Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
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
3.
Mol Neurodegener ; 5: 42, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20977765

ABSTRACT

Overexpression of alpha-synuclein (α-SYN), a protein which plays an important role in the pathogenesis of Parkinson's disease (PD), triggers microglial activation and adaptive immune responses, and leads to neurodegeneration of dopaminergic (DA) neurons. We hypothesized a link between the humoral adaptive immune response and microglial activation in α-SYN induced neurodegeneration. To test this hypothesis, we employed adeno-associated virus serotype 2 (AAV2) to selectively over-express human α-SYN in the substantia nigra (SN) of wild-type mice and FcγR-/- mice, which lack high-affinity receptors for IgG. We found that in wild-type mice, α-SYN induced the expression of NF-κB p65 and pro-inflammatory molecules. In FcγR-/- mice, NF-κB activation was blocked and pro-inflammatory signaling was reduced. Microglial activation was examined using immunohistochemistry for gp91PHOX. At four weeks, microglia were strongly activated in wild-type mice, while microglial activation was attenuated in FcγR-/- mice. Dopaminergic neurodegeneration was examined using immunohistochemistry for tyrosine hydroxylase (TH) and unbiased stereology. α-SYN overexpression led to the appearance of dysmorphic neurites, and a loss of DA neurons in the SN in wild-type animals, while FcγR-/- mice did not exhibit neuritic change and were protected from α-SYN-induced neurodegeneration 24 weeks after injection. Our results suggest that the humoral adaptive immune response triggered by excess α-SYN plays a causative role in microglial activation through IgG-FcγR interaction. This involves NF-κB signaling, and leads to DA neurodegeneration. Therefore, blocking either FcγR signaling or specific intracellular signal transduction events downstream of FcγR-IgG interaction, such as NF-κB activation, may be viable therapeutic strategies in PD.

4.
J Neuropathol Exp Neurol ; 67(12): 1149-58, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19018246

ABSTRACT

Microglial activation and adaptive immunity have been implicated in the neurodegenerative processes in Parkinson disease. It has been proposed that these responses may be triggered by modified forms of alpha-synuclein (alpha-SYN), particularly nitrated species, which are released as a consequence of dopaminergic neurodegeneration. To examine the relationship between alpha-SYN, microglial activation, and adaptive immunity, we used a mouse model of Parkinson disease in which human alpha-SYN is overexpressed by a recombinant adeno-associated virus vector, serotype 2 (AAV2-SYN); this overexpression leads to slow degeneration of dopaminergic neurons. Microglial activation and components of the adaptive immune response were assessed using immunohistochemistry; quantitative polymerase chain reaction was used to examine cytokine expression. Four weeks after injection, there was a marked increase in CD68-positive microglia and greater infiltration of B and T lymphocytes in the substantia nigra pars compacta of the AAV2-SYN group than in controls. At 12 weeks, CD68 staining declined, but B- and T-cell infiltration persisted. Expression of proinflammatory cytokines was enhanced, whereas markers of alternative activation (i.e. arginase I and interleukins 4 and 13) were not altered. Increased immunoreactivity for mouse immunoglobulin was detected at all time points in the AAV2-SYN animals. These data show that overexpression of alpha-SYN alone, in the absence of overt neurodegeneration, is sufficient to trigger neuroinflammation with both microglial activation and stimulation of adaptive immunity.


Subject(s)
Brain/immunology , Immune System Phenomena/immunology , Lymphocyte Activation/immunology , Microglia/immunology , Parkinson Disease/immunology , alpha-Synuclein/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/immunology , Antigens, Differentiation, Myelomonocytic/metabolism , Brain/pathology , Brain/physiopathology , Chemotaxis, Leukocyte/immunology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Dopamine/metabolism , Genetic Vectors , Humans , Immunoglobulins/immunology , Immunoglobulins/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Substantia Nigra/immunology , Substantia Nigra/pathology , Substantia Nigra/physiopathology , Transfection , alpha-Synuclein/genetics
5.
Curr HIV Res ; 5(3): 301-13, 2007 May.
Article in English | MEDLINE | ID: mdl-17504172

ABSTRACT

HIV-1 infection with concurrent methamphetamine (MA) abuse results in exacerbated neurodegenerative changes and rapid progression of a form of sub-cortical dementia termed HIV-1 associated dementia (HAD). A notable feature of HAD is the involvement of the dopaminergic system manifested as parkinsonian like movement abnormalities. The HIV-1 transactivator of transcription (Tat) protein is very often used in experimental studies trying to understand neurotoxic consequences of HIV-1 infection, since the pathophysiological changes induced by Tat mirrors, in part, the means by which HIV-1 infection of the nervous system results in neuronal damage. Understanding the interaction of Tat and MA in the basal ganglia and the resultant injury to the dopaminergic system in rodent models as well as cell culture will shed light on the dopaminergic pathology occurring in HIV-1 infected-MA abusers. The aim of this review is to update the reader on the current knowledge of MA and HIV-1 neurotoxicity, specifically Tat, and discuss the progress in understanding how MA synergizes with the HIV-1 transactivator protein Tat to damage the basal ganglia.


Subject(s)
AIDS Dementia Complex/etiology , Basal Ganglia/physiopathology , Dopamine Agents/pharmacology , HIV Infections/complications , HIV-1 , Methamphetamine/pharmacology , AIDS Dementia Complex/physiopathology , Amphetamine-Related Disorders/complications , Basal Ganglia/drug effects , Basal Ganglia/virology , Central Nervous System Stimulants/pharmacology , Gene Products, tat/physiology , Humans , Neurodegenerative Diseases/etiology , tat Gene Products, Human Immunodeficiency Virus
6.
Ann N Y Acad Sci ; 1074: 178-90, 2006 Aug.
Article in English | MEDLINE | ID: mdl-17105916

ABSTRACT

The human immunodeficiency virus-1 (HIV-1) affects the central nervous system (CNS) in approximately 30% of infected individuals and basal ganglia structures seem to be most affected. The HIV-1-transactivating protein, Tat, has been suggested to be pathogenically relevant in HIV-1-induced neuronal injury. The abuse of methamphetamine (METH), which is great among this patient population, also affects the basal ganglia, causing degeneration of dopaminergic terminals. In previous studies, we demonstrated that coexposure to these two toxins caused a synergistic loss of striatal dopamine and binding to the dopamine transporter (DAT), suggesting a loss of dopamine terminals. Because the loss of dopamine and DAT, however, do not necessarily reflect dopamine terminal degeneration, we have used silver staining and TH immunohistochemistry to further examine this issue. We have also examined the glial reaction using GFAP as a marker of astrocyte activation and OX-42 as a marker of activated microglia. Lastly, we have begun to explore the mechanism of synergy by investigating the role that the cytokine TNF-alpha might play in Tat + METH synergy. Our data indicate that the synergistic loss of dopamine is likely the result of dopamine terminal degeneration. This injury is not a direct result of the number of activated glia but does involve TNF-alpha.


Subject(s)
Corpus Striatum/drug effects , Dopamine/metabolism , Gene Products, tat/pharmacology , Methamphetamine/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Corpus Striatum/metabolism , Drug Synergism , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/drug effects , Rats , Silver Staining , Tumor Necrosis Factor-alpha/genetics
7.
Neurobiol Dis ; 23(3): 663-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16828290

ABSTRACT

Our previous studies demonstrated that the psychostimulant methamphetamine (MA) and the human immunodeficiency virus-1 (HIV-1) protein Tat interacted to cause enhanced dopaminergic neurotoxicity. The present study examined whether tumor necrosis factor-alpha (TNF-alpha) mediates the interaction between Tat and MA. In Sprague-Dawley rats, injections of Tat caused a small but significant increase in striatal TNF-alpha level, whereas MA resulted in no change. The increase in TNF-alpha induced by Tat + MA was not significantly different from that induced by Tat alone. Temporal analysis of TNF-alpha levels revealed a 50-fold increase 4 h after Tat administration. In C57BL/6 mice, Tat + MA induced a 50% decline in striatal dopamine levels, which was significantly attenuated in mice lacking both receptors for TNF-alpha. TNF-alpha synthesis inhibitors significantly attenuated Tat + MA neurotoxicity in hippocampal neuronal culture. The results suggest that Tat-induced elevation of TNF-alpha may predispose the dopaminergic terminals to subsequent damage by MA.


Subject(s)
Encephalitis/immunology , Gene Products, tat/toxicity , Methamphetamine/toxicity , Oxidative Stress/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , AIDS Dementia Complex/immunology , AIDS Dementia Complex/physiopathology , AIDS Dementia Complex/virology , Amphetamine-Related Disorders/immunology , Amphetamine-Related Disorders/metabolism , Amphetamine-Related Disorders/physiopathology , Animals , Basal Ganglia/drug effects , Basal Ganglia/immunology , Basal Ganglia/virology , Cells, Cultured , Disease Models, Animal , Dopamine/metabolism , Dopamine Uptake Inhibitors/toxicity , Encephalitis/physiopathology , Encephalitis/virology , Gene Products, tat/immunology , Gene Products, tat/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/drug effects , Presynaptic Terminals/drug effects , Presynaptic Terminals/immunology , Presynaptic Terminals/virology , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects , Up-Regulation/immunology , tat Gene Products, Human Immunodeficiency Virus
8.
Exp Neurol ; 199(2): 490-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16510141

ABSTRACT

Human immunodeficiency virus-1 (HIV-1) infection of the brain causes elevation in pro-inflammatory cytokines and inflammatory changes in the striatum. HIV-1-infected individuals who also abuse drugs including the psychostimulant methamphetamine (MA) develop more severe encephalitis and neuronal damage compared to HIV-1-infected patients who do not abuse drugs. In previous studies, we demonstrated that the HIV-1 protein Tat and MA interacted to cause enhanced loss of dopamine in the rat striatum via the destruction of dopaminergic terminals. Since both Tat and MA activate glia and induce cytokine production, we investigated the role of cytokines in the synergistic neurotoxicity induced by Tat and MA using cytokine arrays. Significant increases in monocyte chemotactic protein (MCP-1), interleukin-1 alpha (IL-1alpha) and tissue inhibitor of metalloproteinase-1 (TIMP-1) levels were noted 4 h following Tat + MA treatment compared to saline, Tat or MA. MCP-1 and TIMP-1 levels remained elevated 16 h after Tat + MA compared to saline or MA but were not different from the Tat-treated group at this time point. Weak, but significant elevations in cytokine-induced neutrophil chemoattractant-3 (CINC-3), ciliary neurotrophic factor (CNTF) and macrophage inflammatory protein-3 alpha (MIP-3alpha) were also noted with Tat + MA. The interaction of Tat and MA was prevented in mice genetically deficient in MCP-1 with a consequent attenuation of Tat + MA neurotoxicity. Our findings suggest that HIV-1 infection with concurrent drug abuse might profoundly increase chemokine levels in the striatum resulting in enhanced damage to the dopaminergic system.


Subject(s)
Corpus Striatum , Cytokines/metabolism , Dopamine Uptake Inhibitors/administration & dosage , Gene Products, tat/metabolism , HIV-1/chemistry , Methamphetamine/administration & dosage , Analysis of Variance , Animals , Blotting, Western/methods , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/virology , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , HIV Infections/immunology , HIV Infections/virology , HIV-1/pathogenicity , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Time Factors , tat Gene Products, Human Immunodeficiency Virus
9.
Biochemistry ; 41(46): 13483-8, 2002 Nov 19.
Article in English | MEDLINE | ID: mdl-12427008

ABSTRACT

The loss of function of WRN, a DNA helicase and exonuclease, causes the premature aging disease Werner syndrome. A hallmark feature of cells lacking WRN is genomic instability typified by elevated illegitimate recombination events and accelerated loss of telomeric sequences. In this study, the activities of WRN were examined on a displacement loop (D-loop) DNA substrate that mimics an intermediate formed during the strand invasion step of many recombinational processes. Our results indicate that this model substrate is specifically bound by WRN and efficiently disrupted by its helicase activity. In addition, the 3' end of the inserted strand of this D-loop structure is readily attacked by the 3'-->5' exonuclease function of WRN. These results indicate that D-loop structures are favored sites for WRN action. Thus, WRN may participate in DNA metabolic processes that utilize these structures, such as recombination and telomere maintenance pathways.


Subject(s)
DNA Helicases/genetics , DNA Helicases/metabolism , DNA/metabolism , Exonucleases/metabolism , Werner Syndrome/enzymology , Base Sequence , DNA/chemistry , DNA Footprinting , DNA Primers , Deoxyribonuclease I/metabolism , Dimerization , Electrophoretic Mobility Shift Assay , Exodeoxyribonucleases , Humans , In Vitro Techniques , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , RecQ Helicases , Recombinant Proteins/metabolism , Substrate Specificity , Werner Syndrome Helicase
10.
J Biol Chem ; 277(6): 4492-504, 2002 Feb 08.
Article in English | MEDLINE | ID: mdl-11717307

ABSTRACT

Werner syndrome is a premature aging and cancer-prone hereditary disorder caused by deficiency of the WRN protein that harbors 3' -->5' exonuclease and RecQ-type 3' --> 5' helicase activities. To assess the possibility that WRN acts on partially melted DNA intermediates, we constructed a substrate containing a 21-nucleotide noncomplementary region asymmetrically positioned within a duplex DNA fragment. Purified WRN shows an extremely efficient exonuclease activity directed at both blunt ends of this substrate, whereas no activity is observed on a fully duplex substrate. High affinity binding of full-length WRN protects an area surrounding the melted region of the substrate from DNase I digestion. ATP binding stimulates but is not required for WRN binding to this region. Thus, binding of WRN to the melted region underlies the efficient exonuclease activity directed at the nearby ends. In contrast, a WRN deletion mutant containing only the functional exonuclease domain does not detectably bind or degrade this substrate. These experiments indicate a bipartite structure and function for WRN, and we propose a model by which its DNA binding, helicase, and exonuclease activities function coordinately in DNA metabolism. These studies also suggest that partially unwound or noncomplementary regions of DNA could be physiological targets for WRN.


Subject(s)
DNA Footprinting , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Deoxyribonuclease I/metabolism , Exonucleases/metabolism , Werner Syndrome/metabolism , Base Sequence , Exodeoxyribonucleases , Humans , Molecular Sequence Data , RecQ Helicases , Recombinant Proteins/metabolism , Werner Syndrome Helicase
SELECTION OF CITATIONS
SEARCH DETAIL
...