1
|
Nuzhnyi EP, Brsikyan LA, Fedotova EY, Illarioshkin SN. [Cerebellar degeneration associated with HIV infection]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:123-130. [PMID: 37315251 DOI: 10.17116/jnevro2023123051123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
OBJECTIVE To describe the features of the clinical presentation and evaluate the incidence of HIV-associated cerebellar degeneration in patients with progressive cerebellar ataxia. MATERIAL AND METHODS Three hundred and seventy-seven patients with progressive cerebellar ataxia were studied. Brain MRI study, assessment by the Scale for the Assessment and Rating of Ataxia (SARA), screening for cognitive impairment by the Montreal Cognitive Assessment Scale (MoCA) were performed. In patients with HIV infection, autoimmune, deficient and other causes of ataxia, as well as opportunistic infections, multiple system atrophy and frequent forms of hereditary spinocerebellar ataxias were excluded. RESULTS Five patients (1.3%) were identified with a combination of cerebellar ataxia and HIV infection (2 men, 3 women, aged 31 to 52 years). The median duration of HIV infection was 5 years, the duration of ataxia was 1 year. In the clinical findings, in addition to progressive ataxia, pyramidal signs, dysphagia, less often ophthalmoparesis, dystonia, postural hand tremor, affective and mild cognitive impairment were observed. In three patients, brain MRI revealed signs of olivopontocerebellar atrophy, two patients had isolated cerebellar degeneration (mainly of the vermis). All patients received combination of antiretroviral therapy in various regimens, but despite this, ataxia was progressive. CONCLUSION HIV infection is a rare cause of cerebellar degeneration. This diagnosis remains a diagnosis of exclusion to this day. Cerebellar degeneration can occur and progress even after achieving a stable remission of HIV infection while taking highly active antiretroviral therapy.
Collapse
Affiliation(s)
- E P Nuzhnyi
- Research Center of Neurology, Moscow, Russia
| | | | | | | |
Collapse
|
2
|
Fields JA, Ellis RJ. HIV in the cART era and the mitochondrial: immune interface in the CNS. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 145:29-65. [PMID: 31208526 DOI: 10.1016/bs.irn.2019.04.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
HIV-associated neurocognitive disorders (HAND) persist in the era of effective combined antiretroviral therapy (cART). A large body of literature suggests that mitochondrial dysfunction is a prospective etiology of HAND in the cART era. While viral load is often suppressed and the immune system remains intact in HIV+ patients on cART, evidence suggests that the central nervous system (CNS) acts as a reservoir for virus and low-level expression of viral proteins, which interact with mitochondria. In particular, the HIV proteins glycoprotein 120, transactivator of transcription, viral protein R, and negative factor have each been linked to mitochondrial dysfunction in the brain. Moreover, cART drugs have also been shown to have detrimental effects on mitochondrial function. Here, we review the evidence generated from human studies, animal models, and in vitro models that support a role for HIV proteins and/or cART drugs in altered production of adenosine triphosphate, mitochondrial dynamics, mitophagy, calcium signaling and apoptosis, oxidative stress, mitochondrial biogenesis, and immunometabolism in the CNS. When insightful, evidence of HIV or cART-induced mitochondrial dysfunction in the peripheral nervous system or other cell types is discussed. Lastly, therapeutic approaches to targeting mitochondrial dysfunction have been summarized with the aim of guiding new investigations and providing hope that mitochondrial-based drugs may provide relief for those suffering with HAND.
Collapse
Affiliation(s)
- Jerel Adam Fields
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States.
| | - Ronald J Ellis
- Department of Neuroscience, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
3
|
Wächter C, Eiden LE, Naumann N, Depboylu C, Weihe E. Loss of cerebellar neurons in the progression of lentiviral disease: effects of CNS-permeant antiretroviral therapy. J Neuroinflammation 2016; 13:272. [PMID: 27737697 PMCID: PMC5064958 DOI: 10.1186/s12974-016-0726-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/20/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The majority of investigations on HIV-associated neurocognitive disorders (HAND) neglect the cerebellum in spite of emerging evidence for its role in higher cognitive functions and dysfunctions in common neurodegenerative diseases. METHODS We systematically investigated the molecular and cellular responses of the cerebellum as contributors to lentiviral infection-induced neurodegeneration, in the simian immunodeficiency virus (SIV)-infected rhesus macaque model for HIV infection and HAND. Four cohorts of animals were studied: non-infected controls, SIV-infected asymptomatic animals, and SIV-infected AIDS-diseased animals with and without brain-permeant antiretroviral treatment. The antiretroviral utilized was 6-chloro-2',3'-dideoxyguanosine (6-Cl-ddG), a CNS-permeable nucleoside reverse transcriptase inhibitor. Quantitation of granule cells and Purkinje cells, of an established biomarker of SIV infection (gp41), of microglial/monocyte/macrophage markers (IBA-1, CD68, CD163), and of the astroglial marker (GFAP) were used to reveal cell-specific cerebellar responses to lentiviral infection and antiretroviral therapy (ART). The macromolecular integrity of the blood brain barrier was tested by albumin immunohistochemistry. RESULTS Productive CNS infection was observed in the symptomatic stage of disease, and correlated with extensive microglial/macrophage and astrocyte activation, and widespread macromolecular blood brain barrier defects. Signs of productive infection, and inflammation, were reversed upon treatment with 6-Cl-ddG, except for a residual low-grade activation of microglial cells and astrocytes. There was an extensive loss of granule cells in the SIV-infected asymptomatic cohort, which was further increased in the symptomatic stage of the disease and persisted after 6-Cl-ddG (administered after the onset of symptoms of AIDS). In the symptomatic stage, Purkinje cell density was reduced. Purkinje cell loss was likewise unaffected by 6-Cl-ddG treatment at this time. CONCLUSIONS Our findings suggest that neurodegenerative mechanisms are triggered by SIV infection early in the disease process, i. e., preceding large-scale cerebellar productive infection and marked neuroinflammation. These affect primarily granule cells early in disease, with later involvement of Purkinje cells, indicating differential vulnerability of the two neuronal populations. The results presented here indicate a role for the cerebellum in neuro-AIDS. They also support the conclusion that, in order to attenuate the development of motor and cognitive dysfunctions in HIV-positive individuals, CNS-permeant antiretroviral therapy combined with anti-inflammatory and neuroprotective treatment is indicated even before overt signs of CNS inflammation occur.
Collapse
Affiliation(s)
- Christian Wächter
- Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps University Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany
| | - Lee E Eiden
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - Nedye Naumann
- Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps University Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany
| | - Candan Depboylu
- Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps University Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany.,Experimental Neurology, Department of Neurology, Philipps University Marburg, Marburg, Germany
| | - Eberhard Weihe
- Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps University Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany.
| |
Collapse
|
4
|
HCMV Induces Dysregulation of Glutamate Uptake and Transporter Expression in Human Fetal Astrocytes. Neurochem Res 2014; 39:2407-18. [DOI: 10.1007/s11064-014-1445-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 09/23/2014] [Accepted: 09/25/2014] [Indexed: 11/26/2022]
|
5
|
Targeting the glutamatergic system for the treatment of HIV-associated neurocognitive disorders. J Neuroimmune Pharmacol 2013; 8:594-607. [PMID: 23553365 PMCID: PMC3661915 DOI: 10.1007/s11481-013-9442-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 02/08/2013] [Indexed: 12/22/2022]
Abstract
The accumulation of excess glutamate in the extracellular space as a consequence of CNS trauma, neurodegenerative diseases, infection, or deregulation of glutamate clearance results in neuronal damage by excessive excitatory neurotransmission. Glutamate excitotoxicity is thought to be one of several mechanisms by which HIV exerts neurotoxicity that culminates in HIV-associated neurocognitive disorders (HAND). Excess glutamate is released upon HIV infection of macrophage/microglial cells and has been associated with neurotoxicity mediated by gp120, transactivator of transcription (Tat) and other HIV proteins. Several strategies have been used over the years to try to prevent glutamate excitotoxicity. Since the main toxic effects of excess glutamate are thought to be due to excitotoxicity from over activation of glutamate receptors, antagonists of these receptors have been popular therapeutic targets. Early work to ameliorate the effects of excess extracellular glutamate focused on NMDA receptor antagonism, but unfortunately, potent blockade of this receptor has been fraught with side effects. One alternative to direct receptor blockade has been the inhibition of enzymes responsible for the production of glutamate such as glutaminase and glutamate carboxypeptidase II. Another approach has been to regulate the transporters responsible for modulation of extracellular glutamate such as excitatory amino acid transporters and the glutamate-cystine antiporter. There is preliminary experimental evidence that these approaches have potential therapeutic utility for the treatment of HAND. These efforts however, are at an early stage where the next steps are dependent on the identification of drug-like inhibitors as well as the development of predictive neuroAIDS animal models.
Collapse
|
6
|
Buch S, Yao H, Guo M, Mori T, Mathias-Costa B, Singh V, Seth P, Wang J, Su TP. Cocaine and HIV-1 interplay in CNS: cellular and molecular mechanisms. Curr HIV Res 2012; 10:425-8. [PMID: 22591366 DOI: 10.2174/157016212802138823] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 01/12/2012] [Accepted: 04/21/2012] [Indexed: 01/20/2023]
Abstract
Although antiretrovirals are the mainstay of therapy against HIV infection, neurological complications associated with the virus continue to hamper quality of life of the infected individuals. Drugs of abuse in the infected individuals further fuel the epidemic. Epidemiological studies have demonstrated that abuse of cocaine resulted in acceleration of HIV infection and the progression of NeuroAIDS. Cocaine has not only been shown to play a crucial role in promoting virus replication, but also has diverse but often deleterious effects on various cell types of the CNS. In the neuronal system, cocaine exposure results in neuronal toxicity and also potentiates gp120-induced neurotoxicity. In the astroglia and microglia, cocaine exposure leads to up-regulation of pro-inflammatory mediators such as cytokines and chemokines. These in turn, can lead to neuroinflammation and transmission of toxic responses to the neurons. Additionally, cocaine exposure can also lead to leakiness of the blood-brain barrier that manifests as enhanced transmigraiton of leukocytes/monocytes into the CNS. Both in vitro and in vivo studies have provided valuable tools in exploring the role of cocaine in mediating HIV-associated neuropathogenesis. This review summarizes previous studies on the mechanism(s) underlying the interplay of cocaine and HIV as it relates to the CNS.
Collapse
Affiliation(s)
- Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Mithal DS, Banisadr G, Miller RJ. CXCL12 signaling in the development of the nervous system. J Neuroimmune Pharmacol 2012; 7:820-34. [PMID: 22270883 DOI: 10.1007/s11481-011-9336-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 12/14/2011] [Indexed: 11/30/2022]
Abstract
Chemokines are small, secreted proteins that have been shown to be important regulators of leukocyte trafficking and inflammation. All the known effects of chemokines are transduced by action at a family of G protein coupled receptors. Two of these receptors, CCR5 and CXCR4, are also known to be the major cellular receptors for HIV-1. Consideration of the evolution of the chemokine family has demonstrated that the chemokine Stromal cell Derived Factor-1 or SDF1 (CXCL12) and its receptor CXCR4 are the most ancient members of the family and existed in animals prior to the development of a sophisticated immune system. Thus, it appears that the original function of chemokine signaling was in the regulation of stem cell trafficking and development. CXCR4 signaling is important in the development of many tissues including the nervous system. Here we discuss the manner in which CXCR4 signaling can regulate the development of different structures in the central and peripheral nervous systems and the different strategies employed to achieve these effects.
Collapse
Affiliation(s)
- Divakar S Mithal
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | |
Collapse
|
8
|
Zhang J, Liu J, Katafiasz B, Fox H, Xiong H. HIV-1 gp120-induced axonal injury detected by accumulation of β-amyloid precursor protein in adult rat corpus callosum. J Neuroimmune Pharmacol 2011; 6:650-7. [PMID: 21286834 PMCID: PMC3165079 DOI: 10.1007/s11481-011-9259-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 01/18/2011] [Indexed: 10/18/2022]
Abstract
HIV-1 brain infection induces neurodegeneration. While most studies focus on HIV-1-mediated neuronal injury, relatively few have investigated HIV-1-associated white matter damage. Corpus callosum (CC) is one of frequently involved white matter structures in HIV-1-associated white matter damage. Utilizing a model of ex vivo treatment of brain slice containing CC with HIV-1 glycoprotein 120 (gp120), we examined axonal injury by analyzing β-amyloid precursor protein (β-APP) accumulation in the axon. Incubation of CC slice with gp120 produced a significant higher density of β-APP in the CC tissue compared with non-gp120-treated controls, suggesting the presence of axonal damage in the CC. The gp120-induced CC axonal damage was blocked by a chemokine CXCR4 receptor antagonist T140 but not by an NMDA receptor blocker MK801 as demonstrated by Western blot analysis of β-APP, indicating that gp120 evokes the CC axonal injury through CXCR4 receptor. Immunocytochemical studies revealed a surprisingly high density of CXCR4-positive immunoreactivity in the CC. The CXCR4-positive labeling was distributed along the nerve fibers. Moreover, double labeling of anti-CXCR4 with either anti-neuronal nuclei or anti-myelin/oligodendrocyte-specific protein antibody revealed co-localization of CXCR4 and myelin/oligodendrocytes in some fiber-like structures, inferring that some neurons and oligodendrocytes in the CC express CXCR4. Taken together, these results indicate that gp120 induced axonal damage via CXCR4 in the CC.
Collapse
Affiliation(s)
- Jingdong Zhang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | | | | | | | |
Collapse
|
9
|
Buch S, Yao H, Guo M, Mori T, Su TP, Wang J. Cocaine and HIV-1 interplay: molecular mechanisms of action and addiction. J Neuroimmune Pharmacol 2011; 6:503-15. [PMID: 21766222 DOI: 10.1007/s11481-011-9297-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 07/07/2011] [Indexed: 01/16/2023]
Abstract
Human immunodeficiency virus (HIV) infection is now being driven by drug-abusing populations. Epidemiological studies on drug abusers with AIDS link abuse of cocaine, even more than other drugs, to increased incidence of HIV seroprevalence and progression to AIDS. Both cell culture and animal studies demonstrate that cocaine can both potentiate HIV replication and can potentiate HIV proteins to cause enhanced glial cell activation, neurotoxicity, and breakdown of the blood-brain barrier. Based on the ability of both HIV proteins and cocaine to modulate NMDA receptor on neurons, NMDA receptors have been suggested as a common link underlying the crosstalk between drug addiction and HIV infection. While the role of dopamine system as a major target of cocaine cannot be overlooked, recent studies on the role of sigma receptors in mediating the effects of cocaine in both cell and organ systems warrants a deeper understanding of their functional role in the field. In this review, recent findings on the interplay of HIV infection and cocaine abuse and their possible implications in mode of action and/or addiction will be discussed.
Collapse
Affiliation(s)
- Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center (DRC 8011), University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| | | | | | | | | | | |
Collapse
|
10
|
Bjugstad KB, Flitter WD, Arendash GW. Intracerebroventricular infusions of gp120 inhibit weight gain and induce atrophy in the hippocampus and neostriatum without affecting cognition. ACTA ACUST UNITED AC 2006; 2:15-31. [PMID: 16873203 DOI: 10.1300/j128v02n04_02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The HIV envelope protein, gp120, has been proposed to be a key agent in the development of AIDS dementia complex (ADC). To elucidate CNS effects that gp120 alone may be inducing in ADC, the present study investigated changes in weight, motor activity, cognitive function and corresponding neuropathology in rats given daily bilateral infusions of gp120 intracerebroventricularly for 7 days. gp120 inhibited weight gain, but had no measurable effects on motor activity or water maze cognitive performance. Nonetheless, gp120 infusions did induce both hippocampal and neostriatal atrophy. Thus, gp120 alone can cause ADC-related neuropathologic and weight changes, but gp120 alone was not sufficient to induce impairments in spatial learning and memory.
Collapse
Affiliation(s)
- Kimberly B Bjugstad
- Department of Psychiatry, University of Colorado Health Sciences Center, 4200 E Ninth Ave, Denver, CO, USA
| | | | | |
Collapse
|
11
|
Sui Z, Fan S, Sniderhan L, Reisinger E, Litzburg A, Schifitto G, Gelbard HA, Dewhurst S, Maggirwar SB. Inhibition of mixed lineage kinase 3 prevents HIV-1 Tat-mediated neurotoxicity and monocyte activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 177:702-11. [PMID: 16785569 DOI: 10.4049/jimmunol.177.1.702] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The HIV-1 gene products Tat and gp120 are toxic to neurons and can activate cells of myeloid origin, properties that are thought to contribute to the clinical manifestations of HIV-1-associated dementia (HAD). To investigate the intracellular signaling mechanisms involved in these events, the effect of Tat and gp120 on mixed lineage kinase (MLK) 3 activation was examined. Tat and gp120 were shown to induce autophosphorylation of MLK3 in primary rat neurons; this was abolished by the addition of an inhibitor of MLK3 (CEP1347). CEP1347 also enhanced survival of both rat and human neurons and inhibited the activation of human monocytes after exposure to Tat and gp120. Furthermore, overexpression of wild-type MLK3 led to the induction of neuronal death, whereas expression of a dominant negative MLK3 mutant protected neurons from the toxic effects of Tat. MLK3-dependent downstream signaling events were implicated in the neuroprotective and monocyte-deactivating pathways triggered by CEP1347. Thus, the inhibition of p38 MAPK and JNK protected neurons from Tat-induced apoptosis, whereas the inhibition of p38 MAPK, but not of JNK, was sufficient to prevent Tat- and gp120-mediated activation of monocytes. These results suggest that the normal function of MLK3 is compromised by HIV-1 neurotoxins (Tat, gp120), resulting in the activation of downstream signaling events that result in neuronal death and monocyte activation (with release of inflammatory cytokines). In aggregate, our data define MLK3 as a promising therapeutic target for intervention in HAD.
Collapse
Affiliation(s)
- Ziye Sui
- Interdepartmental Program in Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Abstract
Most neurological and psychiatric disorders involve selective or preferential impairments of neurotransmitter systems. Therefore, studies of functional transmitter pathophysiology in human brain are of unique importance in view of the development of effective, mechanism-based, therapeutic modalities. It is well known that central nervous system functional proteins, including receptors, transporters, ion channels, and enzymes, can exhibit high heterogeneity in terms of structure, function, and pharmacological profile. If the existence of types and subtypes of functional proteins amplifies the possibility of developing selective drugs, such heterogeneity certainly increases the likelihood of interspecies differences. It is therefore essential, before choosing animal models to be used in preclinical pharmacology experimentation, to establish whether functionally corresponding proteins in men and animals also display identical pharmacological profiles. Because of evidence that scaffolding proteins, trafficking between plasma membrane and intracellular pools, phosphorylation and allosteric modulators can affect the function of receptors and transporters, experiments with human clones expressed in host cells where the environment of native receptors is rarely reproduced should be interpreted with caution. Thus, the use of neurosurgically removed fresh human brain tissue samples in which receptors, transporters, ion channels, and enzymes essentially retain their natural environment represents a unique experimental approach to enlarge our understanding of human brain processes and to help in the choice of appropriate animal models. Using this experimental approach, many human brain functional proteins, in particular transmitter receptors, have been characterized in terms of localization, function, and pharmacological properties.
Collapse
Affiliation(s)
- Maurizio Raiteri
- Dipartimento di Medicina Sperimentale, Sezione di Farmacologia e Tossicologia, Viale Cembrano 4, 16148 Genova, Italy.
| |
Collapse
|
13
|
Longordo F, Feligioni M, Chiaramonte G, Sbaffi PF, Raiteri M, Pittaluga A. The human immunodeficiency virus-1 protein transactivator of transcription up-regulates N-methyl-D-aspartate receptor function by acting at metabotropic glutamate receptor 1 receptors coexisting on human and rat brain noradrenergic neurones. J Pharmacol Exp Ther 2006; 317:1097-105. [PMID: 16489129 DOI: 10.1124/jpet.105.099630] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We investigated the effects of the human immunodeficiency virus-1 transactivator of transcription (Tat) on the release of norepinephrine (NE) from human and rat brain synaptosomes. Tat could not evoke directly release of [3H]NE. In the presence of Tat (1 nM), N-methyl-D-aspartate (NMDA) concentrations unable to release (human synaptosomes) or slightly releasing (rat synaptosomes) [3H]NE became very effective. The NMDA/Tat-evoked release depends on NMDA receptors (NMDARs) since it was abolished by MK-801 (dizocilpine). Tat binding at NMDARs was excluded. The NMDA-induced release of [3H]NE in the presence of glycine was further potentiated by Tat. The release evoked by NMDA/glycine/Tat depends on metabotropic glutamate receptor 1 (mGluR1) activation, since it was halved by mGluR1 antagonists. Tat seems to act at the glutamate recognition site of mGluR1. Recently, Tat was shown to release [3H]acetylcholine from human cholinergic terminals; here, we demonstrate that this effect is also mediated by presynaptic mGluR1. The peptide sequence Tat41-60, but not Tat61-80, mimicked Tat. Phospholipase C, protein kinase C, and cytosolic tyrosine kinase are involved in the NMDA/glycine/Tat-evoked [3H]NE release. To conclude, Tat can represent a potent pathological agonist at mGlu1 receptors able to release acetylcholine from human cholinergic terminals and up-regulate NMDARs mediating NE release from human and rat noradrenergic terminals.
Collapse
Affiliation(s)
- Fabio Longordo
- Pharmacology and Toxicology Section, Department of Experimental Medicine, University of Genova, Genova, Italy
| | | | | | | | | | | |
Collapse
|
14
|
Kadiu I, Glanzer JG, Kipnis J, Gendelman HE, Thomas MP. Mononuclear phagocytes in the pathogenesis of neurodegenerative diseases. Neurotox Res 2006; 8:25-50. [PMID: 16260384 DOI: 10.1007/bf03033818] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Brain mononuclear phagocytes (MP, bone marrow monocyte-derived macrophages, perivascular macrophages, and microglia) function to protect the nervous system by acting as debris scavengers, killers of microbial pathogens, and regulators of immune responses. MP are activated by a variety of environmental cues and such inflammatory responses elicit cell injury and death in the nervous system. MP immunoregulatory responses include secretion of neurotoxic factors, mobilization of adaptive immunity, and cell chemotaxis. This incites tissue remodelling and blood-brain barrier dysfunction. As disease progresses, MP secretions engage neighboring cells in a vicious cycle of autocrine and paracrine amplification of inflammation leading to tissue injury and ultimately destruction. Such pathogenic processes tilt the balance between the relative production of neurotrophic and neurotoxic factors and to disease progression. The ultimate effects that brain MP play in disease revolves "principally" around their roles in neurodegeneration. Importantly, common functions of brain MP in neuroimmunity link highly divergent diseases (for example, human immunodeficiency virus type-one associated dementia, Alzheimer's disease and Parkinson's disease). Research into this process from our own laboratories and those of others seek to harness MP inflammatory processes with the intent of developing therapeutic interventions that block neurodegenerative processes and improve the quality of life in affected people.
Collapse
Affiliation(s)
- I Kadiu
- Laboratory of Neuroregeneration, Department of Pharmacology and Experimental Neuroscience, Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | | | | | | | |
Collapse
|
15
|
Kaul M, Lipton SA. Experimental and potential future therapeutic approaches for HIV-1 associated dementia targeting receptors for chemokines, glutamate and erythropoietin. Neurotox Res 2005; 8:167-86. [PMID: 16260394 DOI: 10.1007/bf03033828] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Severe and debilitating neurological problems that include behavioral abnormalities, motor dysfunction and frank dementia can occur after infection with the human immunodeficiency virus-1 (HIV-1). Infected peripheral immune-competent cells, in particular macrophages, infiltrate the central nervous system (CNS) and provoke a neuropathological response involving all cell types in the brain. HIV-1 infection results in activation of chemokine receptors, inflammatory mediators, extracellular matrix-degrading enzymes and glutamate receptor-mediated excitotoxicity, all of which can trigger numerous downstream signaling pathways that result in disruption of neuronal and glial function. Despite many major improvements in the control of viral infection in the periphery, a truly effective therapy for HIV-1 associated dementia is currently not available. This review will discuss experimental and potentially future therapeutic strategies based on recently uncovered pathologic mechanisms contributing to neuronal damage induced by HIV-1.
Collapse
Affiliation(s)
- M Kaul
- Center for Neuroscience and Aging Research, The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | |
Collapse
|
16
|
Li W, Galey D, Mattson MP, Nath A. Molecular and cellular mechanisms of neuronal cell death in HIV dementia. Neurotox Res 2005; 8:119-34. [PMID: 16260390 DOI: 10.1007/bf03033824] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The deaths of neurons, astrocytes and endothelial cells have been described in patients with HIV (human immunodeficiency virus) dementia. HIV-1 does not infect neurons; instead, neurotoxic substances shed by infected glia and macrophages can induce a form of programmed cell death called apoptosis in neurons. These neurotoxins include the HIV-1 proteins Tat and gp120, as well as pro-inflammatory cytokines, chemokines, excitotoxins and proteases. In this article we review the evidence for apoptosis of various cell types within the brain of HIV-infected patients, and describe in vitro and in vivo experimental studies that have elucidated the mechanisms by which HIV causes apoptosis of brain cells.
Collapse
Affiliation(s)
- W Li
- RT Johnson Division of Neuroimmunology and Neurological Infection, Department of Neurology, Johns Hopkins University, Baltimore, MD 21287, USA
| | | | | | | |
Collapse
|
17
|
Banks WA, Robinson SM, Nath A. Permeability of the blood–brain barrier to HIV-1 Tat. Exp Neurol 2005; 193:218-27. [PMID: 15817280 DOI: 10.1016/j.expneurol.2004.11.019] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2004] [Revised: 11/09/2004] [Accepted: 11/16/2004] [Indexed: 11/28/2022]
Abstract
Infection with human immunodeficiency virus-1 (HIV-1) is associated with dysfunctions of the central nervous system (CNS). HIV-1 induces its effects on the CNS by a variety of mechanisms, including by shedding the neurotoxic viral proteins such as gp120 and Tat. Both HIV-1 and gp120 have been shown to cross the blood-brain barrier (BBB). It is has not been determined, however, whether blood-borne Tat can cross the BBB. Here, we found that Tat crosses the BBB by a nonsaturable mechanism with a unidirectional influx rate of about 0.490 microl/g/min. About 0.126% of an intravenous dose of Tat enters each g of brain. Radioactively labeled albumin injected simultaneously did not cross the BBB. The hypothalamus, occipital cortex, and hippocampus were the regions of the brain most permeable to Tat. Nonsaturable brain-to-blood efflux also occurred, most likely with reabsorption into the blood of the cerebrospinal fluid. In conclusion, we found that Tat crossed the BBB bidirectionally. Such permeability could provide a mechanism by which Tat produced on one side of the BBB could affect neural or immune function on the other side.
Collapse
Affiliation(s)
- William A Banks
- Division of Geriatrics, Department of Internal Medicine, GRECC, Veterans Affairs Medical Center-St. Louis and Saint Louis University School of Medicine, 915 N. Grand Boulevard, St. Louis, MO 63106, USA.
| | | | | |
Collapse
|
18
|
Walsh KA, Megyesi JF, Wilson JX, Crukley J, Laubach VE, Hammond RR. Antioxidant protection from HIV-1 gp120-induced neuroglial toxicity. J Neuroinflammation 2004; 1:8. [PMID: 15285794 PMCID: PMC483061 DOI: 10.1186/1742-2094-1-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2004] [Accepted: 05/27/2004] [Indexed: 11/17/2022] Open
Abstract
Background The pathogenesis of HIV-1 glycoprotein 120 (gp120) associated neuroglial toxicity remains unresolved, but oxidative injury has been widely implicated as a contributing factor. In previous studies, exposure of primary human central nervous system tissue cultures to gp120 led to a simplification of neuronal dendritic elements as well as astrocytic hypertrophy and hyperplasia; neuropathological features of HIV-1-associated dementia. Gp120 and proinflammatory cytokines upregulate inducible nitric oxide synthase (iNOS), an important source of nitric oxide (NO) and nitrosative stress. Because ascorbate scavenges reactive nitrogen and oxygen species, we studied the effect of ascorbate supplementation on iNOS expression as well as the neuronal and glial structural changes associated with gp120 exposure. Methods Human CNS cultures were derived from 16–18 week gestation post-mortem fetal brain. Cultures were incubated with 400 μM ascorbate-2-O-phosphate (Asc-p) or vehicle for 18 hours then exposed to 1 nM gp120 for 24 hours. The expression of iNOS and neuronal (MAP2) and astrocytic (GFAP) structural proteins was examined by immunohistochemistry and immunofluorescence using confocal scanning laser microscopy (CSLM). Results Following gp120 exposure iNOS was markedly upregulated from undetectable levels at baseline. Double label CSLM studies revealed astrocytes to be the prime source of iNOS with rare neurons expressing iNOS. This upregulation was attenuated by the preincubation with Asc-p, which raised the intracellular concentration of ascorbate. Astrocytic hypertrophy and neuronal injury caused by gp120 were also prevented by preincubation with ascorbate. Conclusions Ascorbate supplementation prevents the deleterious upregulation of iNOS and associated neuronal and astrocytic protein expression and structural changes caused by gp120 in human brain cell cultures.
Collapse
Affiliation(s)
- Kimberley A Walsh
- Department of Pathology, London Health Sciences Centre, University of Western Ontario, London, ON, Canada
| | - Joseph F Megyesi
- Department of Pathology, London Health Sciences Centre, University of Western Ontario, London, ON, Canada
- Department Clinical Neurological Sciences, London Health Sciences Centre, University of Western Ontario, London, ON, Canada
| | - John X Wilson
- Department Physiology, University of Western Ontario, London, ON, Canada
| | - Jeff Crukley
- Department of Pathology, London Health Sciences Centre, University of Western Ontario, London, ON, Canada
| | - Victor E Laubach
- Department of Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Robert R Hammond
- Department of Pathology, London Health Sciences Centre, University of Western Ontario, London, ON, Canada
- Department Clinical Neurological Sciences, London Health Sciences Centre, University of Western Ontario, London, ON, Canada
| |
Collapse
|
19
|
Bachis A, Mocchetti I. The chemokine receptor CXCR4 and not the N-methyl-D-aspartate receptor mediates gp120 neurotoxicity in cerebellar granule cells. J Neurosci Res 2004; 75:75-82. [PMID: 14689450 DOI: 10.1002/jnr.10826] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The human immunodeficiency virus type 1 (HIV-1) glycoprotein gp120 causes neuronal cell death; however, the molecular mechanisms of the neurotoxic effect remain largely unresolved. It has been suggested that gp120 evokes cell death by inducing the release of neurotoxins, including glutamate. The objective of this work was to examine the role of glutamate in gp120-mediated neurotoxicity. We used as an experimental tool cerebellar granule cells prepared from 8-day-old rat cerebella, in which both glutamate and gp120 cause cell death. Cerebellar granule neurons were exposed to gp120 or glutamate alone or in combination with the glutamate receptor antagonist MK801 as well as other antiglutamatergic compounds. Cell viability was measured at various times by using several markers of cell death and apoptosis. MK801, at a concentration that blocked glutamate-induced neuronal cell death, failed to prevent gp120-mediated apoptotic cell death. Moreover, interleukin-10, which has previously been shown to block glutamate toxicity in these neurons, was not neuroprotective against gp120. Because gp120 toxicity is mediated by activation of the chemokine receptor CXCR4, neurons were incubated with the CXCR4 inhibitor AMD3100. This compound prevented gp120- but not glutamate-mediated cell death. These findings suggest that gp120 is toxic to neurons even in the absence of the virus and that the toxic mechanism involves primarily activation of CXCR4 receptor. Therefore, antagonists to the CXCR4 receptor may be more suitable compounds for inhibiting HIV-1 neurotoxicity.
Collapse
Affiliation(s)
- Alessia Bachis
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | | |
Collapse
|
20
|
Acquas E, Bachis A, Nosheny RL, Cernak I, Mocchetti I. Human immunodeficiency virus type 1 protein gp120 causes neuronal cell death in the rat brain by activating caspases. Neurotox Res 2004; 5:605-15. [PMID: 15111237 DOI: 10.1007/bf03033180] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection of the central nervous system is associated with microglia activation and neuronal apoptosis, alterations that are also caused by the HIV-1 envelope glycoprotein 120 (gp120) alone. This study was undertaken to examine the onset of gp120 neurotoxicity, the type of cell death and which cells of the adult rat brain are more sensitive to the toxic action of gp120. Gp120 or vehicle were injected chronically (daily for 3 or 7 days) into the lateral ventricle. Magnetic resonance imaging revealed hypertensive areas in the cortical and hippocampal gray matter in gp120-treated rats 7-10 days after the first injection, suggesting vasogenic edema. This phenomenon was accompanied by an enlargement of the lateral and third ventricles. Immunohistochemical analyses were then carried out to examine the toxic effect of gp120 at a cellular level. Several markers of apoptosis, including activated caspase-3 were observed at both 3 and 7 days throughout brains of gp120-treated rats, especially in the cerebral cortex. In this area, most of the apoptotic cells exhibited a pyramidal shape and were Nissl positive, indicative of neurons. Few non-neuronal cells exhibited signs of apoptosis. The results of the present study support the notion that gp120 is neurotoxic in vivo and provide evidence that gp120 activates a caspase-dependent apoptotic pathway.
Collapse
Affiliation(s)
- Elio Acquas
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | | | | | | | | |
Collapse
|
21
|
The human immunodeficiency virus-1 protein Tat and its discrete fragments evoke selective release of acetylcholine from human and rat cerebrocortical terminals through species-specific mechanisms. J Neurosci 2003. [PMID: 12890775 DOI: 10.1523/jneurosci.23-17-06810.2003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The effect of the human immunodeficiency virus-1 protein Tat was investigated on neurotransmitter release from human and rat cortical nerve endings. Tat failed to affect the release of several neurotransmitters, such as glutamate, GABA, norepinephrine, and others, but it evoked the release of [3H]ACh via increase of cytosolic [Ca2+]. In human nerve terminals, the Tat effect partly depends on Ca2+ entry through voltage-sensitive Ca2+ channels, because Cd2+ halved the Tat-evoked release. Activation of group I metabotropic glutamate receptors (mGluR) and mobilization of Ca2+ from IP3-sensitive intraterminal stores are also involved, because the Tat effect was prevented by mGluR antagonists 2-methyl-6-(phenylethynyl)pyridine hydrochloride and 7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxylate ethyl ester and by the IP3 receptor antagonists heparin and xestospongin C. Furthermore, the group I selective mGlu agonist (RS)-3,5-dihydroxyphenylglycine enhanced [3H]ACh release. In rat nerve terminals, the Tat-evoked release neither depends on external Ca2+ ions entry nor on IP3-mediated mechanisms. Tat seems to cause mobilization of Ca2+ from ryanodine-sensitive internal stores because its effect was prevented by both 8-bromo-cyclic adenosine diphosphate-ribose and dantrolene. The Tat-evoked release from human synaptosomes was mimicked by the peptide sequences Tat 32-62, Tat 49-86, and Tat 41-60. In contrast, the Tat 49-86 and Tat 61-80 fragments, but not the Tat 32-62 fragment, were active in rat synaptosomes. In conclusion, Tat elicits Ca2+-dependent [3H]ACh release by species-specific intraterminal mechanisms by binding via discrete amino acid sequences to different receptive sites on human and rat cholinergic terminals.
Collapse
|
22
|
Brain-derived neurotrophic factor inhibits human immunodeficiency virus-1/gp120-mediated cerebellar granule cell death by preventing gp120 internalization. J Neurosci 2003. [PMID: 12843275 DOI: 10.1523/jneurosci.23-13-05715.2003] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The human immunodeficiency virus type 1 (HIV-1) envelope protein gp120 has been implicated in the pathogenesis of HIV-1 dementia. Thus, inhibition of gp120 activity could reduce HIV toxicity in the brain. We have used primary cultures of rat cerebellar granule cells to examine mechanisms whereby gp120 causes cell death and to characterize neuroprotective agents. gp120 induced a time- and concentration-dependent apoptotic cell death, which was caspase-3-mediated but caspase-1 independent, and was totally blocked by the irreversible caspase-3-like protease inhibitor N-acetyl-Asp-Glu-Val-Asp-chloromethylketone. Caspase-3 activation was observed only in neurons that internalize gp120, indicating that internalization is key to gp120 toxicity. Because brain-derived neurotrophic factor (BDNF) prevents caspase-3-mediated neuronal cell death, we examined whether BDNF could prevent gp120-mediated apoptosis. Preincubation of neurons with BDNF before the addition of gp120 reduced caspase-3 activation, and consequently rescued 80% of neurons from apoptosis. Most importantly, BDNF reduced the levels of CXC chemokine receptor-4 (CXCR4), a receptor that mediates HIV-1 gp120-induced apoptosis. This effect correlated with the ability of BDNF to reduce gp120 internalization and apoptosis. Moreover, BDNF blocked the neurotoxic effect of stromal-derived factor-1alpha, a natural ligand for CXCR4, further establishing a correlation between neuroprotection and downregulation of CXCR4. We propose that BDNF may be a valid therapy to slow down the progression of HIV/gp120-mediated neurotoxicity.
Collapse
|
23
|
Khan MZ, Brandimarti R, Musser BJ, Resue DM, Fatatis A, Meucci O. The chemokine receptor CXCR4 regulates cell-cycle proteins in neurons. J Neurovirol 2003; 9:300-14. [PMID: 12775414 PMCID: PMC2669737 DOI: 10.1080/13550280390201010] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Neurons express a variety of chemokine receptors that regulate neuronal signaling and survival, including CXCR4 and CCR5, the two major human immunodeficiency virus (HIV) coreceptors. However, the role of chemokine receptors in HIV neuropathology and neuroinflammatory disorders is still unclear. This study aims to determine whether chemokine receptors regulate the activity of cell-cycle proteins in neurons and evaluate the possibility that alterations of these proteins are involved in HIV neuropathogenesis. The authors studied the effect of the chemokine stromal cell-derived factor (SDF)-1alpha, the natural CXCR4 ligand, and an X4-using variant of gp120 on the activity of cell-cycle proteins involved in neuronal apoptosis and differentiation, such as Rb and E2F-1. Changes in expression, localization, and phosphorylation/activation of Rb and E2F-1 induced by SDF-1alpha (20 nM) gp120(IIIB) (200 pM) were analyzed in primary cultures of rat neurons and in a human cell line expressing recombinant CXCR4. The data indicate that changes in the nuclear and cytosolic levels of Rb--which result in the functional loss of this protein--are associated with apoptosis in hippocampal or cerebellar granule neurons and in cell lines. SDF-1alpha, which is able to rescue these neurons from apoptosis, induces a time-dependent increase of total Rb expression while decreasing the nuclear content of phosphorylated (Ser780/Ser795) Rb and the transcriptional activity of E2F-1. The HIV envelope protein gp120(IIIB) exerts opposite effects at the nuclear level. These data indicate that CXCR4 affects cell-cycle proteins in neurons and raise the possibility that chemokines may contribute to neuronal survival by repressing the activity of E2F-dependent apoptotic genes and maintaining neurons in a highly differentiated and quiescent state. This state may be altered during neuroinflammatory conditions and/or by HIV-derived proteins.
Collapse
Affiliation(s)
- Muhammad Zafrullah Khan
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Renato Brandimarti
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
- Department of Experimental Pathology, University of Bologna, Italy
| | - Brian Joseph Musser
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Danielle Marie Resue
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Alessandro Fatatis
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
24
|
van de Bovenkamp M, Nottet HSLM, Pereira CF. Interactions of human immunodeficiency virus-1 proteins with neurons: possible role in the development of human immunodeficiency virus-1-associated dementia. Eur J Clin Invest 2002; 32:619-27. [PMID: 12190962 DOI: 10.1046/j.1365-2362.2002.01029.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Human immunodeficiency virus-1 (HIV-1)-associated dementia is a severe neurological complication of HIV-1 infection that affects 15-20% of the patients in the late stages of acquired immunodeficiency syndrome. HIV-1-associated dementia is most probably a consequence of HIV-1 infection of the brain rather than of an opportunistic pathogen. The exact mechanism by which the virus causes this disorder, however, is not completely understood. A number of HIV-1 proteins have been shown to be released from HIV-1-infected cells and/or to be present in the extracellular milieu in the HIV-1-infected brain. Moreover, these proteins have been shown to possess neurotoxic and/or neuromodulatory features in vitro. This review describes the possible direct interactions of the HIV-1 proteins gp120, gp41, vpr, tat, rev, vpu and nef with neurons, which might play a role in the development of HIV-1-associated dementia in vivo.
Collapse
|
25
|
Corasaniti MT, Bilotta A, Strongoli MC, Navarra M, Bagetta G, Di Renzo G. HIV-1 coat protein gp120 stimulates interleukin-1beta secretion from human neuroblastoma cells: evidence for a role in the mechanism of cell death. Br J Pharmacol 2001; 134:1344-50. [PMID: 11704656 PMCID: PMC1573068 DOI: 10.1038/sj.bjp.0704382] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. The role of the pro-inflammatory cytokine interleukin-1beta (IL-1beta) in the mechanism of cell death induced by the human immunodeficiency virus type 1 (HIV-1) recombinant coat glycoprotein, gp120 IIIB, has been studied in the human CHP100 neuroblastoma cell line maintained in culture. 2. Death of neuroblastoma cells typically elicited by 10 pM gp120 or by human recombinant IL-1beta (10 ng x ml(-1)) has been minimized by the antagonist of IL-1 receptor, i.e. IL-1ra (0.5 and 50 ng x ml(-1), respectively), an endogenous molecule that antagonizes most of the biological actions of IL-1beta, or by an antibody (5 and 50 ng x ml(-1)) which blocks the human IL-1 receptor type I (IL-1RI). 3. ELISA experiments have established that gp120 enhances immunoreactive IL-1beta levels in the culture medium and this is prevented by exposure to the IL-1 converting enzyme (ICE) inhibitor t-butoxycarbonyl-L-aspartic acid benzyl ester-chloromethylketone [Boc-Asp(OBzl)-CMK] used at a concentration (2.5 microM) which significantly (P<0.001) reduces cell death. 4. Death of CHP100 cells induced by gp120 is also prevented by acetyl-Tyr-Val-Ala-Asp-chloromethylketone (Ac-YVAD-CMK; 10-100 microM), a second inhibitor of ICE, supporting the concept that the viral protein stimulates the conversion of the 31 kDa pro-IL-1beta in to the 17 kDa mature cytokine which is then secreted to cause death. 5. In conclusion, our present data demonstrate that gp120 stimulates the secretion of IL-1beta which then triggers CHP100 neuroblastoma cell death via stimulation of IL-1 receptor type I.
Collapse
Affiliation(s)
- M T Corasaniti
- Department of Pharmacobiological Sciences, University of Catanzaro Magna Graecia, Catanzaro, Italy.
| | | | | | | | | | | |
Collapse
|
26
|
Navarra M, Romano C, Lorenzon T, Rotiroti D, Di Renzo G. Ethanol exposure inhibits the cytotoxic effect induced by gp120 in CHP100 human neuroblastoma cells. J Neurosci Res 2001; 65:354-61. [PMID: 11494372 DOI: 10.1002/jnr.1161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The aim of present study was to investigate the acute effects of ethanol on cytotoxicity induced by HIV-1 coat protein gp120 in CHP100 human neuroblastoma cell line. We demonstrate that ethanol, within a range of clinically relevant concentrations (15-90 mM) prevents cell death elicited by gp120 (10 pM) in a dose dependent manner. This protective action seems to be mediated by a reduction of free intracellular Ca(2+) levels because ethanol, at concentrations ranging from 0.1-0.5%, is able to decrease gp120-stimulated Ca(2+) uptake up to 24%. Furthermore, our data show an involvement of NO/cGMP messenger system pathway, because ethanol is also able to reduce gp120-stimulated NO release (up to 45%) and cyclic GMP accumulation (up to 73%). These findings suggest that the protective effect of ethanol against gp120-induced cytotoxicity in CHP100 cells underlies a Ca(2+)-activated, NO/cGMP-dependent mechanism.
Collapse
Affiliation(s)
- M Navarra
- Department of Pharmacobiological Sciences, Faculty of Pharmacy, University of Catanzaro Magna Graecia & IBAF-CNR, 88021 Roccelletta di Borgia, Catanzaro, Italy.
| | | | | | | | | |
Collapse
|
27
|
Gosztonyi G, Ludwig H. Interactions of viral proteins with neurotransmitter receptors may protect or destroy neurons. Curr Top Microbiol Immunol 2001; 253:121-44. [PMID: 11417131 DOI: 10.1007/978-3-662-10356-2_6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- G Gosztonyi
- Abteilung für Neuropathologie, Freie Universität Berlin, Universitätsklinikum Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany.
| | | |
Collapse
|
28
|
Sanders VJ, Wiley CA, Hamilton RL. The mechanisms of neuronal damage in retroviral infections of the nervous system. Curr Top Microbiol Immunol 2001; 253:179-201. [PMID: 11417135 DOI: 10.1007/978-3-662-10356-2_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- V J Sanders
- Department of Neuroscience, University of California, San Diego, La Jolla, CA 92302, USA
| | | | | |
Collapse
|
29
|
Phillips TR, Billaud JN, Henriksen SJ. Methamphetamine and HIV-1: potential interactions and the use of the FIV/cat model. J Psychopharmacol 2001; 14:244-50. [PMID: 11106303 DOI: 10.1177/026988110001400309] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The interaction of methamphetamine with human immunodeficiency virus (HIV), the aetiologic agent of Acquired Immune Deficiency Syndrome (AIDS), has not been thoroughly investigated. However, increasingly, a larger proportion of HIV infected individuals acquire the virus through methamphetamine use or are exposed to this drug during their disease course. In certain populations, there is a convergence of methamphetamine use and HIV-1 infection; yet our understanding of the potential effects that simultaneous exposure to these two agents have on disease progression is extremely limited. Studying the interactions between methamphetamine and lentivirus in people is difficult. To thoroughly understand methamphetamine's effects on lentivirus disease progression, an animal model that is both clinically relevant and easily manipulated is essential. In this report, we identified potential problems with methamphetamine abuse in individuals with a concurrent HIV-1 infection, described the Feline Immunodeficiency Virus (FIV)/cat model for HIV-1, and reported our early findings using this modelling system to study the interaction of methamphetamine and lentivirus infections.
Collapse
Affiliation(s)
- T R Phillips
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
30
|
Huang MB, Bond VC. Involvement of protein kinase C in HIV-1 gp120-induced apoptosis in primary endothelium. J Acquir Immune Defic Syndr 2000; 25:375-89. [PMID: 11141237 DOI: 10.1097/00042560-200012150-00001] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We previously showed that HIV-1 gp120-induced apoptosis in primary human umbilical vein endothelial cell cultures (HUVEC), through CCR5 and CXCR4. Here, we have found that agonists of protein kinase C (PKC), basic fibroblast growth factor (bFGF), and short exposure to low concentrations of phorbol esters were found to block gp120-induced apoptosis in HUVEC cultures. PKC antagonists, sphingosine, H7, and extended exposure of cultures to high concentrations of phorbol esters were also found to block gp120-induced apoptosis in HUVEC cultures. A significant increase in the total amount of cellular PKC enzymatic activity was observed on exposure of HUVEC to gp120. No increase in total PKC activity was observed on exposure of HUVECs to the natural ligands SDF-1alpha, or regulated-on-activation normal T-expressed and secreted (RANTES) cells, and gp120-induced PKC induction was found to be totally blocked by CXCR4 antibodies and partially blocked by the caspase 3 inhibitor, DEVD-CHO. Alternatively, CXCR4 antibodies and DEVD-CHO totally blocked apoptosis. Finally, gp120-induced effects were found to be insensitive to pertussis toxin. Accumulated evidence suggests PKC involvement at multiple points in the gp120-induced apoptotic pathway; also suggests involvement of the CXCR4 receptor internalization pathway, and potentially suggests different downstream effects of gp120-receptor interactions and natural ligand-receptor interactions.
Collapse
Affiliation(s)
- M B Huang
- Department of Biochemistry, Morehouse School of Medicine, Atlanta, Georgia 30310-1495, USA
| | | |
Collapse
|
31
|
Involvement of Protein Kinase C in HIV-1 gp120-Induced Apoptosis in Primary Endothelium. J Acquir Immune Defic Syndr 2000. [DOI: 10.1097/00126334-200012150-00001] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Billaud JN, Ly C, Phillips TR, de la Torre JC. Borna disease virus persistence causes inhibition of glutamate uptake by feline primary cortical astrocytes. J Virol 2000; 74:10438-46. [PMID: 11044088 PMCID: PMC110918 DOI: 10.1128/jvi.74.22.10438-10446.2000] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2000] [Accepted: 08/23/2000] [Indexed: 11/20/2022] Open
Abstract
Borna disease virus (BDV), a nonsegmented, negative-stranded (NNS) RNA virus, causes central nervous system (CNS) disease in a broad range of vertebrate species, including felines. Both viral and host factors contribute to very diverse clinical and pathological manifestations associated with BDV infection. BDV persistence in the CNS can cause neurobehavioral and neurodevelopmental abnormalities in the absence of encephalitis. These BDV-induced CNS disturbances are associated with altered cytokine and neurotrophin expression, as well as cell damage that is very restricted to specific brain regions and neuronal subpopulations. BDV also targets astrocytes, resulting in the development of prominent astrocytosis. Astrocytes play essential roles in maintaining CNS homeostasis, and disruption of their normal activities can contribute to altered brain function. Therefore, we have examined the effect of BDV infection on the astrocyte's physiology. We present here evidence that BDV can establish a nonlytic chronic infection in primary cortical feline astrocytes that is associated with a severe impairment in the astrocytes' ability to uptake glutamate. In contrast, the astrocytes' ability to uptake glucose, as well as their protein synthesis, viability, and rate of proliferation, was not affected by BDV infection. These findings suggest that, in vivo, BDV could also affect an important astrocyte function required to prevent neuronal excitotoxicity. This, in turn, might contribute to the neuropathogenesis of BDV.
Collapse
Affiliation(s)
- J N Billaud
- Vaccine Research Institute of San Diego, San Diego, California 92121, USA
| | | | | | | |
Collapse
|
33
|
Abstract
This review examines the interaction of steroid hormones, glucocorticoids and estrogen, and gp120, a possible causal agent of acquired immune deficiency syndrome-related dementia complex. The first part of the review examines the data and mechanisms by which gp120 may cause neurotoxicity and by which these steroid hormones effect cell death in general. The second part of the review summarizes recent experiments that show how these steroid hormones can modulate the toxic effects of gp120 and glucocorticoids exacerbating toxicity, and estrogen decreasing it. We then examine the limited in vivo and clinical data relating acquired immune deficiency syndrome-related dementia complex and steroid hormones and speculate on the possible clinical significance of these findings with respect to acquired immune deficiency syndrome-related dementia complex.
Collapse
Affiliation(s)
- S M Brooke
- Department of Biological Sciences, Stanford University, Stanford, California 94305, USA
| | | |
Collapse
|
34
|
Gemignani A, Paudice P, Pittaluga A, Raiteri M. The HIV-1 coat protein gp120 and some of its fragments potently activate native cerebral NMDA receptors mediating neuropeptide release. Eur J Neurosci 2000; 12:2839-46. [PMID: 10971626 DOI: 10.1046/j.1460-9568.2000.00172.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The objective of this study was to investigate the effects of the HIV-1 envelope protein gp120 and its peptide fragments on the function of N-methyl-D-aspartate (NMDA) receptors mediating release of cholecystokinin (CCK) and somatostatin (SRIF). These are nonconventional NMDA receptors recently found to be activated by glycine or D-serine 'only'. The release of cholecystokinin-like immunoreactivity (CCK-LI) and of somatostatin-like immunoreactivity (SRIF-LI) elicited by 12 mM K+ from superfused rat neocortex synaptosomes was potently increased by gp120, its cyclic V3 loop and the linear V3 sequence BRU-C-34-A, but not by RP-135 (a central portion of BRU-C-34-A). The EC50 values of gp120 were 0.02 nM (CCK-LI release) and 0.01 nM (SRIF-LI release). The releasing effect of gp120 was prevented by blocking the glycine site or the ion channel of NMDA receptors, but not the glutamate recognition site; in addition, the gp120 effect was strongly inhibited by nanomolar concentrations of Zn2+ ions and by low micromolar concentrations of ifenprodil. It is concluded that gp120 acts as a very potent agonist at the glycine site of NMDA receptors sited on CCK- and SRIF-releasing nerve endings; the protein is able to activate the receptor channel in the absence of glutamate. Gp120 activates the receptors through its V3 loop as peptide fragments related to V3 retain near-maximal activity. The sensitivity of the gp120 effect to both Zn2+ and ifenprodil would not be incompatible with the idea that these NMDA receptors contain the triple subunit combination NR1/NR2A/NR2B.
Collapse
Affiliation(s)
- A Gemignani
- Department of Experimental Medicine, Pharmacology and Toxicology Section, Viale Cembrano 4, 16148 Genova, Italy
| | | | | | | |
Collapse
|
35
|
Müller WE, Laplanche JL, Ushijima H, Schröder HC. Novel approaches in diagnosis and therapy of Creutzfeldt-Jakob disease. Mech Ageing Dev 2000; 116:193-218. [PMID: 10996019 DOI: 10.1016/s0047-6374(00)00112-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The scrapie prion protein, PrP(Sc), as well as its peptide fragment, PrP106-126, are toxic on neuronal cells, resulting in cell death by an apoptotic, rather than necrotic mechanism. The apoptotic process of neuronal cells induced by prion protein supports diagnosis and offers potential targets for therapeutic intervention of the prion diseases. Among the cerebrospinal fluid (CSF) proteins, which may serve as markers of neuronal cell death associated with prion diseases, the 14-3-3 protein(s) turned out to be the most promising one. A new sensitive assay allows the detection of even small changes in the normally low levels of these proteins. In vitro, the toxic effects displayed by PrP(Sc) and its peptide fragment can be blocked by antagonists of N-methyl-D-aspartate (NMDA) receptor channels, like Memantine. Also Flupirtine, a non-opiod analgesic drug, which is already in clinical use, was found to display in vitro a strong cytoprotective effect on neurons treated with PrP(Sc) or PrP106-126. This drug acts like a NMDA receptor antagonists, but does not bind to the receptor. Clinical trials on prion diseases with Flupirtine are in progress. Flupirtine was found to enhance the intracellular levels of the antiapoptotic protein Bcl-2 and the antioxidative agent glutathione (GSH). Due to its favourable pharmacokinetic profile, Flupirtine is considered to be a promising drug to prevent neuronal death in Creutzfeldt-Jakob disease (CJD) and other neurodegenerative disorders occurring with age, e.g. Alzheimer's disease.
Collapse
Affiliation(s)
- W E Müller
- Institut für Physiologische Chemie, Abteilung Angewandte Molekularbiologie, Universität Mainz, Duesbergweg 6, D-55099, Mainz, Germany.
| | | | | | | |
Collapse
|
36
|
Bräuner-Osborne H, Egebjerg J, Nielsen EO, Madsen U, Krogsgaard-Larsen P. Ligands for glutamate receptors: design and therapeutic prospects. J Med Chem 2000; 43:2609-45. [PMID: 10893301 DOI: 10.1021/jm000007r] [Citation(s) in RCA: 435] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
MESH Headings
- Animals
- Drug Design
- Excitatory Amino Acid Agonists/chemistry
- Excitatory Amino Acid Agonists/metabolism
- Excitatory Amino Acid Agonists/pharmacology
- Excitatory Amino Acid Agonists/therapeutic use
- Excitatory Amino Acid Antagonists/chemistry
- Excitatory Amino Acid Antagonists/metabolism
- Excitatory Amino Acid Antagonists/pharmacology
- Excitatory Amino Acid Antagonists/therapeutic use
- Humans
- Ligands
- N-Methylaspartate/agonists
- N-Methylaspartate/antagonists & inhibitors
- N-Methylaspartate/chemistry
- N-Methylaspartate/metabolism
- Receptors, AMPA/agonists
- Receptors, AMPA/antagonists & inhibitors
- Receptors, AMPA/chemistry
- Receptors, AMPA/metabolism
- Receptors, Glutamate/chemistry
- Receptors, Glutamate/drug effects
- Receptors, Glutamate/metabolism
- Receptors, Kainic Acid/agonists
- Receptors, Kainic Acid/antagonists & inhibitors
- Receptors, Kainic Acid/chemistry
- Receptors, Kainic Acid/metabolism
- Receptors, Metabotropic Glutamate/agonists
- Receptors, Metabotropic Glutamate/antagonists & inhibitors
- Receptors, Metabotropic Glutamate/chemistry
- Receptors, Metabotropic Glutamate/metabolism
- Synapses/metabolism
Collapse
Affiliation(s)
- H Bräuner-Osborne
- NeuroScience PharmaBiotec Research Center, Department of Medicinal Chemistry, The Royal Danish School of Pharmacy, DK-2100 Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
37
|
Manion MK, Su Z, Villain M, Blalock JE. A new type of Ca
2+
channel blocker that targets Ca
2+
sensors and prevents Ca
2+
‐mediated apoptosis. FASEB J 2000. [DOI: 10.1096/fasebj.14.10.1297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Michael K. Manion
- University of Alabama at BirminghamDepartment of Physiology and Biophysics Birmingham Alabama 35294 USA
| | - Zhengchang Su
- University of Alabama at BirminghamDepartment of Physiology and Biophysics Birmingham Alabama 35294 USA
| | - Matteo Villain
- University of Alabama at BirminghamDepartment of Physiology and Biophysics Birmingham Alabama 35294 USA
| | - J. Edwin Blalock
- University of Alabama at BirminghamDepartment of Physiology and Biophysics Birmingham Alabama 35294 USA
| |
Collapse
|
38
|
Catani MV, Corasaniti MT, Navarra M, Nisticò G, Finazzi-Agrò A, Melino G. gp120 induces cell death in human neuroblastoma cells through the CXCR4 and CCR5 chemokine receptors. J Neurochem 2000; 74:2373-9. [PMID: 10820198 DOI: 10.1046/j.1471-4159.2000.0742373.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To infect target cells, the human immunodeficiency virus (HIV) type I (HIV-1) must engage not only the well-known CD4 molecule, but it also requires one of several recently described coreceptors. In particular, the CXCR4 (LESTR/fusin) receptor allows fusion and entry of T-tropic strains of HIV, whereas CCR5 is the major coreceptor used by primary HIV-1 strains that infect macrophages and CD4(+) T-helper cells (M-tropic viruses). In addition, the alpha chemokine SDF1alpha and the beta chemokines MIP1alpha, MIP1beta, and RANTES, natural ligands of CXCR4 and CCR5, respectively, are potent soluble inhibitors of HIV infection by blocking the binding between the viral envelope glycoprotein gp120 and the coreceptors. Approximately two-thirds of individuals with acquired immunodeficiency syndrome (AIDS) show neurologic complications, which are referred to a syndrome called AIDS dementia complex or HIV-1-associated cognitive/motor complex. The HIV-1 coat glycoprotein gp120 has been proposed as the major etiologic agent for neuronal damage, mediating both direct and indirect effects on the CNS. Furthermore, recent findings showing the presence of chemokine receptors on the surface of different cell types resident in the CNS raise the possibility that the association of gp120 with these receptors may contribute to the pathogenesis of neurological dysfunction. Here, we address the possible role of alpha and beta chemokines in inhibiting gp120-mediated neurotoxicity using the human neuroblastoma CHP100 cell line as an experimental model. We have previously shown that, in CHP100 cells, picomolar concentrations of gp120 produce a significant increase in cell death, which seems to proceed through a Ca(2+) - and NMDA receptor-dependent cascade. In this study, we gained insight into the mechanism(s) of neurotoxicity elicited by the viral glycoprotein. We found that CHP100 cells constitutively express both CXCR4 and CCR5 receptors and that stimulation with phorbol 12-myristate 13-acetate down-regulates their expression, thus preventing gp120-induced cell death. Furthermore, all the natural ligands of these receptors exerted protective effects against gp120-mediated neuronal damage, although with different efficiencies. These findings, together with our previous reports, suggest that the neuronal injury observed in HIV-1 infection could be due to direct (or indirect) interactions between the viral protein gp120 and chemokine and/or NMDA receptors.
Collapse
Affiliation(s)
- M V Catani
- Biochemistry Laboratory, IDI-IRCCS, c/o Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | | | | | | | | |
Collapse
|
39
|
Yeh MW, Kaul M, Zheng J, Nottet HS, Thylin M, Gendelman HE, Lipton SA. Cytokine-stimulated, but not HIV-infected, human monocyte-derived macrophages produce neurotoxic levels of l -cysteine. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:4265-70. [PMID: 10754324 DOI: 10.4049/jimmunol.164.8.4265] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Approximately one-quarter of individuals with AIDS develop neuropathological symptoms that are attributable to infection of the brain with HIV. The cognitive manifestations have been termed HIV-associated dementia. The mechanisms underlying HIV-associated neuronal injury are incompletely understood, but various studies have confirmed the release of neurotoxins by macrophages/microglia infected with HIV-1 or stimulated by viral proteins, including the envelope glycoprotein gp120. In the present study, we investigated the possibility that l -cysteine, a neurotoxin acting at the N-methyl-d -aspartate subtype of glutamate receptor, could contribute to HIV-associated neuronal injury. Picomolar concentrations of gp120 were found to stimulate cysteine release from human monocyte-derived macrophages (hMDM) in amounts sufficient to injure cultured rat cerebrocortical neurons. TNF-alpha and IL-1beta, known to be increased in HIV-encephalitic brains, as well as a cellular product of cytokine stimulation, ceramide, were also shown to induce release of cysteine from hMDM in a dose-dependent manner. A TNF-alpha-neutralizing Ab and an IL-1betaR antagonist partially blocked gp120-induced cysteine release, suggesting that these cytokines may mediate the actions of gp120. Interestingly, hMDM infected with HIV-1 produced significantly less cysteine than uninfected cells following stimulation with TNF-alpha. Our findings imply that cysteine may play a role in the pathogenesis of neuronal injury in HIV-associated dementia due to its release from immune-activated macrophages but not virus-infected macrophages. Such uninfected cells comprise the vast majority of mononuclear phagocytes (macrophages and microglia) found in HIV-encephalitic brains.
Collapse
Affiliation(s)
- M W Yeh
- Cerebrovascular and Neuroscience Research Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Billaud JN, Selway D, Yu N, Phillips TR. Replication rate of feline immunodeficiency virus in astrocytes is envelope dependent: implications for glutamate uptake. Virology 2000; 266:180-8. [PMID: 10612672 DOI: 10.1006/viro.1999.0079] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Feline immunodeficiency virus (FIV) induces neurological abnormalities in domestic cats. Previously, we demonstrated that two disparate strains of FIV (FIV-34TF10 and FIV-PPR) varied greatly in the ability to replicate in feline cortical astrocytes. To investigate the impact of the env region on the replication efficiency of these strains, we constructed two env chimera viruses, FIV-34TF10-PPRenv and FIV-PPR-34TF10env, to infect feline cortical astrocytes in vitro. Although all of these viruses infected cortical astrocytes, the efficiency of replication depended on strain, and the env region played an essential role. The viruses containing the env of 34TF10, FIV-34TF10, and FIV-PPR-34TF10env had the greatest replication rate, whereas the viruses containing the env of PPR replicated at a lower level. Other viral regions had modulatory effects on the replication rate, with the FIV-PPR genome providing a slight replication advantage over the FIV-34TF10 genome. We also monitored the effects of these viruses on an important astrocyte function, glutamate uptake; all viruses significantly decreased this activity, but only the viruses containing the env of PPR significantly impaired glutamate uptake without altering the culture viability. These results may be particularly relevant in the context of lentivirus-induced central nervous system disease in which a selective breakdown of astroglial function may contribute to neurodegeneration.
Collapse
Affiliation(s)
- J N Billaud
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California, 92037, USA
| | | | | | | |
Collapse
|
41
|
Medina I, Ghose S, Ben-Ari Y. Mobilization of intracellular calcium stores participates in the rise of [Ca2+]i and the toxic actions of the HIV coat protein GP120. Eur J Neurosci 1999; 11:1167-78. [PMID: 10103113 DOI: 10.1046/j.1460-9568.1999.00550.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The HIV envelope glycoprotein, GP120, increases intracellular Ca2+ concentration and induces degeneration of human and animal neurons in culture. Using patch-clamp recordings and Ca2+ imaging techniques, we have now examined the contribution of intracellular stores of calcium in the effects of GP120. We report that in rat hippocampal neuronal cultures, GP120 induces a dramatic and persistent increase in [Ca2+]i which is prevented by drugs that either deplete (caffeine, carbachol, thapsigargin) or block (dantrolene) Ca2+ release from intracellular stores. In contrast, N-methyl-d-aspartate (NMDA) receptors or voltage-dependent calcium channels do not participate in these effects, as: (i) the increase in [Ca2+]i was not affected by NMDA receptor antagonists or calcium channel blockers; and (ii) and GP120 did not generate any current in whole-cell recording. Dantrolene, a ryanodine stores inhibitor, also prevented neuronal death induced by GP120. Our results show that the GP120-induced rise in [Ca2+]i originates from intracellular calcium stores, and suggest that intracellular stores of calcium may play a determinant role in the pathological actions of GP120.
Collapse
Affiliation(s)
- I Medina
- INSERM, Unit 29, Hôpital de Port-Royal, Paris, France
| | | | | |
Collapse
|
42
|
Abstract
Glutamic acid is the principal excitatory neurotransmitter in the mammalian central nervous system. Glutamic acid binds to a variety of excitatory amino acid receptors, which are ligand-gated ion channels. It is activation of these receptors that leads to depolarisation and neuronal excitation. In normal synaptic functioning, activation of excitatory amino acid receptors is transitory. However, if, for any reason, receptor activation becomes excessive or prolonged, the target neurones become damaged and eventually die. This process of neuronal death is called excitotoxicity and appears to involve sustained elevations of intracellular calcium levels. Impairment of neuronal energy metabolism may sensitise neurones to excitotoxic cell death. The principle of excitotoxicity has been well-established experimentally, both in in vitro systems and in vivo, following administration of excitatory amino acids into the nervous system. A role for excitotoxicity in the aetiology or progression of several human neurodegenerative diseases has been proposed, which has stimulated much research recently. This has led to the hope that compounds that interfere with glutamatergic neurotransmission may be of clinical benefit in treating such diseases. However, except in the case of a few very rare conditions, direct evidence for a pathogenic role for excitotoxicity in neurological disease is missing. Much attention has been directed at obtaining evidence for a role for excitotoxicity in the neurological sequelae of stroke, and there now seems to be little doubt that such a process is indeed a determining factor in the extent of the lesions observed. Several clinical trials have evaluated the potential of antiglutamate drugs to improve outcome following acute ischaemic stroke, but to date, the results of these have been disappointing. In amyotrophic lateral sclerosis, neurolathyrism, and human immunodeficiency virus dementia complex, several lines of circumstantial evidence suggest that excitotoxicity may contribute to the pathogenic process. An antiglutamate drug, riluzole, recently has been shown to provide some therapeutic benefit in the treatment of amyotrophic lateral sclerosis. Parkinson's disease and Huntington's disease are examples of neurodegenerative diseases where mitochondrial dysfunction may sensitise specific populations of neurones to excitotoxicity from synaptic glutamic acid. The first clinical trials aimed at providing neuroprotection with antiglutamate drugs are currently in progress for these two diseases.
Collapse
Affiliation(s)
- A Doble
- Neuroscience Dept. Rhŏne-Poulenc Rorer S.A., Antony, France
| |
Collapse
|
43
|
Budd SL. Mechanisms of neuronal damage in brain hypoxia/ischemia: focus on the role of mitochondrial calcium accumulation. Pharmacol Ther 1998; 80:203-29. [PMID: 9839772 DOI: 10.1016/s0163-7258(98)00029-1] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Following a hypoxic-ischemic insult, the collapse of ion gradients results in the inappropriate release of excitatory neurotransmitters. Although excitatory amino acids such as glutamate are the likely extracellular mediators of the ensuing neuronal cell death, the intracellular events occurring downstream of glutamate receptor activation are much less clear. The present review attempts to summarize how Ca2+ overload of neurons following a hypoxic-ischemic insult is neurotoxic. In particular, the interlocked relation between mitochondrial Ca2+ accumulation and subsequent neuronal cell death is examined.
Collapse
Affiliation(s)
- S L Budd
- Neurosciences Institute, Ninewells Medical School, University of Dundee, Scotland, UK
| |
Collapse
|
44
|
Pattarini R, Pittaluga A, Raiteri M. The human immunodeficiency virus-1 envelope protein gp120 binds through its V3 sequence to the glycine site of N-methyl-D-aspartate receptors mediating noradrenaline release in the hippocampus. Neuroscience 1998; 87:147-57. [PMID: 9722148 DOI: 10.1016/s0306-4522(98)00125-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Recent results show that the HIV-1 protein gp120 can enhance N-methyl-D-aspartate receptor-mediated release of noradrenaline from CNS nerve endings. We now investigate the mechanism of this action, including the structural determinants of the gp120 effect and the nature of its binding sites. The N-methyl-D-aspartate-evoked release of [3H]noradrenaline from rat hippocampal synaptosomes was potentiated similarly by gp120 and gp160; gp41 was ineffective. The regions of gp120 involved appear to be outside the CD4-binding domain of the protein, because gp120 retained its activity after pretreatment with N-carbomethoxycarbonyl-D-prolyl-D-phenylalanine, a compound known to inhibit binding of gp120 to CD4 receptors. Moreover, sequences of gp120 critical for binding to CD4 did not mimic the effect of gp120. Preincubation of synaptosomes with anti-galactocerebroside antibodies did not affect gp120 activity. The protein effect was retained by peptides mimicking its V3 sequence, including the cyclic V3 "universal peptide" and the linear V3 sequence BRU-C-34-A, but not RP-135 (a central portion of BRU-C-34-A). The block of the N-methyl-D-aspartate-induced [3H]noradrenaline release by 7-chlorokynurenate, an antagonist at the N-methyl-D-aspartate receptor glycine site, was competitively reversed by glycine, by V3 and by BRU-C-34-A. When added with N-methyl-D-aspartate, V3 was three to four orders of magnitude more potent than glycine (EC50 values: about 20 pM and 150 nM, respectively) in enhancing [3H]noradrenaline release. Gp120 did not release glycine or serine from synaptosomes, thus excluding indirect actions through these agents. To conclude, gp120 may act following recognition by its V3 sequence of a high-affinity site possibly coincident with the glycine site of N-methyl-D-aspartate receptors present on hippocampal terminals of noradrenergic neurons. Considering the importance of N-methyl-D-aspartate receptor activation and of noradrenaline in cognitive processes, the effects of gp120 and V3 described here may be relevant to the pathology of AIDS dementia.
Collapse
Affiliation(s)
- R Pattarini
- Department of Experimental Medicine, University of Genoa, Italy
| | | | | |
Collapse
|
45
|
Corasaniti MT, Bagetta G, Rotiroti D, Nisticò G. The HIV envelope protein gp120 in the nervous system: interactions with nitric oxide, interleukin-1beta and nerve growth factor signalling, with pathological implications in vivo and in vitro. Biochem Pharmacol 1998; 56:153-6. [PMID: 9698067 DOI: 10.1016/s0006-2952(98)00044-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The neuronal loss often described at post-mortem in the brain neocortex of patients suffering from AIDS has been proposed to be responsible for the development of the AIDS dementia complex. Neuroinvasive strains of the HIV virus infect macrophages, microglial cells, and multinucleated giant cells, but not neurones. Processing of the virus by cells of the myelomonocytic lineage yields viral products known to initiate a complex network of events that may lead to the death of neurones and to the development of AIDS-associated neurological syndrome. The HIV-1 coat protein gp120, in particular, has been proposed as a likely etiologic agent of the described neuronal loss because it causes the death of neurones in culture. More recently, it has been shown that brain cortical cell death caused in rats by intracerebroventricular injection of gp120 occurs via apoptosis. This observation broadens our knowledge of the pathophysiology of the reported neuronal cell loss and opens a new avenue of experimental research for the development of novel therapeutic strategies for the treatment of patients suffering from AIDS-associated neurological syndrome.
Collapse
|
46
|
|
47
|
Corasaniti MT, Navarra M, Nisticò S, Rotiroti D, Maccarrone M, Melino G, Finazzi-Agrò A. Requirement for membrane lipid peroxidation in HIV-1 gp120-induced neuroblastoma cell death. Biochem Biophys Res Commun 1998; 246:686-9. [PMID: 9618273 DOI: 10.1006/bbrc.1998.8687] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The HIV-1 envelope protein gp120 engenders Ca(2+)-mediated, excitotoxic damage of rodent neuronal and human neuroblastoma cells in culture. Here we report that human CHP100 neuroblastoma cells undergo early peroxidation of membrane lipids following a brief exposure to gp120. This effect is prevented by preincubating cell cultures with the 21-aminosteroid U-74389G, an inhibitor of membrane lipid peroxidation, which also rescues neuroblastoma cultures from gp120-induced cell death; conversely, no protection from cell death is observed when the 21-aminosteroid is added to neuroblastoma cultures after the induction of membrane lipid peroxidation by gp120. These data indicate that membrane lipid peroxidation has a causative role in the expression of cell death produced by the viral protein.
Collapse
|
48
|
Abstract
Mounting evidence suggests that cognitive dysfunction developing as a result of HIV-1 infection is mediated at least in part by generation of excitotoxins and free radicals in the brain. This syndrome is currently designated HIV-1-associated cognitive/motor complex, was originally termed the AIDS Dementia Complex, and for simplicity, is called AIDS dementia in this review. Recently, brains of patients with AIDS have been shown to manifest neuronal injury and apoptotic-like cell death. How can HIV-1 result in neuronal damage if neurons themselves are only rarely, if ever, infected by the virus? Experiments from several different laboratories have lent support to the existence of HIV- and immune-related toxins in a variety of in vitro and in vivo paradigms. In one recently defined pathway to neuronal injury, HIV-infected macrophages and microglia, or immune-activated macrophages and astrocytes (activated by the shed HIV-1 envelope protein, gp120, or other viral proteins and cytokines), appear to secrete excitants and neurotoxins. These substances may include arachidonic acid, platelet-activating factor, free radicals (NO. and O2.-), glutamate, quinolinate, cysteine, amines, and as yet unidentified factors emanating from stimulated macrophages and reactive astrocytes. A final common pathway for neuronal susceptibility is operative, similar to that observed in stroke and several neurodegenerative diseases. This mechanism involves excessive activation of N-methyl-D-aspartate (NMDA) receptor-operated channels, with resultant excessive influx of Ca2+ and the generation of free radicals, leading to neuronal damage. With the very recent development of clinically tolerated NMDA antagonists, there is hope for future pharmacological intervention.
Collapse
Affiliation(s)
- S A Lipton
- Cerebrovascular and NeuroScience Research Institute, Brigham & Women's Hospital, Boston, Massachusetts 02115, USA.
| |
Collapse
|
49
|
Yu N, Billaud JN, Phillips TR. Effects of feline immunodeficiency virus on astrocyte glutamate uptake: implications for lentivirus-induced central nervous system diseases. Proc Natl Acad Sci U S A 1998; 95:2624-9. [PMID: 9482937 PMCID: PMC19438 DOI: 10.1073/pnas.95.5.2624] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Feline immunodeficiency virus (FIV) is a lentivirus of domestic cats that causes a spectrum of diseases remarkably similar to AIDS in HIV-infected humans. As part of this spectrum, both HIV-1 and FIV induce neurologic disorders. Because astrocytes are essential in maintaining the homeostasis of the central nervous system, we analyzed FIV for the ability to infect feline astrocytes. Through immunocytochemistry and reverse transcriptase activity, it was demonstrated that two molecular clones of FIV (FIV-34TF10 and FIV-PPR) produce a chronic low level productive infection of feline astrocyte cultures. To investigate the consequences of this infection, selected astrocyte functions were examined. Infection with FIV-34TF10 significantly decreased the ability of astrocytes to scavenge extracellular glutamate (with a peak inhibition of 74%). The effects of the infection did not appear to be a result of toxicity but rather were more selective in nature because the glucose uptake function of the infected astrocyte cultures was not altered. Our data demonstrate that FIV productively infected, at a low level, feline astrocyte cultures, and as a consequence of this infection, an important astroglial function was altered. These findings suggest that a chronic low grade infection of astrocytes may impair the ability of these cells to maintain homeostasis of the central nervous system that, in turn, may contribute to a neurodegenerative disease process that is often associated with lentivirus infections.
Collapse
Affiliation(s)
- N Yu
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
50
|
Scorziello A, Florio T, Bajetto A, Schettini G. Intracellular signalling mediating HIV-1 gp120 neurotoxicity. Cell Signal 1998; 10:75-84. [PMID: 9481481 DOI: 10.1016/s0898-6568(97)00093-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
During the last few years several studies have been undertaken to characterise the role of gp120, the HIV-1 envelope glycoprotein, in the pathogenesis of neurological defects associated with AIDS. However, neurons did not appear to be the main target of the virus, since the widespread neuronal damage is not associated with a productive viral infection in neurons. The current opinion supports the hypothesis that an indirect mechanism exists to explain the neuronal cell death which occurs in patients infected by HIV-1. In particular, several reports suggest that gp120 may be the main candidate as mediator of the neurological deficits during HIV-1 infection and demonstrate that this molecule affects neuronal survival through a direct interaction with non-neuronal cell types such as monocytes, macrophages/microglia and astrocytes.
Collapse
Affiliation(s)
- A Scorziello
- Institute of Pharmacology, Medical School, University of Genova, Italy
| | | | | | | |
Collapse
|