1
|
Tse JKY. Gut Microbiota, Nitric Oxide, and Microglia as Prerequisites for Neurodegenerative Disorders. ACS Chem Neurosci 2017. [PMID: 28640632 DOI: 10.1021/acschemneuro.7b00176] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Regulating fluctuating endogenous nitric oxide (NO) levels is necessary for proper physiological functions. Aberrant NO pathways are implicated in a number of neurological disorders, including Alzheimer's disease (AD) and Parkinson's disease. The mechanism of NO in oxidative and nitrosative stress with pathological consequences involves reactions with reactive oxygen species (e.g., superoxide) to form the highly reactive peroxynitrite, hydrogen peroxide, hypochloride ions and hydroxyl radical. NO levels are typically regulated by endogenous nitric oxide synthases (NOS), and inflammatory iNOS is implicated in the pathogenesis of neurodegenerative diseases, in which elevated NO mediates axonal degeneration and activates cyclooxygenases to provoke neuroinflammation. NO also instigates a down-regulated secretion of brain-derived neurotrophic factor, which is essential for neuronal survival, development and differentiation, synaptogenesis, and learning and memory. The gut-brain axis denotes communication between the enteric nervous system (ENS) of the GI tract and the central nervous system (CNS) of the brain, and the modes of communication include the vagus nerve, passive diffusion and carrier by oxyhemoglobin. Amyloid precursor protein that forms amyloid beta plaques in AD is normally expressed in the ENS by gut bacteria, but when amyloid beta accumulates, it compromises CNS functions. Escherichia coli and Salmonella enterica are among the many bacterial strains that express and secrete amyloid proteins and contribute to AD pathogenesis. Gut microbiota is essential for regulating microglia maturation and activation, and activated microglia secrete significant amounts of iNOS. Pharmacological interventions and lifestyle modifications to rectify aberrant NO signaling in AD include NOS inhibitors, NMDA receptor antagonists, potassium channel modulators, probiotics, diet, and exercise.
Collapse
Affiliation(s)
- Joyce K. Y. Tse
- University Research Facility in Chemical
and Environmental Analysis, and Department of Civil and Environmental
Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| |
Collapse
|
2
|
Odiase FE, Ogunrin OA, Ogunniyi AA. Memory Performance in HIV/AIDS - A Prospective Case Control Study. Can J Neurol Sci 2014; 34:154-9. [PMID: 17598591 DOI: 10.1017/s0317167100005977] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Background:Memory impairment, usually impaired retrieval of information, has been described in HIV/AIDS, especially among those with severe illness. Neuro-cognitive disturbances in HIV/AIDS have been linked to poor quality of life and medication adherence. This prospective, case-control study was designed to assess the verbal and non-verbal memory as well as the attention abilities of Nigerian Africans with HIV/AIDS and correlate their performances with their CD4+ T lymphocytes (CD4+) counts.Methods:A total of 288 randomly selected subjects, comprising 96 HIV-positive symptomatic patients, 96 HIV-positive asymptomatic patients and 96 HIV-negative controls, participated in the study. The subjects were age-, sex-, and level of education matched. The Recognition Memory Test and Choice Reaction Time tasks, components of the computer-assisted neuropsychological tests battery- the Iron Psychology ‘FePsy’ were used for cognitive assessments.Results:The mean memory scores of the HIV-positive asymptomatic subjects did not differ significantly from the controls (p>0.05) but the HIV-positive symptomatic subjects' scores were significantly lower than the controls (p<0.05). Both HIV-positive groups had psychomotor slowing and impaired attention (p<0.05). The HIV-positive subjects with CD4+ counts <200/μl and between 200 and 499/μl had significant memory impairment (p<0.001 and p<0.001 respectively) but there was no significant impairment among those with count ≥500/μl. Impaired ability for sustained attention was however present irrespective of the CD4+ level relative to controls (p<0.001).Conclusions:We concluded that there was no significant memory disturbance among HIV-positive asymptomatic subjects despite the presence of impaired attention and psychomotor slowing, and that the severity of immune suppression (as indicated by the CD4+ T lymphocytes count) is a strong determinant of cognitive decline in HIV/AIDS.
Collapse
Affiliation(s)
- Francis E Odiase
- Neurology Unit, Department of Medicine, University of Benin Teaching Hospital, Benin City, Nigeria
| | | | | |
Collapse
|
3
|
Fields J, Dumaop W, Langford TD, Rockenstein E, Masliah E. Role of neurotrophic factor alterations in the neurodegenerative process in HIV associated neurocognitive disorders. J Neuroimmune Pharmacol 2014; 9:102-16. [PMID: 24510686 PMCID: PMC3973421 DOI: 10.1007/s11481-013-9520-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 12/26/2013] [Indexed: 12/30/2022]
Abstract
Migration of HIV infected cells into the CNS is associated with a spectrum of neurological disorders, ranging from milder forms of HIV-associated neurocognitive disorders (HAND) to HIV-associated dementia (HAD). These neuro-psychiatric syndromes are related to the neurodegenerative pathology triggered by the release of HIV proteins and cytokine/chemokines from monocytes/macrophages into the CNS -a condition known as HIV encephalitis (HIVE). As a result of more effective combined anti-retroviral therapy patients with HIV are living longer and thus the frequency of HAND has increased considerably, resulting in an overlap between the neurodegenerative pathology associated with HIV and that related to aging. In fact, HIV infection is believed to hasten the aging process. The mechanisms through which HIV and aging lead to neurodegeneration include: abnormal calcium flux, excitotoxicity, signaling abnormalities, oxidative stress and autophagy defects. Moreover, recent studies have shown that defects in the processing and transport of neurotrophic factors such as fibroblast growth factors (FGFs), neural growth factor (NGF) and brain-derived growth factor (BDNF) might also play a role. Recent evidence implicates alterations in neurotrophins in the pathogenesis of neurodegeneration associated with HAND in the context of aging. Here, we report FGF overexpression curtails gp120-induced neurotoxicity in a double transgenic mouse model. Furthermore, our data show disparities in brain neurotrophic factor levels may be exacerbated in HIV patients over 50 years of age. In this review, we discuss the most recent findings on neurotrophins and HAND in the context of developing new therapies to combat HIV infection in the aging population.
Collapse
Affiliation(s)
- Jerel Fields
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | | | | | | | | |
Collapse
|
4
|
Winston A, Duncombe C, Li PCK, Gill JM, Kerr SJ, Puls RL, Taylor-Robinson SD, Emery S, Cooper DA. Two patterns of cerebral metabolite abnormalities are detected on proton magnetic resonance spectroscopy in HIV-infected subjects commencing antiretroviral therapy. Neuroradiology 2012; 54:1331-9. [PMID: 22772471 DOI: 10.1007/s00234-012-1061-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 06/20/2012] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Cerebral function impairment remains problematic in subjects with chronic human immunodeficiency virus (HIV) infection despite effective combination antiretroviral therapy (cART). Using cerebral proton magnetic resonance spectroscopy ((1)H MRS), we aimed to determine if abnormalities could be detected in neurologically asymptomatic HIV-infected subjects electively commencing cART. METHODS Therapy-naive, HIV-infected individuals and HIV-uninfected controls underwent (1)H MRS in several anatomical voxels including the mid-frontal grey matter (FGM) and right basal ganglia (RBG). Differences in cerebral metabolite ratios between groups and correlations between immune and virological status were assessed. RESULTS Forty-six subjects were recruited (26 HIV-infected and 20 control subjects). In the HIV-infected group, mean CD4+ count (SD, cells per microlitre) and plasma HIV RNA (SD, log10 copies per millilitre) were 192 (86) and 4.71 (0.64), respectively. Choline (Cho)/Creatine (Cr) and myoinositol (MI)/Cr ratios were significantly lower in the FGM in HIV-infected subjects compared to controls (0.67 (0.14) versus 0.88 (0.49), p = 0.036, and 0.94 (0.28) and 1.17 (0.26), p = 0.008, for Cho/Cr and MI/Cr, respectively) and Cho/Cr ratio associated with CD4+ lymphocyte count (p = 0.041). N-Acetyl-aspartate (NAA)/Cho ratio was significantly lower in the RBG in HIV-infected subjects compared to controls (2.27 (0.54) versus 2.63 (0.68), p = 0.002), and this was associated with greater plasma HIV RNA load (p = 0.014). CONCLUSIONS Two patterns of cerebral metabolite abnormalities were observed in HIV-infected subjects electively commencing cART. Greater inflammatory metabolite ratios (Cho/Cr and MI/Cr) associated with lower markers of peripheral immune markers (CD4+ lymphocyte count) in the FGM and lower neuronal metabolite ratios (NAA/Cho) associated with greater HIV viraemia in the RBG were present in HIV-infected subjects.
Collapse
Affiliation(s)
- Alan Winston
- St. Mary's Hospital, Imperial College London, Ground Floor, Clinical Trials, Winston Churchill Wing, Praed Street, London, W2 1NY, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Abstract
HIV enters the brain during the early stages of initial infection and can result in a complicated array of diverse neurological dysfunctions. While neuronal injury and loss are at the heart of neurological decline and HIV-associated neuropathology, HIV does not productively infect neurons and the effects of HIV on neurons may be described as largely indirect. Viral proteins released from infected cells in the CNS are a well-characterized source of neuronal toxicity. Likewise, host-derived inflammatory cytokines and chemokines released from infected and/or activated glial cells can damage neurons, as well. Newly identified host-virus interactions and the current state of our knowledge regarding HIV-associated neuronal toxicity will be addressed in this review. Aspects of HIV-associated neurotoxic mechanisms, patterns of neuronal damage, viral effects on neurotrophic signaling, clade variations and comorbid substance abuse will be discussed. Recent advances in our understanding of the impact of HIV infection of the CNS on neuronal dysfunction and cell death will also be highlighted.
Collapse
Affiliation(s)
- Jane Kovalevich
- Department of Neuroscience, Temple University School of Medicine, Education & Research Building, 3500 North Broad Street, Philadelphia, PA 19140-5104, USA
| | | |
Collapse
|
6
|
Bertin J, Barat C, Méthot S, Tremblay MJ. Interactions between prostaglandins, leukotrienes and HIV-1: possible implications for the central nervous system. Retrovirology 2012; 9:4. [PMID: 22236409 PMCID: PMC3268096 DOI: 10.1186/1742-4690-9-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 01/11/2012] [Indexed: 12/29/2022] Open
Abstract
In HIV-1-infected individuals, there is often discordance between viremia in peripheral blood and viral load found in the central nervous system (CNS). Although the viral burden is often lower in the CNS compartment than in the plasma, neuroinflammation is present in most infected individuals, albeit attenuated by the current combined antiretroviral therapy. The HIV-1-associated neurological complications are thought to result not only from direct viral replication, but also from the subsequent neuroinflammatory processes. The eicosanoids - prostanoids and leukotrienes - are known as potent inflammatory lipid mediators. They are often present in neuroinflammatory diseases, notably HIV-1 infection. Their exact modulatory role in HIV-1 infection is, however, still poorly understood, especially in the CNS compartment. Nonetheless, a handful of studies have provided evidence as to how these lipid mediators can modulate HIV-1 infection. This review summarizes findings indicating how eicosanoids may influence the progression of neuroAIDS.
Collapse
Affiliation(s)
- Jonathan Bertin
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec - CHUL, 2705 boul, Laurier, Québec (QC), Canada, G1V 4G2
| | | | | | | |
Collapse
|
7
|
The nerve growth factor reduces APOBEC3G synthesis and enhances HIV-1 transcription and replication in human primary macrophages. Blood 2011; 117:2944-52. [PMID: 21217078 DOI: 10.1182/blood-2010-05-287193] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Macrophages infected with HIV-1 sustain viral replication for long periods of time, functioning as viral reservoirs. Therefore, recognition of factors that maintain macrophage survival and influence HIV-1 replication is critical to understanding the mechanisms that regulate the HIV-1-replicative cycle. Because HIV-1-infected macrophages release the nerve growth factor (NGF), and NGF neutralization reduces viral production, we further analyzed how this molecule affects HIV-1 replication. In the present study, we show that NGF stimulates HIV-1 replication in primary macrophages by signaling through its high-affinity receptor Tropomyosin-related Kinase A (TrKA), and with the involvement of reticular calcium, protein kinase C, extracellular signal-regulated kinase, p38 kinase, and nuclear factor-κB. NGF-induced enhancement of HIV-1 replication occurred during the late events of the HIV-1-replicative cycle, with a concomitant increase in viral transcription and production. In addition, NGF reduced the synthesis of the cellular HIV-1 restriction factor APOBEC3G and also overrode its interferon-γ-induced up-regulation, allowing the production of a well-fitted virus. Because NGF-TrKA signaling is a crucial event for macrophage survival, it is possible that NGF-induced HIV-1 replication plays a role in the maintenance of HIV-1 reservoirs. Our study may contribute to the understanding of the immunopathogenesis of HIV-1 infection and provide insights about approaches aimed at limiting viral replication in HIV-1 reservoirs.
Collapse
|
8
|
Hazleton JE, Berman JW, Eugenin EA. Novel mechanisms of central nervous system damage in HIV infection. HIV AIDS-RESEARCH AND PALLIATIVE CARE 2010; 2:39-49. [PMID: 22096383 PMCID: PMC3218694 DOI: 10.2147/hiv.s9186] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Human immunodeficiency virus-1 infection of the central nervous system is an early event after primary infection, resulting in motor and cognitive defects in a significant number of individuals despite successful antiretroviral therapy. The pathology of the infected brain is characterized by enhanced leukocyte infiltration, microglial activation and nodules, aberrant expression of inflammatory factors, neuronal dysregulation and loss, and blood–brain barrier disruption. Months to years following the primary infection, these central nervous system insults result in a spectrum of motor and cognitive dysfunction, ranging from mild impairment to frank dementia. The mechanisms that mediate impairment are still not fully defined. In this review we discuss the cellular and molecular mechanisms that facilitate impairment and new data that implicate intercellular communication systems, gap junctions and tunneling nanotubes, as mediators of human immunodeficiency virus-1 toxicity and infection within the central nervous system. These data suggest potential targets for novel therapeutics.
Collapse
Affiliation(s)
- Joy E Hazleton
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | |
Collapse
|
9
|
Dopaminergic neurotoxicity of HIV-1 gp120: Reactive oxygen species as signaling intermediates. Brain Res 2010; 1306:116-30. [DOI: 10.1016/j.brainres.2009.09.113] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 09/25/2009] [Accepted: 09/29/2009] [Indexed: 11/19/2022]
|
10
|
Yadav A, Collman RG. CNS inflammation and macrophage/microglial biology associated with HIV-1 infection. J Neuroimmune Pharmacol 2009; 4:430-47. [PMID: 19768553 PMCID: PMC5935112 DOI: 10.1007/s11481-009-9174-2] [Citation(s) in RCA: 151] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Accepted: 09/03/2009] [Indexed: 10/20/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection of the central nervous system (CNS) can result in neurological dysfunction with devastating consequences in a significant proportion of individuals with acquired immune deficiency syndrome. HIV-1 does not infect neurons directly but induces damage indirectly through the accumulation of activated macrophage/microglia (M/M) cells, some of which are infected, that release neurotoxic mediators including both cellular activation products and viral proteins. One mechanism for the accumulation of activated M/M involves the development in infected individuals of an activated peripheral blood monocyte population that traffics through the blood-brain barrier, a process that also serves to carry virus into CNS and establish local infection. A second mechanism involves the release by infected and activated M/M in the CNS of chemotactic mediators that recruit additional monocytes from the periphery. These activated M/M, some of which are infected, release a number of cytokines and small molecule mediators as well as viral proteins that act on bystander cells and in turn activate them, thus amplifying the cascade. These viral proteins and cellular products have neurotoxic properties as well, both directly and through induction of astrocyte dysfunction, which ultimately lead to neuronal injury and death. In patients effectively treated with antiretroviral therapy, frank dementia is now uncommon and has been replaced by milder forms of neurocognitive impairment, with less frequent and more focal neuropathology. This review summarizes key findings that support the critical role and mechanisms of monocyte/macrophage activation and inflammation as a major component for HIV-1 encephalitis or HIV-1 associated dementia.
Collapse
Affiliation(s)
- Anjana Yadav
- Department of Medicine and Center for AIDS Research, University of Pennsylvania School of Medicine, 522 Johnson Pavilion, 36th & Hamilton Walk, Philadelphia, PA 19104, USA
| | | |
Collapse
|
11
|
Xing HQ, Hayakawa H, Izumo K, Kubota R, Gelpi E, Budka H, Izumo S. In vivo expression of proinflammatory cytokines in HIV encephalitis: an analysis of 11 autopsy cases. Neuropathology 2009; 29:433-42. [PMID: 19170891 DOI: 10.1111/j.1440-1789.2008.00996.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
As the pathogenesis of AIDS dementia complex (ADC), cytokines such as TNF-alpha and IL-1beta have been thought to have toxic effects on CNS cells and induce neuronal cell death. However, many of the discussions have been based on the studies done by in vitro experiments. There are only a few reports which demonstrate proinflammatory cytokines directly in vivo in HIV encephalitis (HIVE) brains, and roles of these cytokines with relation to HIV-1 infection are not yet clarified. In the present study, we examined 11 autopsy cases of HIVE using immunohistochemistry, and explored which cell types expressed these cytokines and whether expression of cytokines was related to viral infection. IL-1beta was detected in the frontal white matter of all 11 cases where microglial nodules were observed to varying degrees, whereas TNF-alpha was detected in seven cases. IL-1beta- or TNF-alpha-positive cells were almost restricted to CD68-positive macrophages/microglia and mild expression of these cytokines by astrocytes was observed in two cases with severe HIVE. IL-1beta was detected in some HIVp24-positive multinucleated giant cells. However, we could not detect TNF-alpha expression in the HIVp24-positive cells, which indicates that IL-1beta is induced by HIV-1 infection. In conclusion, a macrophage/microglia lineage is the main cell type to release cytokines in HIVE, and IL-1beta expression by HIV-1-infected cells may be one of the important factors for induction of HIVE. In addition, many non-infected macrophages/microglia as well as some astrocytes express IL-1beta and TNF-alpha, which might contribute to pathogenesis of ADC.
Collapse
Affiliation(s)
- Hui Qin Xing
- Division of Molecular Pathology, Center for Chronic Viral Diseases, Graduate School of Medical and Dental Sciences, Kagoshima University, Sakuragaoka, Kagoshima, Japan.
| | | | | | | | | | | | | |
Collapse
|
12
|
Calì C, Marchaland J, Regazzi R, Bezzi P. SDF 1-alpha (CXCL12) triggers glutamate exocytosis from astrocytes on a millisecond time scale: Imaging analysis at the single-vesicle level with TIRF microscopy. J Neuroimmunol 2008; 198:82-91. [DOI: 10.1016/j.jneuroim.2008.04.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Accepted: 04/10/2008] [Indexed: 12/01/2022]
|
13
|
BACHIS ALESSIA, MOCCHETTI ITALO. Brain-Derived Neurotrophic Factor Is Neuroprotective against Human Immunodeficiency Virus-1 Envelope Proteins. Ann N Y Acad Sci 2008. [DOI: 10.1111/j.1749-6632.2005.tb00032.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
14
|
Xing HQ, Moritoyo T, Mori K, Sugimoto C, Ono F, Izumo S. Expression of proinflammatory cytokines and its relationship with virus infection in the brain of macaques inoculated with macrophage-tropic simian immunodeficiency virus. Neuropathology 2008; 29:13-9. [PMID: 18507770 DOI: 10.1111/j.1440-1789.2008.00929.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The pathogenesis of acquired immunodeficiency syndrome dementia complex (ADC) is still poorly understood. Many studies suggest that proinflammatory cytokines such as IL-1beta and TNF-alpha released by microglia/macrophages or astrocytes play a role in CNS injury. A microscopic finding of a microglial nodule with multinucleated giant cells (MNGCs) is a histopathologic hallmark of ADC and named HIV encephalitis. However, in vivo expression of these cytokines in this microenvironment of HIV encephalitis is not yet clarified. One of the main reasons is complexities of brain pathology in patients who have died from terminal AIDS. In this study, we infected two macaques with macrophage-tropic Simian immunodeficiency virus SIV239env/MERT and examined expression of TNF-alpha and IL-1beta in inflammatory lesions with MNGCs and its relation to virus-infected cells using immunohistochemistry. One macaque showed typical inflammatory lesions with MNGCs in the frontal white matter. Small microglial nodules were also detected in the basal ganglia and the spinal cord. SIVenv positive cells were detected mainly in inflammatory lesions, and seemed to be microglia/macrophages and MNGCs based on their morphology. Expression of IL-1beta and TNF-alpha were detected in the inflammatory lesions with MNGCs, and these positive cells were found to be negative for SIVenv by double-labeling immunohistochemistry or immunohistochemistry of serial sections. There were a few TNF-alpha positive cells and almost no IL-1beta positive cells in the area other than inflammatory lesions. Another macaque showed scattered CD3+ cells and CD68+ cells in the perivascular regions of the white matter. SIVenv and TNF-alpha was demonstrated in a few perivascular macrophages. These findings indicate that virus-infected microglia/macrophages do not always express IL-1beta and TNF-alpha, which suggests an indirect role of HIV-1-infected cells in cytokine-mediated pathogenesis of ADC. Our macaque model for human ADC may be useful for better understanding of its pathogenesis.
Collapse
Affiliation(s)
- Hui Qin Xing
- Division of Molecular Pathology, Center for Chronic Viral Diseases, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.
| | | | | | | | | | | |
Collapse
|
15
|
Fernandes SP, Edwards TM, Ng KT, Robinson SR. HIV-1 protein gp120 rapidly impairs memory in chicks by interrupting the glutamate–glutamine cycle. Neurobiol Learn Mem 2007; 87:1-8. [PMID: 16714124 DOI: 10.1016/j.nlm.2006.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Revised: 03/27/2006] [Accepted: 03/28/2006] [Indexed: 10/24/2022]
Abstract
Learning and memory impairments are frequently observed in patients suffering from AIDS Dementia Complex (ADC). These effects have been linked to the presence of gp120, an HIV viral coat glycoprotein. The present study investigated the possibility that gp120 prevents the uptake of extracellular glutamate by astrocytes, leading to an interruption of the glutamate-glutamine cycle and a subsequent impairment of memory. Ten microliters of 10nM gp120 was bilaterally injected into the region of the intermediate medial mesopallium of day-old chicks at various times before, or after, training using a single-trial passive avoidance task. Gp120 was found to significantly impair memory retention when injected 10-40 min after training. Memory impairments were evident within 5 min of gp120 administration and remained evident 24h later. Further, the amnestic effect of gp120 could be overcome with glutamine or with precursors of glutamate synthesis, but only weakly by glutamate. These results support the conclusion that the amnestic effect of gp120 is due to an impaired uptake of glutamate by astrocytes and a subsequent interruption of glutamine supply to neurones. The data indicate that the glutamate-glutamine cycle may be a useful therapeutic target in the treatment of ADC.
Collapse
Affiliation(s)
- S P Fernandes
- School of Psychology, Psychiatry and Psychological Medicine, Monash University, 3800 Vic., Australia
| | | | | | | |
Collapse
|
16
|
Abstract
Human immunodeficiency virus-1 (HIV-1)-infected and immune-activated macrophages and microglia secrete neurotoxins. Two of these neurotoxins are the pro-inflammatory cytokines tumour necrosis factor-alpha (TNF-alpha) and interleukin-1beta (IL-1beta), which are thought to play a major role in inducing neuronal death. Both TNF-alpha and IL-1beta increase the permeability of the blood-brain barrier, through which subsequently HIV-infected monocytes can enter the brain. They both induce over-stimulation of the NMDA-receptor via several pathways, resulting in a lethal neuronal increase in Ca(2+) levels. Additionally, TNF-alpha co-operates with several other proinflammatory mediators to enhance their toxic effects. Although most research has focused on the neurotoxic effects of TNF-alpha and IL-1beta in HAD, there is also evidence that these cytokines can be neuroprotective. In this paper the effect of TNF-alpha and IL-1beta on neuronal life and death in HAD is discussed.
Collapse
Affiliation(s)
- N A C H Brabers
- Department of Virology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | | |
Collapse
|
17
|
Price TO, Uras F, Banks WA, Ercal N. A novel antioxidant N-acetylcysteine amide prevents gp120- and Tat-induced oxidative stress in brain endothelial cells. Exp Neurol 2006; 201:193-202. [PMID: 16750528 DOI: 10.1016/j.expneurol.2006.03.030] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2006] [Accepted: 03/23/2006] [Indexed: 02/07/2023]
Abstract
Free radical production and, consequently, oxidative stress play an important role in the pathogenesis of AIDS and cause damage to lipids, proteins, and DNA. In our previous study, the HIV-1 envelope glycoprotein (gp120) and transregulatory protein (Tat) of HIV-1 have been found to induce oxidative stress in an immortalized endothelial cell line from rat brain capillaries, RBE4 (in vitro model of the blood-brain barrier). Here, we have determined the effects of a novel antioxidant, N-acetylcysteine amide (NACA), on gp120- and Tat-induced oxidative stress. Various oxidative stress parameters, including reduced glutathione (GSH), oxidized glutathione (GSSG), catalase (CAT) activity, and glutathione reductase (GR) activity, as well as malondialdehyde (MDA) levels, were used as measures of oxidative stress. NACA significantly increased the levels of intracellular GSH, CAT, and GR and decreased the levels of MDA in RBE4 cells, showing that oxidatively challenged cells were protected. Gp120- and Tat-induced increases in intracellular reactive oxygen species (ROS) were observed by using the 2',7'-DCF assay; the ROS scavenger, NACA, blocked ROS generation. A well-known apoptosis indicator, caspase-3 activity, was measured and was also found to have been returned to its control levels by NACA. Treatment of RBE4 cells with gp120 and Tat caused an increase in toxicity, as measured by lactate dehydrogenase (LDH) and tetrazolium reduction (MTS) assays. HIV-1 protein-induced toxicity in these cells was blocked by treatment with NACA. These studies show that NACA reverses gp120- and Tat-induced oxidative stress in immortalized endothelial cells.
Collapse
Affiliation(s)
- Tulin Otamis Price
- Department of Biochemistry, Faculty of Pharmacy, Marmara University, Istanbul 81010, Turkey
| | | | | | | |
Collapse
|
18
|
Wallace DR, Dodson S, Nath A, Booze RM. Estrogen attenuates gp120- and tat1-72-induced oxidative stress and prevents loss of dopamine transporter function. Synapse 2006; 59:51-60. [PMID: 16237680 DOI: 10.1002/syn.20214] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Postmenopausal women who are infected with HIV are at risk for experiencing dementia and Parkinson's-like symptoms associated with low levels of estrogen. Neurotoxic damage leading to these symptoms may involve HIV-associated proteins gp120 and/or tat(1-72) (tat). Our hypothesis is that 17beta-Estradiol (E(2)) is an effective agent for protection against gp120/tat-induced damage associated with increased oxidative stress, with particular focus on peroxynitrite-induced oxidative stress. We used SK-N-SH cells and striatal synaptosomes from Sprague-Dawley rats as model systems to assess neuroprotection by E(2). Cells coincubated with SIN-1(3-morpholinosydnonimine) or tat and gp120, together or separately, significantly increased oxidative stress on the SK-N-SH cells, as indicated by the increase in the levels of dichlorofluorescein (DCFH) fluorescence. These data suggest that a component of tat and gp120 neurotoxicity may be due to increased oxidative stress. Coincubation with E(2) attenuated tat- and gp120-induced increase in fluorescence. Coincubation with progesterone had no effect on tat-induced fluorescence, whereas coincubation with the E(2) antagonist ICI 182,780 and E(2) completely prevented the effects observed with E(2) alone. Both gp120 and tat decreased [(3)H] dopamine uptake into striatal synaptosomes by decreasing the V(max) of the dopamine transporter (DAT). Pretreatment of synaptosomes with E(2) (100 nM) partially reversed this reduction. In conclusion, E(2) appears to be effective for preventing the oxidative stress and loss of DAT function associated with gp120/tat neurotoxicity.
Collapse
Affiliation(s)
- David R Wallace
- Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, 74107, USA.
| | | | | | | |
Collapse
|
19
|
Nosheny RL, Bachis A, Aden SA, De Bernardi MA, Mocchetti I. Intrastriatal administration of human immunodeficiency virus-1 glycoprotein 120 reduces glial cell-line derived neurotrophic factor levels and causes apoptosis in the substantia nigra. ACTA ACUST UNITED AC 2006; 66:1311-21. [PMID: 16967504 DOI: 10.1002/neu.20288] [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: 01/08/2023]
Abstract
Uninfected neurons of the substantia nigra (SN) degenerate in human immunodeficiency virus (HIV)-positive patients through an unknown etiology. The HIV envelope glycoprotein 120 (gp120) causes apoptotic neuronal cell death in the rodent striatum, but its primary neurotoxic mechanism is still under investigation. Previous studies have shown that gp120 causes neurotoxicity in the rat striatum by reducing brain-derived neurotrophic factor (BDNF). Because glial cell line-derived neurotrophic factor (GDNF) and BDNF are neurotrophic factors crucial for the survival of dopaminergic neurons of the SN, we investigated whether gp120 reduces GDNF and BDNF levels concomitantly to induce apoptosis. Rats received a microinjection of gp120 or vehicle into the striatum and were sacrificed at various time intervals. GDNF but not BDNF immunoreactivity was decreased in the SN by 4 days in gp120-treated rats. In these animals, a significant increase in the number of caspase-3- positive neurons, both tyrosine hydroxylase (TH)-positive and -negative, was observed. Analysis of TH immunoreactivity revealed fewer TH-positive neurons and fibers in a medial and lateral portion of cell group A9 of the SN, an area that projects to the striatum, suggesting that gp120 induces retrograde degeneration of nigrostriatal neurons. We propose that dysfunction of the nigrostriatal dopaminergic system associated with HIV may be caused by a reduction of neurotrophic factor expression by gp120.
Collapse
Affiliation(s)
- Rachel L Nosheny
- Department of Neuroscience, Georgetown University Medical Center, Washington DC 20057, USA
| | | | | | | | | |
Collapse
|
20
|
Geiger KD, Stoldt P, Schlote W, Derouiche A. Ezrin Immunoreactivity Reveals Specific Astrocyte Activation in Cerebral HIV. J Neuropathol Exp Neurol 2006; 65:87-96. [PMID: 16410752 DOI: 10.1097/01.jnen.0000195943.32786.39] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The actin-binding protein ezrin is associated with cellular shape changes, motility, tumor invasion, and lymphocyte activation. We have earlier shown that ezrin immunoreactivity (IR) is faintly present in normal astrocytes but increased in malignant human astrogliomas. We studied the role of ezrin in astrocyte activation, applying immunostaining on serial paraffin sections from human autopsied brain tissues (51 cases). Cerebral HIV infection was chosen as a model displaying consistent exemplary astrocyte activation. Semiquantitative ezrin-IR was compared with the common glial markers GFAP, ferritin, and HLA-DR in relation to clinical and morphologic criteria of HIV encephalopathy. In all cases with HIV infection, GFAP-, HLA-DR-, and ferritin-IR were elevated in comparison to normal brain tissues. In contrast, high ezrin-IR in HIV infection strictly correlated with additional HIV encephalopathy. HIV encephalopathy with particularly high ezrin-IR was correlated with neuronal apoptosis (TUNEL). Combined ezrin-IR and GFAP-IR thus reveals 2 distinct states of astrocytic activation. Normal ezrin-IR, when paralleled by upregulated GFAP, reflects astroglial activation not associated with neuronal apoptosis. High ezrin-IR indicates specific astrocyte stressors related to cellular damage within the central nervous system. Ezrin-IR might also provide a diagnostic tool for the classification of HIV encephalopathy.
Collapse
|
21
|
Nosheny RL, Mocchetti I, Bachis A. Brain-derived neurotrophic factor as a prototype neuroprotective factor against HIV-1-associated neuronal degeneration. Neurotox Res 2005; 8:187-98. [PMID: 16260395 DOI: 10.1007/bf03033829] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Patients with human immunodeficiency virus type 1 (HIV-1) infection develop a broad spectrum of motor impairments and cognitive deficits, which follow or parallel cellular loss and atrophy in their brains. The viral envelope glycoprotein 120 (gp120) has been suggested to be a causal agent of neuronal loss. Therefore, reducing gp120 neurotoxicity may prevent neuronal degeneration seen in these patients. Here, we describe in vitro and in vivo experimental evidence that gp120 toxicity can be reduced by brain-derived neurotrophic factor (BDNF), a naturally occurring peptide that has been shown to block neurotoxin and trauma-induced neuronal injury. Moreover, we review the survival promoting properties of BDNF and the issues concerning its delivery into the brain, in an attempt to explain the rationale for exploring BDNF as a prototype trophic factor for a therapy to reduce neuronal cell death in HIV-1 infected patients.
Collapse
Affiliation(s)
- R L Nosheny
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | | | | |
Collapse
|
22
|
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: 2.9] [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
|
23
|
Price TO, Ercal N, Nakaoke R, Banks WA. HIV-1 viral proteins gp120 and Tat induce oxidative stress in brain endothelial cells. Brain Res 2005; 1045:57-63. [PMID: 15910762 DOI: 10.1016/j.brainres.2005.03.031] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2004] [Revised: 03/09/2005] [Accepted: 03/15/2005] [Indexed: 11/28/2022]
Abstract
The blood-brain barrier (BBB) has an important role in the development of AIDS dementia. The HIV-1 envelope glycoprotein (gp120) and transregulatory protein (Tat) of HIV-1 are neurotoxic and cytotoxic and have been implicated in the development of HIV dementia. They are known to cause oxidative stress and are associated with disruption of the BBB. Here, we used an immortalized endothelial cell line from rat brain capillaries, RBE4, to determine whether gp120 and Tat can induce oxidative stress in an in vitro model of the BBB. RBE4 cells were exposed to gp120 or Tat and the levels of reduced glutathione (GSH), oxidized glutathione (GSSG), catalase (CAT) activity, glutathione peroxidase (GPx) activity, and glutathione reductase (GR) activity, and malondialdehyde (MDA) used as measures of oxidative stress. Both gp120 and Tat significantly decreased the levels of intracellular GSH, GPx, and GR and increased the levels of MDA in RBE4 cells, showing that the cells were oxidatively challenged. The ratio of GSH/GSSG, a widely accepted indicator of oxidative stress, was also significantly decreased. These studies show that both of these viral proteins can induce oxidative stress in immortalized BBB endothelial cells.
Collapse
Affiliation(s)
- Tulin Otamis Price
- Department of Biochemistry, Faculty of Pharmacy, Marmara University, Istanbul 81010, Turkey
| | | | | | | |
Collapse
|
24
|
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: 1.5] [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
|
25
|
Khan NA, Di Cello F, Nath A, Kim KS. Human immunodeficiency virus type 1 tat-mediated cytotoxicity of human brain microvascular endothelial cells. J Neurovirol 2004; 9:584-93. [PMID: 14602571 DOI: 10.1080/13550280390218760] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2022]
Abstract
Human immunodeficiency virus (HIV)-1 infection is often complicated with neurologic disorders, but the pathogenesis of HIV-1 encephalopathy is incompletely understood. Tat (HIV-1 transactivator protein) is released from HIV-1-infected cells and has been detected in the sera and cerebrospinal fluid of HIV-1-infected patients. Tat, along with increased inflammatory cytokines such as interferon-gamma (IFN-gamma), have been implicated in the pathogenesis of HIV-1-associated blood-brain barrier dysfunction. The present study examined the effects of Tat and IFN-gamma on human brain microvascular endothelial cells (HBMECs), which constitute the blood-brain barrier. Tat produced cytotoxicity of HBMECs, but required IFN-gamma. IFN-gamma treatment of HBMECs up-regulates vascular endothelial growth factor receptor-2 (VEGFR2/KDR), which is known to be the receptor for Tat. Tat activated KDR in the presence of IFN-gamma, and Tat-mediated cytopathic changes involve its interaction with KDR and phosphatidylinositol 3-kinase (PI3K). Further understanding and characterization of Tat-HBMEC interactions should help us understand HIV-1 neuropathogenesis and develop strategies to prevent HIV-1 encephalopathy.
Collapse
Affiliation(s)
- Naveed Ahmed Khan
- Division of Pediatric Infectious Diseases, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|
26
|
Corasaniti MT, Piccirilli S, Paoletti A, Nisticò R, Stringaro A, Malorni W, Finazzi-Agrò A, Bagetta G. Evidence that the HIV-1 coat protein gp120 causes neuronal apoptosis in the neocortex of rat via a mechanism involving CXCR4 chemokine receptor. Neurosci Lett 2001; 312:67-70. [PMID: 11595336 DOI: 10.1016/s0304-3940(01)02191-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The HIV-1 coat protein, gp120 (100 ng given intracerebroventricularly (i.c.v.) daily for seven consecutive days) causes DNA fragmentation in the brain neocortex of rat. In neocortical cells bearing ultrastructural features typical of apoptosis, electron microscopy revealed specific immunopositivity for neurofilament cytoskeletal proteins, suggesting the neuronal nature of dying cells. Neuronal apoptosis by gp120 implicates CXCR4 chemokine receptors; in fact, in rats receiving a single daily, non-neurotoxic, dose of SDF-1alpha (0.25 pmoles given i.c.v. for 7 days before gp120), the natural ligand of CXCR4 receptor, apoptosis was significantly hindered. The mechanism of SDF-1alpha protection involves inhibition of gp120-enhanced expression of IL-1beta, a cytokine implicated in the mechanisms of apoptosis induced by the viral protein in the neocortex of rat.
Collapse
Affiliation(s)
- M T Corasaniti
- Department of Pharmacobiological Sciences, University of Catanzaro Magna Graecia and IBAF-CNR, Catanzaro, Italy
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Corasaniti MT, Maccarrone M, Nistico R, Malorni W, Rotiroti D, Bagetta G. Exploitation of the HIV-1 coat glycoprotein, gp120, in neurodegenerative studies in vivo. J Neurochem 2001; 79:1-8. [PMID: 11595751 DOI: 10.1046/j.1471-4159.2001.00537.x] [Citation(s) in RCA: 22] [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
Neuronal loss has often been described at post-mortem in the brain neocortex of patients suffering from AIDS. Neuroinvasive strains of HIV infect macrophages, microglial cells and multinucleated giant cells, but not neurones. Processing of the virus by cells of the myelomonocytic lineage yields viral products that, in conjunction with potentially neurotoxic molecules generated by the host, might initiate a complex network of events which lead neurones to death. In particular, the HIV-1 coat glycoprotein, gp120, has been proposed as a likely aetiologic agent of the described neuronal loss because it causes death of neurones in culture. More recently, it has been shown that brain neocortical cell death is caused in rat by intracerebroventricular injection of a recombinant gp120 coat protein, and that this occurs via apoptosis. The latter observation broadens our knowledge in the pathophysiology of the reported neuronal cell loss and opens a new lane of experimental research for the development of novel therapeutic strategies to limit damage to the brain of patients suffering from HIV-associated dementia.
Collapse
Affiliation(s)
- M T Corasaniti
- Faculty of Pharmacy, University of Catanzaro Magna Graecia and IBAF-CNR, Catanzaro, Italy
| | | | | | | | | | | |
Collapse
|
28
|
Belmadani A, Zou JY, Schipma MJ, Neafsey EJ, Collins MA. Ethanol pre-exposure suppresses HIV-1 glycoprotein 120-induced neuronal degeneration by abrogating endogenous glutamate/Ca2+-mediated neurotoxicity. Neuroscience 2001; 104:769-81. [PMID: 11440808 DOI: 10.1016/s0306-4522(01)00139-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The neurotoxic mechanism of HIV-1 envelope glycoprotein 120 (gp120) involves glutamatergic (NMDA) receptor/Ca2+-dependent excitotoxicity, mediated in part via glia. Pro-inflammatory cytokines also may have roles. We have reported that pre-exposure of brain cultures to 'physiological' ethanol concentrations (20-30 mM) protects against neuronal damage from HIV-1 gp120, but not from the direct receptor agonist, NMDA. Using lactate dehydrogenase assays and propidium iodide staining of rat organotypic hippocampal-entorhinal cortical slice cultures we determined that ethanol's suppression of gp120 neurotoxicity required at least 4 days of pretreatment. The gp120-induced neurotoxicity was accompanied by interleukin-6 elevations that were not affected by the pretreatment. However, gp120 induced substantial, early increases in extracellular glutamate levels that were blocked by ethanol pretreatment, conceivably abrogating excitotoxicity. Consistent with abrogation of excitotoxic pathways, fura-2 imaging showed selective deficits in gp120-dependent intracellular Ca2+ responses in ethanol-pretreated slices. Gp120 is believed to increase glutamate levels by both stimulating release and inhibiting (re)uptake. Results with a labeled glutamate analog, D-[3H]aspartate, revealed that gp120's inhibition of glutamate uptake, rather than its stimulation of release, was abolished after ethanol. Further studies indicated that two converging effects of ethanol pretreatment may underlie the abolishment of gp120-mediated glutamate uptake inhibition: (a) blockade of gp120-induced release (ostensibly from glia) of arachidonic acid, an inhibitor of astroglial glutamate reuptake, and (b) modest proliferation and activation of astroglia upon gp120 stimulation--which are likely to augment glutamate transporters. Thus, as with gp120 itself, glia and glutamate/arachidonic acid regulation appear to be important targets for ethanol. Since moderate ethanol consumption is as common among HIV-infected individuals as in the general population, this newly recognized neuroprotective (and apparently anti-excitotoxic) effect of ethanol withdrawal in vitro could be important, but it requires further study before its significance, if any, is understood.
Collapse
Affiliation(s)
- A Belmadani
- Department of Cell Biology, Neurobiology and Anatomy, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA
| | | | | | | | | |
Collapse
|
29
|
Corasaniti MT, Turano P, Bilotta A, Malorni W, Stringaro AR, Nisticò R, Finazzi-Agró A, Bagetta G. Evidence that increases of mitochondrial immunoreactive IL-1beta by HIV-1 gp120 implicate in situ cleavage of pro-IL-1beta in the neocortex of rat. J Neurochem 2001; 78:611-8. [PMID: 11483664 DOI: 10.1046/j.1471-4159.2001.00441.x] [Citation(s) in RCA: 22] [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
Immunoelectron microscopy analysis of brain tissue sections and rat-specific sandwich ELISA allowed the localization of interleukin-1beta (IL-1beta) immunoreactivity in the mitochondria and cytosol of neocortical tissue preparations from the brain of naive, untreated, rats and rats receiving a single daily injection into one lateral cerebral ventricle (i.c.v.) of bovine serum albumin (BSA; 100 ng/day) for seven consecutive days. Interestingly, seven days i.c.v. treatment with the HIV-1 coat protein gp120 (100 ng/day) enhances IL-1beta immunoreactivity in the cellular fractions studied. Elevation of mitochondrial immunoreactive IL-1beta levels seems to originate from the conversion operated by the interleukin converting enzyme (ICE) of mitochondrial pro-IL-1beta; in fact, IL-1beta increases reported in the ELISA experiments were paralleled by a decrease of the mitochondrial pro-IL-1beta 31-kDa band in conjunction with enhanced expression of the p20 component of activated ICE. In conclusion, the present results demonstrate that gp120-enhanced neocortical expression of IL-1beta originates, at least in part, from in situ cleavage of mitochondrial pro-IL-1beta and suggest that this, together with the central role of the mitochondrion in the expression of programmed cell death, may be important for apoptosis induced by the viral coat protein in the brain of rats.
Collapse
Affiliation(s)
- M T Corasaniti
- Department of Pharmacological Sciences, Faculty of Pharmacy, Catanzaro, Italy
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Mollace V, Nottet HS, Clayette P, Turco MC, Muscoli C, Salvemini D, Perno CF. Oxidative stress and neuroAIDS: triggers, modulators and novel antioxidants. Trends Neurosci 2001; 24:411-6. [PMID: 11410272 DOI: 10.1016/s0166-2236(00)01819-1] [Citation(s) in RCA: 154] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neurological disorders represent one of the most common disturbances accompanying HIV infection. In the past few years, highly antiretroviral active therapy has significantly reduced the incidence of HIV-related diseases. However, neurological dysfunction in AIDS patients still remains an unresolved problem. Oxidative stress, which occurs in brain tissues of patients undergoing HIV infection and is implicated in cell death of both astroglia and neurones, has recently been suggested to play a role in the pathogenesis of neuroAIDS. Thus, a better understanding of the processes that trigger and modulate free radical formation in brain tissues of AIDS patients might help in a successful therapeutic approach to the neuropathogenesis of HIV infection.
Collapse
Affiliation(s)
- V Mollace
- Faculty of Pharmacy, University of Catanzaro 'Magna Graecia', Roccelletta di Borgia, Catanzaro, Italy.
| | | | | | | | | | | | | |
Collapse
|
31
|
Speth C, Stöckl G, Mohsenipour I, Würzner R, Stoiber H, Lass-Flörl C, Dierich MP. Human immunodeficiency virus type 1 induces expression of complement factors in human astrocytes. J Virol 2001; 75:2604-15. [PMID: 11222683 PMCID: PMC115884 DOI: 10.1128/jvi.75.6.2604-2516.2001] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Since the brain is separated from the blood immune system by a tight barrier, the brain-resident complement system may represent a central player in the immune defense of this compartment against human immunodeficiency virus (HIV). Chronic complement activation, however, may participate in HIV-associated neurodegeneration. Since the level of complement factors in the cerebrospinal fluid is known to be elevated in AIDS-associated neurological disorders, we evaluated the effect of HIV type 1 (HIV-1) on the complement synthesis of brain astrocytes. Incubation of different astrocytic cell lines and primary astrocytes with HIV-1 induced a marked upregulation of the expression of the complement factors C2 and C3. The synthesis of other secreted or membrane-bound complement proteins was not found to be altered. The enhancement of C3 production was measured both on the mRNA level and as secreted protein in the culture supernatants. HIV-1 laboratory strains as well as primary isolates were capable of inducing C3 production with varied effectiveness. The usage of viral coreceptors by HIV-1 was proved to be a prerequisite for the upregulation of C3 synthesis, which was modulated by the simultaneous addition of cytokines. The C3 protein which is secreted after incubation of the cells with HIV was shown to be biologically active as it can participate in the complement cascade.
Collapse
Affiliation(s)
- C Speth
- Ludwig Boltzmann Institute for AIDS Research and Institute for Hygiene and Social Medicine, University of Innsbruck, Innsbruck, Austria.
| | | | | | | | | | | | | |
Collapse
|
32
|
Costa A, Nappi RE, Polatti F, Poma A, Grossman AB, Nappi G. Stimulating effect of HIV-1 coat protein gp120 on corticotropin-releasing hormone and arginine vasopressin in the rat hypothalamus: involvement of nitric oxide. Exp Neurol 2000; 166:376-84. [PMID: 11085902 DOI: 10.1006/exnr.2000.7502] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Subjects with human immunodeficiency virus type 1 (HIV-1) infection display increased activity of the hypothalamo-pituitary-adrenal (HPA) axis, which may play a role in both HIV-related neurodegenerative processes and disease progression. It has been speculated that the HIV coat protein gp120 may be responsible for these changes, and previous experimental evidence in both transgenic and nontransgenic mice supports this view. We speculated that one of the effects of gp120 in the CNS is to act within the hypothalamus to affect both corticotropin-releasing hormone (CRH) and arginine vasopressin (AVP), the principal regulators of HPA axis. We therefore administered i.p. gp120 (100 ng/rat) or vehicle to male Wistar rats and then detected Fos protein (an index of neuronal activation), CRH, and AVP immunoreactivity in the cellular compartments of the hypothalamic paraventricular nucleus (PVN). In addition, we tested the direct effect of various concentrations of gp120 on the release of CRH and AVP from rat hypothalamic explants maintained in vitro. Any modulation of gp120 effects by nitric oxide (NO) pathways was also sought by coadministering i.p. to rats or adding to the hypothalamic preparations the NO synthase inhibitor N(G)-methyl-l-arginine (l-NMMA). Gp120 induced the expression of Fos protein in both the parvo- and the magnocellular PVN, which was significantly attenuated by l-NMMA 10(-6) nM/L (P < 0.001 vs gp120 alone). Double immunochemistry showed costaining for Fos protein and CRH or AVP in the PVN following gp120; the number of double-labeled CRH and AVP cells for Fos protein was markedly reduced (P < 0.001) by coadministration of l-NMMA 10(-6) nM/L. In the in vitro studies, addition of gp120 to the hypothalamic explants in the dose range of 10 pM-1 nM resulted in a clear stimulation of both CRH and AVP release (P < 0.05-0.001 compared to control); in the presence of l-NMMA at 10-fold higher concentrations the stimulatory effect of gp120 on the release of both peptides was completely lost. It would therefore appear that gp120 activates CRH and AVP-producing neurons in the hypothalamic PVN and stimulates the release of both peptides in vitro via NO-dependent mechanisms. These findings, in line with previous evidence, further suggest that the increased activity of the HPA axis associated with HIV infection may be of central origin, due to the effects of gp120 on hypothalamic CRH and AVP release.
Collapse
Affiliation(s)
- A Costa
- Laboratory of Neuroendocrinology, Institute of Neurology IRCCS C. Mondino, Italy
| | | | | | | | | | | |
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
|
Wang YS, White TD. The HIV glycoproteins gp41 and gp120 cause rapid excitation in rat cortical slices. Neurosci Lett 2000; 291:13-6. [PMID: 10962142 DOI: 10.1016/s0304-3940(00)01385-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Inflammation and associated excitotoxicity may play important roles in various neurodegenerative diseases including AIDS dementia. Here we show that exposure of rat parietal cortical slices to the HIV glycoproteins gp120 and gp41 triggered very rapid releases of the neurotransmitters glutamate and [(3)H]noradrenaline (NA), and of the neuromodulator adenosine. Gp41 was more effective than gp120 at releasing glutamate and [(3)H]NA, while both glycoproteins were equi-effective at releasing adenosine. The responses to gp120 and gp41 declined rapidly to basal levels following their removal. It seems possible that rapid, inappropriate excitation may occur in the immediate vicinity of HIV infections in the brain, possibly producing some of the transient neurological and psychiatric symptoms associated with AIDS dementia.
Collapse
Affiliation(s)
- Y S Wang
- Department of Pharmacology, Dalhousie University, Halifax, B3H 4H7, Nova Scotia, Canada
| | | |
Collapse
|
35
|
Gavriil ES, Cooney R, Weeks BS. Tat mediates apoptosis in vivo in the rat central nervous system. Biochem Biophys Res Commun 2000; 267:252-6. [PMID: 10623606 DOI: 10.1006/bbrc.1999.1894] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The HIV-1 transactivator of transcription, Tat, mediates apoptosis in neurons and may cause AIDS-associated neurologic disorders, which include dementia and loss of motor control. Here we investigate the ability of Tat to stimulate apoptosis in the rat central nervous system in vivo. Using the TUNEL assay, treatment of rat pheochromocytoma (PC12) cells with 1.0 microgram/ml of Tat for 24 h was found to stimulate apoptosis. Further, electrophoresis of DNA from Tat-treated PC12 cells demonstrated fragmentation. To investigate Tat mediated apoptosis in vivo, 20 microgram of Tat was infused into the striatum of Sprague-Dawley rats. Histochemical analysis (TUNEL) of the Tat-infused area demonstrated apoptosis. Further, these rats demonstrated postural deviation ipsilateral to the infusion. These data demonstrate that Tat stimulates apoptosis in vivo and causes neurologic dysfunction in the intact animal.
Collapse
Affiliation(s)
- E S Gavriil
- Department of Biology, Adelphi University, Garden City, New York, 11530, USA
| | | | | |
Collapse
|