DA neurons against 1-methyl-4-phenyl-pyridinium neurotoxicity in cocultures of mesencephalic neurons and microglia, but not in neuron-enriched mesencephalic cultures devoid of microglia. The observed neuroprotection and inhibition of microglial activation were reversed upon treatment with CB(1) receptor selective antagonists AM251 and/or SR14,716A, confirming the involvement of the CB(1) receptor. The present in vivo and in vitro findings clearly indicate that the CB(1) receptor possesses anti-inflammatory properties and inhibits microglia-mediated oxidative stress. Our results collectively suggest that the cannabinoid system is beneficial for the treatment of Parkinson's disease and other disorders associated with neuroinflammation and microglia-derived oxidative damage.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Corpus Striatum/immunology , Growth Inhibitors/physiology , Microglia/drug effects , Microglia/immunology , Neurotoxins/adverse effects , Receptor, Cannabinoid, CB1/physiology , Substantia Nigra/immunology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Animals , Benzoxazines/pharmacology , Cells, Cultured , Coculture Techniques , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine Agents/administration & dosage , Dopamine Agents/adverse effects , Dronabinol/analogs & derivatives , Dronabinol/pharmacology , Male , Mice , Mice, Inbred C57BL , Microglia/pathology , Morpholines/pharmacology , Naphthalenes/pharmacology , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Neurotoxins/administration & dosage , Parkinsonian Disorders/immunology , Parkinsonian Disorders/pathology , Parkinsonian Disorders/prevention & control , Receptor, Cannabinoid, CB1/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism
16.
Neurol Sci ; 34(9): 1559-70, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23354599

ABSTRACT

It has been suggested that DNA vaccine plays a protective effect on degenerative diseases in the central nervous system (CNS), the Parkinson's disease (PD) included. In this study, we assessed the immune effects of optimized DNA vaccine (pVAX1-IL-4/SYN-B) in the C57BL/6 mice by ELISA, and immunohistochemistry. We also evaluated the neuroprotective effect of pVAX1-IL-4/SYN-B in MPTP model of Parkinson's disease, using behavioral methods, immunohistochemistry and western blot. We found that alphα-synuclein (α-syn) antibody significantly increased, IL-4 increased and IFN-r reduced in the serum of immunized C57BL/6 mice in optimized DNA vaccine group. The immune serum of mice specifically combined with the α-syn positive inclusion bodies in the brain of PD model mice. The preventive immunization with optimized DNA vaccine made the motor symptoms improved significantly, the apoptosis of tyrosine hydroxylase (TH) neuron and cyclooxygenase-2 (COX-2) expression significantly decreased in MPTP model mice. These results suggest that optimized DNA vaccine can make immunized mice produce high titers of specific α-syn antibody, mainly causing the humoral immune response; preventive immunization with optimized DNA vaccine can play neuroprotective and anti-inflammatory effects on mice suffering from the sub-acute MPTP Parkinson's disease.


Subject(s)
Autoantibodies/immunology , Parkinsonian Disorders/immunology , Vaccines, DNA/pharmacology , alpha-Synuclein/immunology , Animals , Autoantibodies/blood , Autoantibodies/pharmacology , Blotting, Western , Cytokines/blood , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/blood , Parkinsonian Disorders/prevention & control , Vaccines, DNA/immunology , alpha-Synuclein/antagonists & inhibitors
17.
J Clin Invest ; 119(1): 13-5, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19104142

ABSTRACT

Neuroinflammation, characterized by activated microglia and infiltrating T cells, is a prominent pathological feature in neurodegenerative diseases. However, whether this inflammation contributes to neuronal injury or is a late consequence of neuronal injury is unclear. In this issue of the JCI, Brochard et al. report that CD4+ T cells are cytotoxic in a mouse model of Parkinson disease (PD) (see the related article beginning on page 182). Specifically, invading T lymphocytes contributed to neuronal cell death via the Fas/FasL pathway. The results implicate the adaptive immune system in the pathogenesis of Parkinson neurodegeneration and provide a meaningful rationale for immune-based therapies for PD.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Parkinsonian Disorders/immunology , T-Lymphocyte Subsets/immunology , Animals , Apoptosis/physiology , CD4-Positive T-Lymphocytes/cytology , Disease Models, Animal , Dopamine/metabolism , Humans , Inflammation/immunology , Mice , Microglia/immunology , Parkinsonian Disorders/pathology , T-Lymphocyte Subsets/cytology , alpha-Synuclein/metabolism
18.
J Clin Invest ; 119(1): 182-92, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19104149

ABSTRACT

Parkinson disease (PD) is a neurodegenerative disorder characterized by a loss of dopamine-containing neurons. Mounting evidence suggests that dopaminergic cell death is influenced by the innate immune system. However, the pathogenic role of the adaptive immune system in PD remains enigmatic. Here we showed that CD8+ and CD4+ T cells but not B cells had invaded the brain in both postmortem human PD specimens and in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD during the course of neuronal degeneration. We further demonstrated that MPTP-induced dopaminergic cell death was markedly attenuated in the absence of mature T lymphocytes in 2 different immunodeficient mouse strains (Rag1-/- and Tcrb-/- mice). Importantly, similar attenuation of MPTP-induced dopaminergic cell death was seen in mice lacking CD4 as well as in Rag1-/- mice reconstituted with FasL-deficient splenocytes. However, mice lacking CD8 and Rag1-/- mice reconstituted with IFN-gamma-deficient splenocytes were not protected. These data indicate that T cell-mediated dopaminergic toxicity is almost exclusively arbitrated by CD4+ T cells and requires the expression of FasL but not IFNgamma. Further, our data may provide a rationale for targeting the adaptive arm of the immune system as a therapeutic strategy in PD.


Subject(s)
Brain , CD4-Positive T-Lymphocytes/immunology , Nerve Degeneration , Parkinson Disease , Parkinsonian Disorders , Aged , Aged, 80 and over , Animals , Brain/cytology , Brain/immunology , Brain/pathology , CD4-Positive T-Lymphocytes/cytology , Cell Death/physiology , Disease Models, Animal , Dopamine/metabolism , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immune System/physiology , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Parkinson Disease/immunology , Parkinson Disease/pathology , Parkinsonian Disorders/immunology , Parkinsonian Disorders/pathology
19.
Bull Acad Natl Med ; 196(7): 1369-77; discussion 1377-9, 2012 Oct.
Article in French | MEDLINE | ID: mdl-23815021

ABSTRACT

Parkinson's disease is characterized by a triad of cardinal motor symptoms (bradykinesia, rigidity and tremor) resulting from the loss of dopaminergic neurons in the substantia nigra. This synucleinopathy is classified in the larger group of Lewy body disorders. Currently, these symptoms are relatively well alleviated by drugs that restore dopaminergic neurotransmission, andlor by deep brain stimulation. It is not yet possible to halt the underlying degeneration, or to treat symptoms due to non-dopaminergic neuron damage. This review examines the mechanisms of neuronal degeneration in Parkinson's disease, new targets for neuroprotection, and the mechanisms causing symptoms resistant to current treatments.


Subject(s)
Antiparkinson Agents/therapeutic use , Molecular Targeted Therapy , Parkinson Disease/drug therapy , Adaptive Immunity , Animals , Antiparkinson Agents/pharmacology , Brain/pathology , Cation Transport Proteins/physiology , Cell Death , Cholinergic Neurons/pathology , Deep Brain Stimulation , Dopamine Agonists/therapeutic use , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Forecasting , Haplorhini , Humans , Magnetic Resonance Imaging , Mice , Nerve Degeneration/physiopathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress , Parkinson Disease/therapy , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/immunology , Parkinsonian Disorders/pathology
20.
J Pharmacol Exp Ther ; 339(2): 618-23, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21831964

ABSTRACT

Biologic tumor necrosis factor (TNF)-α inhibitors do not cross the blood-brain barrier (BBB). A BBB-penetrating TNF-α inhibitor was engineered by fusion of the extracellular domain of the type II human TNF receptor (TNFR) to the carboxyl terminus of the heavy chain of a mouse/rat chimeric monoclonal antibody (MAb) against the mouse transferrin receptor (TfR), and this fusion protein is designated cTfRMAb-TNFR. The cTfRMAb-TNFR fusion protein and etanercept bound human TNF-α with high affinity and K(D) values of 374 ± 77 and 280 ± 80 pM, respectively. Neuroprotection in brain in vivo after intravenous administration of the fusion protein was examined in a mouse model of Parkinson's disease. Mice were also treated with saline or a non-BBB-penetrating TNF decoy receptor, etanercept. After intracerebral injection of the nigral-striatal toxin, 6-hydroxydopamine, mice were treated every other day for 3 weeks. Treatment with the cTfRMAb-TNFR fusion protein caused an 83% decrease in apomorphine-induced rotation, a 67% decrease in amphetamine-induced rotation, a 82% increase in vibrissae-elicited forelimb placing, and a 130% increase in striatal tyrosine hydroxylase (TH) enzyme activity. In contrast, chronic treatment with etanercept, which does not cross the BBB, had no effect on neurobehavior or striatal TH enzyme activity. A bridging enzyme-linked immunosorbent assay specific for the cTfRMAb-TNFR fusion protein showed that the immune response generated in the mice was low titer. In conclusion, a biologic TNF inhibitor is neuroprotective after intravenous administration in a mouse model of neurodegeneration, providing that the TNF decoy receptor is reengineered to cross the BBB.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Blood-Brain Barrier/metabolism , Neuroprotective Agents/therapeutic use , Parkinsonian Disorders/drug therapy , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Behavior, Animal/drug effects , Brain/immunology , Brain/metabolism , CHO Cells , Control Groups , Corpus Striatum/drug effects , Cricetinae , Etanercept , Humans , Immunoglobulin G/pharmacology , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacokinetics , Parkinsonian Disorders/immunology , Rats , Receptors, Transferrin/immunology , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Substantia Nigra/drug effects , Tumor Necrosis Factor Decoy Receptors/pharmacokinetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Tyrosine 3-Monooxygenase/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL