Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Neurology ; 66(6): 919-21, 2006 Mar 28.
Article in English | MEDLINE | ID: mdl-16510768

ABSTRACT

In vitro and animal model data demonstrate that valproic acid (VPA) can ameliorate HIV-associated neurotoxicity. The authors conducted a pilot 10-week placebo-controlled study of VPA 250 mg twice daily in 22 HIV-infected individuals with (n = 16) and without (n = 6) cognitive impairment. VPA was safe and well tolerated, with trends toward improved neuropsychological performance and brain metabolism in the impaired subjects.


Subject(s)
AIDS Dementia Complex/drug therapy , HIV Infections/drug therapy , HIV-1 , Valproic Acid/therapeutic use , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/psychology , Adult , Anti-Retroviral Agents/therapeutic use , CD4 Lymphocyte Count , Double-Blind Method , Drug Therapy, Combination , Female , HIV Infections/metabolism , HIV Infections/psychology , Humans , Male , Middle Aged , Pilot Projects
2.
Neurotox Res ; 8(1-2): 161-6, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16260393

ABSTRACT

In the past decade we have seen a milder phenotype and decreased incidence of HIV-1 associated dementia (HAD), largely due to the widespread use of combination chemotherapy to reduce viral burden. However, the prevalence of neurologic disease in people living with HIV-1 has actually increased, raising significant concerns that new therapeutic strategies, directed at restoring neuronal and glial homeostasis and signaling in the central nervous system (CNS), as opposed to directly interfering with the life cycle of HIV-1, must be developed. In this review, we focus briefly on previous Phase 1 clinical trials for adjunctive (i.e., chemotherapeutic agents that do not have a primary antiretroviral mechanism of action) therapy in patients with HAD, followed by an overview of key molecular events in the neuropathogenesis of HAD, and then discuss in more detail our rationale for investigating the effects of therapeutic agents that restore impaired mitochondrial bioenergetics in the CNS. Specifically, we focus on agents that either work in part through K-ATP channels, present in both mitochondria and plasma membranes, and agents that work to weakly uncouple the respiratory capacity of the electron transport chain in mitochondria from ATP production. We propose these agents may be complementary to currently available antiretroviral agents and may significantly improve the capacity of CNS infected with HIV-1 to meet increased bioenergetic demands involved in normal synaptic communication.


Subject(s)
AIDS Dementia Complex/drug therapy , HIV-1 , Neuroprotective Agents/therapeutic use , AIDS Dementia Complex/metabolism , Animals , Antiretroviral Therapy, Highly Active , Energy Metabolism/drug effects , Humans
3.
J Neurosci Res ; 80(4): 562-75, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15825192

ABSTRACT

Relatively few immune-activated and virus-infected mononuclear phagocytes (MP; perivascular macrophages and microglia) may affect widespread neuronal dysfunction during human immunodeficiency virus type 1 (HIV-1)-associated dementia (HAD). Indeed, histopathological evidence of neuronal dropout often belies the extent of cognitive impairment. To define relationships between neuronal function and histopathology, proton magnetic resonance spectroscopic imaging (1H MRSI) and hippocampal long-term potentiation (LTP) were compared with neuronal and glial immunohistology in a murine model of HIV-1 encephalitis (HIVE). HIV-1(ADA)-infected human monocyte-derived macrophages (MDM) were stereotactically injected into the subcortex of severe combined immunodeficient (SCID) mice. Sham-operated and unmanipulated mice served as controls. Seven days after cell injection, brain histological analyses revealed a focal giant cell encephalitis, with reactive astrocytes, microgliosis, and neuronal dropout. Strikingly, significant reductions in N-acetyl aspartate concentration ([NAA]) and LTP levels in HIVE mice were in both injected and contralateral hemispheres and in brain subregions, including the hippocampus, where neuropathology was limited or absent. The data support the importance of 1H MRSI as a tool for assessing neuronal function for HAD. The data also demonstrate that a highly focal encephalitis can produce global deficits for neuronal function and metabolism.


Subject(s)
AIDS Dementia Complex/pathology , Aspartic Acid/analogs & derivatives , Cognition Disorders/pathology , HIV-1 , Magnetic Resonance Spectroscopy , AIDS Dementia Complex/complications , AIDS Dementia Complex/physiopathology , Animals , Aspartic Acid/metabolism , Brain Mapping , Calcium-Binding Proteins/metabolism , Capsid Proteins/metabolism , Choline/metabolism , Cognition Disorders/etiology , Cognition Disorders/virology , Creatine/metabolism , Disease Models, Animal , Electric Stimulation/methods , Functional Laterality , Glial Fibrillary Acidic Protein/metabolism , HIV Infections/pathology , Hippocampus/physiopathology , Hippocampus/virology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Long-Term Potentiation/physiology , Long-Term Potentiation/radiation effects , Magnetic Resonance Imaging/methods , Male , Mice , Mice, SCID , Microfilament Proteins , Microtubule-Associated Proteins/metabolism , Phosphopyruvate Hydratase/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/metabolism , Protons , Time Factors , Vimentin/metabolism
4.
J Neurochem ; 78(4): 874-89, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11520908

ABSTRACT

HIV-1 associated dementia is thought to be caused by neuronal damage and death in response to the production of soluble neurotoxic factors by virally infected mononuclear phagocytes. These neurotoxins include HIV-1 Tat. The ability of neurotrophins to promote cell survival prompted us to examine whether neurotrophins might also be capable of opposing the pro-apoptotic effects of Tat. Here, we show that Tat-induced neuronal apoptosis in primary cultures of rat cerebellar granule cells and in neuronally differentiated human SK-N-MC cells is profoundly inhibited by brain-derived neurotrophic factor, nerve growth factor and activity-dependent neurotrophic factor nonamer peptide. These neurotrophins activated the transcription factor NF-kappaB, and inhibition of NF-kappaB activation using a super-repressor IkappaB-alpha mutant was found to block the survival-promoting activity of the neurotrophins. Reporter gene assays and immunoblot experiments revealed that the neurotrophins also up-regulated the expression of Bcl-2, at both the transcriptional and protein levels. Overexpression of the super-repressor IkappaB-alpha mutant prevented this induction of Bcl-2 expression. Moreover, overexpression of either Bcl-2, alone, or the RelA subunit of NF-kappaB, alone, protected neurons from Tat-induced apoptosis. These findings suggest that the activation of NF-kappaB by neurotrophic factors may promote survival of neurons exposed to Tat, via regulation of anti-apoptotic genes including Bcl-2.


Subject(s)
Apoptosis/physiology , Gene Products, tat/pharmacology , NF-kappa B/metabolism , Nerve Growth Factors/pharmacology , Neurons/drug effects , AIDS Dementia Complex/physiopathology , Animals , Apoptosis/drug effects , Cell Fractionation , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cells, Cultured , Cerebellum/cytology , Dose-Response Relationship, Drug , Genes, Reporter , Humans , In Situ Nick-End Labeling , Microscopy, Fluorescence , NF-kappa B/genetics , Neurons/metabolism , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factor RelA , tat Gene Products, Human Immunodeficiency Virus
5.
Eur J Neurosci ; 13(10): 1913-22, 2001 May.
Article in English | MEDLINE | ID: mdl-11403684

ABSTRACT

Children with vertically acquired HIV-1 can present with a rapidly progressive encephalopathy and neuronal apoptosis in the first 12-18 months of life. Furthermore, abnormal prenatal platelet activating factor (PAF) signalling may result in lissencephaly, a disorder of neuronal migration. PAF, produced from human immunodeficiency virus type 1 (HIV-1) -infected brain-resident macrophages, induces neuronal apoptosis in cultured cerebellar granule neurons (CGNs) in part by activating glycogen synthase kinase 3 beta (GSK-3beta). However, PAF can also inhibit migration of CGNs that are dispersed and allowed to reaggregate. Therefore, we investigated the biological effects following activation of GSK-3beta by PAF, and whether these effects were dependent on the culture conditions of the CGNs. We show here that activation of neuronal GSK-3beta by PAF is receptor-specific, with similar kinetics of activation in both monolayer cultures of CGNs that have ceased to migrate and reaggregate cultures of CGNs that are actively migrating. However, PAF receptor activation in reaggregated CGNs inhibits neuronal migration and induces approximately half the level of neuronal apoptosis compared with PAF-treated CGN cultures that have ceased to migrate. PAF-mediated inhibition of neuronal migration in reaggregated CGNs or induction of apoptosis in CGNs that have ceased to migrate can be reversed by either PAF receptor antagonists, or the GSK-3beta inhibitors lithium or valproic acid, in a dose-dependent manner. Abnormal PAF signalling that results in GSK-3beta overactivation may represent a common mechanism for pathological defects in neuronal migration in the prenatal period and neuronal apoptosis in the postnatal period.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cerebellum/physiology , Neurons/physiology , Platelet Activating Factor/pharmacology , Animals , Apoptosis/physiology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Aggregation , Cell Death/drug effects , Cell Movement/drug effects , Cells, Cultured , Cerebellum/cytology , Cerebellum/drug effects , Enzyme Activation , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Kinetics , Platelet Activating Factor/analogs & derivatives , Rats , Rats, Sprague-Dawley
6.
J Virol ; 75(8): 3791-801, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11264368

ABSTRACT

All primate lentiviruses known to date contain one or two open reading frames with homology to the human immunodeficiency virus type 1 (HIV-1) vpr gene. HIV-1 vpr encodes a 96-amino-acid protein with multiple functions in the viral life cycle. These functions include modulation of the viral replication kinetics, transactivation of the long terminal repeat, participation in the nuclear import of preintegration complexes, induction of G2 arrest, and induction of apoptosis. The simian immunodeficiency virus (SIV) that infects African green monkeys (SIVagm) contains a vpr homologue, which encodes a 118-amino-acid protein. SIVagm vpr is structurally and functionally related to HIV-1 vpr. The present study focuses on how three specific functions (transactivation, induction of G2 arrest, and induction of apoptosis) are related to one another at a functional level, for HIV-1 and SIVagm vpr. While our study supports previous reports demonstrating a causal relationship between induction of G2 arrest and transactivation for HIV-1 vpr, we demonstrate that the same is not true for SIVagm vpr. Transactivation by SIVagm vpr is independent of cell cycle perturbation. In addition, we show that induction of G2 arrest is necessary for the induction of apoptosis by HIV-1 vpr but that the induction of apoptosis by SIVagm vpr is cell cycle independent. Finally, while SIVagm vpr retains its transactivation function in human cells, it is unable to induce G2 arrest or apoptosis in such cells, suggesting that the cytopathic effects of SIVagm vpr are species specific. Taken together, our results suggest that while the multiple functions of vpr are conserved between HIV-1 and SIVagm, the mechanisms leading to the execution of such functions are divergent.


Subject(s)
Apoptosis , Cell Cycle , Gene Products, vpr/metabolism , HIV-1 , Simian Immunodeficiency Virus , Transcriptional Activation , Active Transport, Cell Nucleus , Animals , Apoptosis/drug effects , COS Cells , Caffeine/pharmacology , Cell Cycle/drug effects , Chlorocebus aethiops , Flow Cytometry , G2 Phase/drug effects , Gene Products, vpr/genetics , Genes, vpr/genetics , HIV Long Terminal Repeat/genetics , HIV-1/genetics , Humans , Mitosis/drug effects , Models, Biological , Paclitaxel/pharmacology , Simian Immunodeficiency Virus/genetics , Transcriptional Activation/drug effects , Transduction, Genetic , Tumor Cells, Cultured , vpr Gene Products, Human Immunodeficiency Virus
7.
Ann Neurol ; 48(4): 590-602, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11026442

ABSTRACT

Intercellular adhesion molecule (ICAM)-5 (telencephalin) is unique among the ICAMs, because it is only expressed in somatodendritic membranes of telencephalic neurons. To investigate the fate of ICAM-5 during focal brain injury, we induced hypoxia-ischemia (HI) damage in adult mice by right common carotid artery ligation followed by hypoxia. ICAM-5 was detectable in serum within a 48-hour window after HI injury. In HI brain, dendritic ICAM-5 immunore-activity was abolished, but it was present in the neuropil and soma of hippocampal pyramidal, dentate granule, and some cortical and striatal neurons. After HI injury, levels of ICAM-5 protein and messenger RNA initially increased, and ICAM-5 messenger RNA expression then decreased, although protein levels continued to increase. Because HI injury induces microglial activation with increases in CD11a/CD18 (lymphocyte function antigen [LFA]-1) counterreceptors to ICAM-5, we investigated whether modulation of interactions between LFA-1 receptors and brain ICAM-5 during HI injury are associated with changes in levels of serum ICAM-5. Intracerebroventricular administration of lipopolysaccharide to activate microglia before HI injury resulted in elevated serum ICAM-5 levels compared with those in mice with only HI injury. Pretreatment with anti-LFA-1 antibodies before HI injury or LFA-1 receptor knockout mice with HI injury had markedly reduced levels of serum ICAM-5. Lipopolysaccharide levels increased, whereas LFA-1 receptor blockade or LFA-1 knockout decreased HI injury in the first 12 hours. These data suggest that during the necrotic phase of HI injury, serum ICAM-5 may be a potential marker for somatodendritic neuronal damage.


Subject(s)
Hypoxia-Ischemia, Brain/blood , Hypoxia-Ischemia, Brain/immunology , Immunoglobulins/blood , Immunoglobulins/immunology , Membrane Glycoproteins/blood , Membrane Glycoproteins/immunology , Nerve Tissue Proteins/blood , Nerve Tissue Proteins/immunology , Animals , Brain/immunology , Brain/pathology , Enzyme-Linked Immunosorbent Assay , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Male , Mice , Time Factors
8.
J Neurochem ; 74(2): 527-39, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10646503

ABSTRACT

Nerve growth factor (NGF) activates the transcription factors nuclear factor kappaB (NF-kappaB) and activator protein-1 (AP-1) in sympathetic neurons. Whereas NGF-inducible NF-kappaB is required for the survival of neurons, c-Jun has the ability to promote neuronal death. In this report, we have examined the effect of NGF withdrawal on c-Jun and NF-kappaB transcription factors in PC12 cells differentiated to a neuronal phenotype. We show that the withdrawal of NGF from these cultures results in de novo synthesis of c-Jun, increase in AP-1 activity, and down-regulation of NF-kappaB activity. To investigate how the signal transduction pathways activating c-Jun and NF-kappaB are differentially regulated by NGF, we performed transcriptional analyses. Expression of ReIA (NF-kappaB) suppressed the c-Jun-dependent transcription of c-jun, and this effect was reversed by overexpression of the coactivator p300. RelA's effects on c-Jun transcription were mediated by competitive binding of the carboxy-terminal region of RelA to the CH1 domain of p300, which also binds to c-Jun; deletion of this region abrogated the ability of RelA to inhibit c-Jun activity. Furthermore, the inhibition of endogenous NF-kappaB in NGF-maintained neuronal PC12 cells led to the induction of c-Jun synthesis and a marked increase in cell death. Together, these studies demonstrate a functional interaction between NF-kappaB and c-Jun and suggest a novel mechanism of NF-kappaB-mediated neuroprotection.


Subject(s)
NF-kappa B/physiology , Nerve Growth Factor/physiology , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-jun/physiology , Trans-Activators/physiology , Animals , Apoptosis/physiology , Cell Survival/physiology , E1A-Associated p300 Protein , NF-kappa B/antagonists & inhibitors , NF-kappa B/pharmacology , Nuclear Proteins/chemistry , Nuclear Proteins/pharmacology , PC12 Cells , Proto-Oncogene Proteins c-jun/biosynthesis , Rats , Trans-Activators/chemistry , Trans-Activators/pharmacology , Transcription, Genetic/drug effects
9.
J Immunol ; 164(3): 1333-9, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10640747

ABSTRACT

HIV-1 infection of the brain results in chronic inflammation, contributing to the neuropathogenesis of HIV-1 associated neurologic disease. HIV-1-infected mononuclear phagocytes (MP) present in inflammatory infiltrates produce neurotoxins that mediate inflammation, dysfunction, and neuronal apoptosis. Neurologic disease is correlated with the relative number of MP in and around inflammatory infiltrates and not viral burden. It is unclear whether these cells also play a neuroprotective role. We show that the chemokine, fractalkine (FKN), is markedly up-regulated in neurons and neuropil in brain tissue from pediatric patients with HIV-1 encephalitis (HIVE) compared with those without HIVE, or that were HIV-1 seronegative. FKN receptors are expressed on both neurons and microglia in patients with HIVE. These receptors are localized to cytoplasmic structures which are characterized by a vesicular appearance in neurons which may be in cell-to-cell contact with MPs. FKN colocalizes with glutamate in these neurons. Similar findings are observed in brain tissue from an adult patient with HIVE. FKN is able to potently induce the migration of primary human monocytes across an endothelial cell/primary human fetal astrocyte trans-well bilayer, and is neuroprotective to cultured neurons when coadministered with either the HIV-1 neurotoxin platelet activating factor (PAF) or the regulatory HIV-1 gene product Tat. Thus focal inflammation in brain tissue with HIVE may up-regulate neuronal FKN levels, which in turn may be a neuroimmune modulator recruiting peripheral macrophages into the brain, and in a paracrine fashion protecting glutamatergic neurons.


Subject(s)
Brain/immunology , Chemokines, CX3C/biosynthesis , Encephalitis, Viral/immunology , HIV Infections/immunology , HIV-1/immunology , Macrophage Activation/immunology , Membrane Proteins/biosynthesis , Neurons/metabolism , Neuroprotective Agents/pharmacology , Adult , Animals , Astrocytes/immunology , Brain/metabolism , Brain/pathology , Cell Movement/immunology , Cells, Cultured , Chemokine CX3CL1 , Chemokines, CX3C/administration & dosage , Chemokines, CX3C/physiology , Child , Cytoplasm/metabolism , Encephalitis, Viral/pathology , Endothelium, Vascular/immunology , Gene Products, tat/administration & dosage , HIV Infections/pathology , HIV Seronegativity/immunology , Humans , Male , Membrane Proteins/administration & dosage , Membrane Proteins/physiology , Microglia/metabolism , Microglia/pathology , Monocytes/immunology , Neurons/pathology , Platelet Activating Factor/administration & dosage , Rats , Rats, Sprague-Dawley , Up-Regulation/immunology , tat Gene Products, Human Immunodeficiency Virus
10.
Neuropathol Appl Neurobiol ; 25(5): 380-6, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10564527

ABSTRACT

Apoptosis of neurones, macrophages, and microglia occurs in the brains of paediatric patients with human immunodeficiency virus (HIV) type 1 encephalitis, which is often associated with pre-mortem neurological disease (progressive encephalopathy). We have previously reported that TUNEL-positive neurones in brain tissue from paediatric patients with HIV type 1 encephalitis and progressive encephalopathy are strikingly devoid of the pro-apoptotic gene product Bax, in marked contrast to brain-resident macrophages and microglia. Using immunocytochemical methods, the present study demonstrate that neurones in patients with HIV type 1 encephalitis and progressive encephalopathy, as well as macrophages and microglia, but not astrocytes, overexpress caspase-3, a pro-apoptotic enzyme that is proteolytically activated downstream of Bax-Bcl-2 dysregulation. Co-localization of neuronal cytoplasmic caspase-3 and nuclear TUNEL staining, a marker for fragmented DNA, was also infrequently observed in brain tissue from patients with HIV type 1 encephalitis and progressive encephalopathy. These findings suggest that vulnerable neurones in brain tissue from patients with HIV virus type 1 encephalitis and progressive encephalopathy undergo apoptosis by a mechanism that involves upregulation of caspase-3 in a pathway that is independent of Bax-Bcl-2 dysregulation. Furthermore, caspase-3 upregulation in apoptotic neurones likely occurs prior to DNA fragmentation.


Subject(s)
Brain/enzymology , Caspases/metabolism , Encephalitis/enzymology , Encephalitis/virology , HIV Infections/enzymology , HIV-1 , AIDS Dementia Complex/enzymology , AIDS Dementia Complex/genetics , Adolescent , Brain/pathology , Caspase 3 , Child , Child, Preschool , Cytoplasm/enzymology , DNA Fragmentation , Encephalitis/genetics , Encephalitis/pathology , Female , HIV Infections/pathology , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Infant , Infant, Newborn , Male , Microglia/enzymology , Neurons/enzymology
11.
J Neuroimmunol ; 98(2): 185-200, 1999 Aug 03.
Article in English | MEDLINE | ID: mdl-10430052

ABSTRACT

The mechanism(s) by which HIV-1 affects neural injury in HIV-1-associated dementia (HAD) remains unknown. To ascertain the role that cellular and viral macrophage products play in HAD neurotoxicity, we explored one potential route for neuronal demise, CXCR4. CXCR4, expressed on lymphocytes and neurons, is both a part of neural development and a co-receptor for HIV-1. Its ligand, stromal cell-derived factor-1alpha (SDF-1alpha), affects neuronal viability. GTP binding protein (G-protein) linked signaling after neuronal exposure to SDF-1alpha, virus-infected monocyte-derived macrophage (MDM) secretory products, and virus was determined. In both human and rat neurons, CXCR4 was expressed at high levels. SDF-1alpha/beta was detected predominantly in astrocytes and at low levels in MDM. SDF-1beta/beta was expressed in HAD brain tissue and upregulated in astrocytes exposed to virus infected and/or immune activated MDM conditioned media (fluids). HIV-1-infected MDM secretions, virus and SDF-1beta induced a G inhibitory (Gi) protein-linked decrease in cyclic AMP (cAMP) and increase inositol 1,4, 5-trisphosphate (IP3) and intracellular calcium. Such effects were partially blocked by antibodies to CXCR4 or removal of virus from MDM fluids. Changes in G-protein-coupled signaling correlated, but were not directly linked, to increased neuronal synaptic transmission, Caspase 3 activation and apoptosis. These data, taken together, suggest that CXCR4-mediated signal transduction may be a potential mechanism for neuronal dysfunction during HAD.


Subject(s)
AIDS Dementia Complex/immunology , Apoptosis/immunology , Neurons/cytology , Receptors, CXCR4/immunology , Signal Transduction/immunology , Animals , Astrocytes/chemistry , Astrocytes/cytology , Astrocytes/virology , Calcium/metabolism , Cell Nucleus/ultrastructure , Cell Nucleus/virology , Cells, Cultured , Chemokine CXCL12 , Chemokines, CXC/genetics , Chemokines, CXC/immunology , Excitatory Postsynaptic Potentials/immunology , Fetus/cytology , Gene Expression/immunology , HIV Envelope Protein gp120/immunology , HIV-1/growth & development , HIV-1/immunology , Hippocampus/cytology , Hippocampus/immunology , Hippocampus/virology , Humans , In Situ Nick-End Labeling , Macrophages/immunology , Macrophages/virology , Microscopy, Electron , Monocytes/immunology , Monocytes/virology , Neurons/chemistry , Neurons/virology , Oligonucleotide Probes , RNA, Messenger/analysis , Rats , Receptors, CXCR4/genetics , Reverse Transcriptase Polymerase Chain Reaction , Synaptic Transmission/immunology
12.
J Neurochem ; 73(2): 578-86, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10428053

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) Tat induces neuronal apoptosis. To examine the mechanism(s) that contribute to this process, we studied Tat's effects on glycogen synthase kinase-3beta (GSK-3beta), an enzyme that has been implicated in the regulation of apoptosis. Addition of Tat to rat cerebellar granule neurons resulted in an increase in GSK-3beta activity, which was not associated with a change in protein expression and could be abolished by the addition of an inhibitor of GSK-3beta (lithium). Lithium also enhanced neuronal survival following exposure to Tat. Coprecipitation experiments revealed that Tat can associate with GSK-3beta, but direct addition of Tat to purified GSK-3beta had no effect on enzyme activity, suggesting that Tat's effects might be mediated indirectly. As the activation of platelet activating factor (PAF) receptors is critical for the induction of neuronal death by several candidate HIV-1 neurotoxins, we determined whether PAF can also activate GSK-3beta. Application of PAF to neuronal cultures activated GSK-3beta, and coincubation with lithium ameliorated PAF-induced neuronal apoptosis. These findings are consistent with the existence of one or more pathways that can lead to GSK-3beta activation in neurons, and they suggest that the dysregulation of this enzyme could contribute to HIV-induced neuronal apoptosis.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Gene Products, tat/pharmacology , HIV-1 , Neurons/enzymology , Neurons/virology , Animals , Antimanic Agents/pharmacology , Azepines/pharmacology , Cells, Cultured , Cerebellum/cytology , Enzyme Activation/drug effects , Excitatory Amino Acid Agonists/pharmacology , Gene Products, tat/metabolism , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Lithium Chloride/pharmacology , N-Methylaspartate/pharmacology , Neurons/cytology , Neurotoxins/pharmacology , Platelet Activating Factor/metabolism , Platelet Aggregation Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Triazoles/pharmacology , tat Gene Products, Human Immunodeficiency Virus
13.
J Neuroimmunol ; 95(1-2): 55-64, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10229115

ABSTRACT

HIV-1 infection of the central nervous system can cause severe neurologic disease although only microglial cells and brain macrophages are susceptible to productive viral infection. Substances secreted by infected cells are thought to cause disease indirectly. Tumor necrosis factor alpha (TNF-alpha) is one candidate neurotoxin and is upregulated during HIV-1 infection of the brain, likely via activation of the transcription factor NF-kappaB. We used the proteasome inhibitors, MG132 and ALLN (N-acetyl-Leu-Leu-Norleucinal), to inhibit NF-kappaB activation in primary human fetal microglia (PHFM) and primary monocyte derived-macrophages, and showed that they could block TNF-alpha release stimulated by lipopolysaccharide (LPS) or TNF-alpha. In addition, we performed electrophoretic mobility shift analysis and determined that in microglia, the p50/p65 heterodimer of NF-kappaB is activated by LPS stimulation, and is inhibited by MG132. Thus, blockade of NF-kappaB activation in microglia in vitro can decrease production of TNF-alpha and may prove to be a novel strategy for treating HIV-1 dementia.


Subject(s)
AIDS Dementia Complex/immunology , Cysteine Endopeptidases/metabolism , HIV-1 , Macrophages/immunology , Microglia/immunology , Multienzyme Complexes/metabolism , Tumor Necrosis Factor-alpha/metabolism , AIDS Dementia Complex/metabolism , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , DNA Probes , Enzyme Activation/drug effects , Fetus/cytology , Gene Expression/immunology , Humans , Leupeptins/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/enzymology , Macrophages/virology , Microglia/enzymology , Microglia/virology , NF-kappa B/genetics , NF-kappa B/immunology , Phagocytosis/immunology , Proteasome Endopeptidase Complex , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
14.
J Leukoc Biol ; 65(4): 453-7, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10204573

ABSTRACT

HIV-1 infection of the nervous system causes neuronal injury and death, resulting in cognitive, motor, and behavioral dysfunction in both adults and children. In infants a characteristic feature of HIV-1 infection is impaired brain growth resulting in secondary microcephaly with onset between 2 and 4 months of age. This post-natal period of brain development is particularly vulnerable to excitotoxic neuronal injury due to the active synaptogenesis and pruning that takes place at this age associated with over-expression of excitatory amino acid (EAA) receptors. HIV-1 infection of brain microglia and perivascular macrophages results in chronic inflammation manifest pathologically as diffuse microglial activation and reactive astrogliosis. Several inflammatory products of activated microglia, including tumor necrosis factor alpha (TNF-alpha) and platelet-activating factor (PAF) have been shown to act as neuronal toxins. This toxic effect can be antagonized by blocking NMDA (or AMPA) glutamate receptors, suggesting that (weak) excitotoxicity leads to oxidative stress, neuronal injury, and apoptosis. HIV-1 infection and chronic inflammation may also contribute disruption of the blood-brain barrier and could result in further entry into the CNS of toxic viral or cellular products or additional HIV-1-infected cells. We hypothesize that prolonged microglial activation during HIV-1 infection underlies the neuronal injury and impaired brain growth in affected infants. Further investigation of the interaction between HIV-1-infected/activated microglia and developing neurons seems warranted. The current understanding of HIV neuropathogenesis implies that therapeutic strategies should target the sustained immune activation in microglia, attempt to repair the integrity of the blood-brain barrier, and provide "neuroprotection" from excitotoxic neuronal injury.


Subject(s)
Brain/growth & development , Brain/pathology , HIV Infections/pathology , HIV-1/pathogenicity , Neurons/pathology , Animals , Brain/immunology , Brain/virology , HIV Infections/complications , HIV Infections/immunology , HIV-1/immunology , Humans , Infant , Infant, Newborn , Neurons/immunology , Neurons/virology
15.
Brain Res ; 823(1-2): 1-10, 1999 Mar 27.
Article in English | MEDLINE | ID: mdl-10095006

ABSTRACT

The herbicide paraquat, bearing structural similarity to the known dopaminergic neurotoxicant MPTP, has been suggested as a potential etiologic factor in Parkinson's disease. Consideration of paraquat as a candidate neurotoxicant requires demonstration that systemic delivery produces substantia nigra dopaminergic neuron loss and the attendant neurobehavioral syndrome reflecting depletion of dopamine terminals within the striatum. To address these issues paraquat was administered systemically into adult C57 bl/6 mice, ambulatory behavior monitored, substantia nigra dopamine neuron number and striatal dopamine terminal density quantified. The data indicate that paraquat like MPTP elicits a dose-dependent decrease in substantia nigra dopaminergic neurons assessed by a Fluoro-gold prelabeling method, a decline in striatal dopamine nerve terminal density assessed by measurement of tyrosine hydroxylase immunoreactivity; and neurobehavioral syndrome characterized by reduced ambulatory activity. Taken together, these data suggest that systemically absorbed paraquat crosses the blood-brain barrier to cause destruction of dopamine neurons in the substantia nigra, consequent reduction of dopaminergic innervation of the striatum and a neurobehavioral syndrome similar to the well characterized and bona fide dopaminergic toxin MPTP.


Subject(s)
Dopamine/physiology , Herbicides , Neurons/physiology , Paraquat , Parkinson Disease, Secondary/chemically induced , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Behavior, Animal/drug effects , Cell Death/drug effects , Cell Death/physiology , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Dose-Response Relationship, Drug , Herbicides/pharmacology , Male , Mice , Mice, Inbred C57BL , Nerve Endings/pathology , Nervous System/drug effects , Paraquat/pharmacology , Parkinson Disease, Secondary/pathology , Parkinson Disease, Secondary/physiopathology , Substantia Nigra/pathology , Substantia Nigra/physiopathology , Synaptic Transmission/physiology
17.
J Neurosci Methods ; 83(2): 159-64, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9765129

ABSTRACT

A novel method for quantitative analysis of blood-brain barrier (BBB) disruption is described, using luciferase as a probe in a murine model system. Purified luciferase was delivered to mouse brain by osmotic BBB disruption with hypertonic mannitol; control animals received an intracarotid inoculation of saline prior to infusion of luciferase. Delivery of luciferase to brain tissue was then assessed by enzyme assay of tissue extracts, and by immunohistochemical staining. Luciferase activity in the brain of mannitol-treated animals was found to be significantly elevated (approx. sevenfold), when compared to activity in control (saline-treated) mice. This finding was confirmed by quantitative immunohistochemical staining of tissue sections, using a luciferase-specific antibody. These studies showed that there was an eight-fold elevation in the level of extravascular luciferase particles within the brain of mannitol-treated animals, as compared to controls. Taken together these data show that purified recombinant luciferase can be used as a sensitive probe, with which to study the integrity of the BBB.


Subject(s)
Blood-Brain Barrier/physiology , Luciferases/pharmacokinetics , Animals , Antibodies , Blood-Brain Barrier/drug effects , Brain Chemistry/physiology , Diuretics, Osmotic/pharmacology , Immunohistochemistry/methods , Liver/chemistry , Luciferases/analysis , Luciferases/immunology , Male , Mannitol/pharmacology , Mice , Mice, Inbred BALB C
18.
J Biol Chem ; 273(28): 17660-4, 1998 Jul 10.
Article in English | MEDLINE | ID: mdl-9651362

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) infection of the central nervous system results in neuronal apoptosis. Activated HIV-1-infected monocytes secrete high levels of the proinflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) and the phospholipid mediator platelet-activating factor (PAF). TNF-alpha and PAF are elevated in the central nervous system of patients with HIV-1-associated dementia. We now demonstrate that conditioned media from activated HIV-1-infected monocytes induces neuronal apoptosis, which can be prevented by co-incubation with PAF acetylhydrolase, the enzyme that catabolizes PAF in the central nervous system. Preceding apoptosis is a TNF-alpha-induced increase in neuronal ceramide levels. TNF-alpha-mediated neuronal apoptosis can also be blocked by co-incubation with PAF acetylhydrolase, or a PAF receptor antagonist. Blocking pathologic activation of PAF receptors may therefore be a pivotal step in the treatment of HIV-1-associated dementia.


Subject(s)
Central Nervous System/virology , HIV Infections/metabolism , HIV-1/pathogenicity , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, Cell Surface , Receptors, G-Protein-Coupled , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Apoptosis/drug effects , Central Nervous System/pathology , Ceramides/metabolism , Culture Media, Conditioned , HIV Infections/pathology , Humans , Monocytes/drug effects , Monocytes/virology , Neurons/drug effects , Neurons/virology , Phospholipases A/metabolism , Platelet Activating Factor/antagonists & inhibitors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
19.
J Biol Chem ; 273(28): 17852-8, 1998 Jul 10.
Article in English | MEDLINE | ID: mdl-9651389

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) infection of the central nervous system may result in neuronal apoptosis in vulnerable brain regions, including cerebral cortex and basal ganglia. The mechanisms for neuronal loss are likely to be multifactorial and indirect, since HIV-1 productively infects brain-resident macrophages and microglia but does not cause cytolytic infection of neurons in the central nervous system. HIV-1 infection of macrophages and microglia leads to production and release of diffusible factors that result in neuronal cell death, including the HIV-1 regulatory protein Tat. We demonstrate in this report that recombinant Tat1-86 and Tat peptides containing the basic region induce neuronal apoptosis in approximately 50% of vulnerable neurons in both rat and human neuronal cultures, and this apoptotic cell death is mediated by release of the pro-inflammatory cytokine tumor necrosis factor alpha, and by activation of glutamate receptors of the non-N-methyl-D-aspartate subtype. Finally, we show that Tat-induced apoptosis of human neuronal cell cultures occurs in the absence of activation of the transcription factor NFkappaB. These findings further define cellular pathways activated by Tat, that dysregulate production of tumor necrosis factor alpha, and lead to activation of glutamate receptors and neuronal death during HIV-1 infection of the central nervous system.


Subject(s)
Apoptosis/physiology , Gene Products, tat/physiology , HIV-1/metabolism , NF-kappa B/metabolism , Neurons/pathology , Tumor Necrosis Factor-alpha/physiology , Animals , Base Sequence , Cells, Cultured , DNA Probes , Humans , Oxidative Stress , Rats , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction , Tumor Cells, Cultured , tat Gene Products, Human Immunodeficiency Virus
20.
Neurosci Lett ; 247(2-3): 99-102, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9655602

ABSTRACT

Parkinson's disease is associated with progressive loss of nigrostriatal dopamine (DA). Models of the disorder, produced with neurotoxins (N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine or 6-hydroxydopamine) that selectively lesion DA neurons, are characterized by acute removal and gradual recovery of DA. We report slowly progressive loss of DA in ipsilateral nucleus accumbens following profound (>90%) acute unilateral depletion of DA in the caudate-putamen of neonatal rats, from 50% at age 27 days to 94% by 100 days. Metabolic turnover of DA markedly increased in ipsilateral accumbens, and may yield tissue-damaging neurotoxic by-products. This paradigm may help in elucidating mechanisms responsible for gradual degeneration of DA neurons and for screening potential neuroprotective agents.


Subject(s)
Nerve Degeneration/chemically induced , Neurotoxins/toxicity , Nucleus Accumbens/drug effects , Oxidopamine/toxicity , 3,4-Dihydroxyphenylacetic Acid/analysis , Animals , Animals, Newborn , Caudate Nucleus/drug effects , Chromatography, High Pressure Liquid , Disease Progression , Dopamine/physiology , Homovanillic Acid/analysis , Nucleus Accumbens/pathology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/pathology , Putamen/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...