NeuroAIDS Banner
 
NeuroAids Vol. 2, Issue 11 (December 1999)          

The SIV Model of HIV-associated Dementia

Dianne M. Rausch1

1Center for Mental Health Research on AIDS, National Institute of Mental Health
6100 Executive Blvd., Room 6212, MSC 9623, Bethesda, Maryland 20892-9623; USA
E-mail: dr89b@nih.gov

Keywords: animal model, neuropathogenesis, HIV-associated dementia, SIV.


Q: What is the value of the SIV Model of HIV-associated dementia?
HIV-associated dementia (HAD) refers to a spectrum of progressive cognitive and motor abnormalities of the central nervous system (CNS) resulting from HIV infection (1). HAD eventually develops in about 15-30 percent of infected patients (2). While the advent of highly active antiretroviral therapy (HAART) has dramatically decreased the death rate from AIDS, its impact on HAD is less clear. Most of the medications in this combination therapy have poor penetration into the CNS (3). While it appears that, at least in some cases, treatment with HAART results in improvements in HAD and significant decreases in plasma viral load (4)(5), some evidence suggests that HAART is less effective for HAD than for other AIDS-defining diseases (6). The neuropathogenesis of HAD is not well understood, in part because it cannot be readily studied in humans. Yet insights about pathogenic mechanisms are crucial to improved drug targeting and prevention strategies.

The simian immunodeficiency virus (SIV) model of HAD is widely considered the preeminent animal model in which to study the neuropathogenesis of HAD. Rhesus monkeys infected with SIV become immunosuppressed and display neuropathological (7)(8) and behavioral (9) features resembling those in HIV-infected humans. SIV is genetically, antigenically and morphologically close to HIV (10). The principal targets of SIV infection, like those of HIV, are monocytes and lymphocytes (11). There is evidence of HIV infection of astrocytes (12). SIV and HIV also infect brain capillary endothelial cells in vitro (13)(14), and there is evidence of SIV infection of these cells in vivo (15), although the latter finding is more controversial. The major advantage of the SIV model is that it permits sacrifice of animals, and thus access to CNS tissue, at any point during disease progression (9). In contrast, human HIV-infected CNS tissue is only available post-mortem. Another advantage of the model is the elimination of treatment-related effects. The major difference between SIV- and HIV-infection is that the former is more rapid in its progression (16), yet this very feature makes the model advantageous for research. Although the principal drawback of using rhesus monkeys for research is their expense, the SIV model has the potential to answer some key questions about HAD neuropathogenesis presented below.

Q: What are the mechanisms of HIV trafficking into the central nervous system?
It is generally agreed that microglia/macrophages (MG/MP) are the sites of productive infection within the CNS, but the mechanisms of viral entry into the CNS are still debated (17)(18). One of the foremost mechanisms of entry is thought to be through trafficking of HIV-infected lymphocytes or monocytes, the so-called Trojan Horse hypothesis, although several other mechanisms are possible (19).

Studies with the SIV model have shown that neuroinvasion across the blood brain barrier (BBB) is dependent on host factors and viral strain (20). Neuroinvasive strains yield a higher number of perivascular macrophages (derived from monocytes) in the CNS (21). The infiltrating cells in encephalitic vs. non-encephalitic brains of SIV-infected macaques display increased expression of several chemokines and corresponding chemokine receptors (22). Since chemokines serve as potent chemoattractants, this suggests that normal monocyte trafficking into the CNS is enhanced in HAD through upregulation of chemokines and their receptors. The roles of specific cell types and the mechanisms through which upregulation occur have been the subject of theoretical models (22), as well as in vitro studies. With an artificial BBB using co-cultures of endothelial cells and astrocytes, the astrocyte-derived chemokine, monocyte-chemoattractant protein-1 (MCP-1), appears to play a vital role in causing enhanced migration of leukocytes across the BBB (23). This process, which appears to be under the influence of the viral protein tat (24), may also include upregulated expression of adhesion molecules on endothelial cells (25). A major advantage of the SIV model is the ability to determine the in vivo relevance of in vitro findings about mechanisms of viral entry.

Q: What are the functional consequences of HIV infection in the central nervous system?
According to the prevailing model of HAD pathogenesis, neuronal dysfunction and death are the result of indirect mechanisms of neurotoxicity, namely excessive local concentrations of soluble neurotoxic factors released by infected MG/MP or astrocytes, or viral components (18)(26). The indirect model of HAD is the only way to account for neurotoxicity and death in the absence of productive HIV infection of neurons. Findings from the SIV model have substantiated the indirect model of HAD and have established early neurochemical markers of toxicity that correlate with the onset of behavioral impairment. These markers include quinolinic acid in macrophages, glial fibrillary acidic protein in astrocytes, and somatostatin in neurons (27). The SIV model also has uncovered an early electrophysiological marker—a delay in sensory evoked potentials—that precedes the onset of behavioral impairments (28). Identification of neurochemical or electrophysiological markers has tremendous utility for monitoring the efficacy of new strategies for treatment of HAD. These strategies are emerging from in vitro studies showing that prevention of neurotoxic damage can be achieved by blocking chemokine or NMDA receptors on neurons. Interaction with these receptors is the first step by which soluble factors from HIV-infected cells mediate abnormalities in neuronal signaling and apoptosis (29)(30).

Q: Is there a reservoir of HIV-infected cells in the CNS and what is its physiological significance?
Peripheral tissues contain viral reservoirs of latently infected, resting T-cells that are resistant to HAART (31). If activated, these reservoirs present serious consequences for controlling HIV. In the CNS, the existence of a reservoir of HIV-infected MG/MP is difficult to verify but widely assumed because HIV penetrates into the CNS soon after infection yet symptoms may not become manifest until years later (18). A CNS reservoir is considered even more refractory to treatment than are peripheral reservoirs because of poor treatment penetration across the BBB. A CNS reservoir may eventually account for an increase in HAD incidence among individuals now living longer (3). Moreover, evidence from the SIV model also suggests that a CNS reservoir may be responsible for reemergence of systemic infection via trafficking of infected cells from the CNS into the peripheral circulation (20). The possibility of a CNS reservoir contributing to peripheral disease has been overlooked, and thus represents a critical area of inquiry. Much needs to be learned about viral clearance (14) and the trafficking of distinct immune cells types into and out of the CNS with normal physiology and HIV infection (32). The SIV model is beneficial for studying the roles of a CNS reservoir and the types of cells infected through the ability to control viral type and time of inoculation and to evaluate the impact via bodily fluid sampling, peripheral tissue biopsy, behavioral assessment, and CNS tissue pathology.


References


Use the Back button in your browser to continue reading the article.

(1) Navia BA, Jordan BD, Price RW (1986). The AIDS dementia complex: I. Clinical features. Ann Neurol 19(6):517-524. Medline

(2) McArthur JC, Hoover DR, Bacellar H, Miller EN, Cohen BA, Becker JT, Graham NMH, McArthur JH, Selnes OA, Jacobson LP, Visscher BR, Concha M, Saah A (1993). Dementia in AIDS patients: incidence and risk factors. Neurology 43:2245-2252. Medline

(3) Lipton SA (1998). Neuronal injury associated with HIV-1: approaches to treatment. Annu Rev Pharmacol Toxicol 38:159-77. Medline

(4) Tozzi V, Balestra P, Galgani S, Narciso P, Sebastian, G, D'Amato C, Affticano C, Pigorini F, Pau FM, De Felici A, Benedetto A (1999). Positive and sustained effects of highly active antiretroviral therapy on HIV-1 associated neurocognitive impairment. AIDS 13(14):1889-1997. Medline

(5) Tozzi V, Balestra P, Galgani S, Narciso P, Sebastiani G, D'Amato C, Affticano C, Pigorini F, Pau FM, De Felici A, Benedetto A (1999). Positive and sustained effects of highly active antiretroviral therapy on HIV-1 associated neurocognitive impairment. AIDS 13(14):1889-1997. Medline

(6) Dore GJ, Correll PK, Li Y, Kaldor JM, Cooper DA, Brew BJ (1999). Changes to AIDS dementia complex in the era of highly active antiretroviral therapy. AIDS 13(10):1249-53. Medline

(7) Sharer LR (1992). Pathology of HIV-1 infection of the central nervous system. J Neuropathol Exp Neurol 51(1):3-11. Medline

(8) Lackner AA, Dandekar S, Gardner MB (1991) Neurobiology of simian and feline immunodeficiency virus infections. Brain Pathol 1: 201-212. Medline

(9) Murray EA, Rausch DM, Lendvay J, Sharer LR, Eiden LE (1992). Cognitive and motor impairments associated with SIV infection in rhesus monkeys. Science 255: 1246-1249. Medline

(10) Desrosiers RC, Daniel MD, Li Y (1989). HIV-related lentiviruses of nonhuman primates. AIDS Res Hum Retroviruses 5 (5):465-73. Medline

(11) Fox HS, Gold LH, Henrikson SJ, Bloom FE (1997). Simian immunodeficiency virus: a model for neuroAIDS. Neurobiol Dis 4(3-4):265-274. Medline

(12) Tornatore C, Chandra R, Berger JR, Major EO (1994). HIV-1 infection of subcortical astrocytes in the pediatric central nervous system. Neurology 44(3 pt 1):481-487. Medline

(13) Strelow LI, Watry DD, Fox HS, Nelson JA (1998). Efficient infection of brain microvascular endothelial cells by an in vivo-selected neuroinvasive SIVmac variant. J Neurovirol 4(3):269-280. Medline

(14) Moses, AV, Stenglein, SG, Strussenberg, JG, Wehrly, K, Chesebro, B, Nelson, JA (1996). Sequences regulating tropism of human immunodeficiency virus type 1 for brain capillary endothelial cells map to a unique region on the viral genome. J Virol 70(6):3401-3406. Medline

(15) Zink MC, Spelman JP, Robinson RB, Clements J (1998). SIV infection of macaques--modeling the progression to AIDS dementia. J Neurovirol 4(3):249-59. Medline

(16) Westmoreland SV, Halpern E, Lackner AA (1998). Simian immunodeficiency virus encephalitis in rhesus macaques is associated with rapid disease progression. J Neurovirol 4(3):260-8. Medline

(17) Kolson DL, Lavi E, Gonzalez-Scarano F (1998). The effects of human immunodeficiency virus in the central nervous system. Adv Virus Res 50:1-47. Medline

(18) Wiley CA, Achim CL, Christopherson C, Kidane Y, Keok S, Masliah E, Mellors J, Radhakrishnan L, Wang G, Soontorniyomkij V (1999). HIV mediates a productive infection of the brain. AIDS 13(15):2055-2059. Medline

(19) Hickey W and Williams KC (1998). Neuroimmunologic mechanisms for disease in AIDS: the role of the astrocyte. In The Neurology of AIDS (HE Gendelman, SA Lipton, L Epstein, and S Swindells, eds.), Chapman and Hall, New York, N.Y. 61-72.

(20) Zink MC, Amedee AM, Mankowski JL, Craig L, Didier P, Carter DL, Munoz A, Murphey-Corb M, Clements JE (1997). Pathogenesis of SIV encephalitis. Selection and replication of neurovirulent SIV. Am J Pathol 151(3):793-803. Medline

(21) Lane JH, Sasseville VG, Smith MO, Vogel P, Pauley DR, Heyes MP, Lackner AA (1996). Neuroinvasion by simian immunodeficiency virus coincides with increased numbers of perivascular macrophages/microglia and intrathecal immune activation. J Neurovirol 2(6):423-32. Medline

(22) Sasseville VG, Lackner AA (1997 ) Neuropathogenesis of simian immunodeficiency virus infection in macaque monkeys. J Neurovirol 3(1):1-9. Medline

(23) Weiss JM, Downie SA, Lyman WD, Berman JW (1998). Astrocyte-derived monocyte-chemoattractant protein-1 directs the transmigration of leukocytes across a model of the human blood-brain barrier. J Immunol 161(12):6896-903. Medline

(24) Weiss JM, Nath A, Major EO, Berman JW (1999). HIV-1 Tat induces monocyte chemoattractant protein-1-mediated monocyte transmigration across a model of the human blood-brain barrier and up-regulates CCR5 expression on human monocytes. J Immunol 163(5):2953-9. Medline

(25) Dhawan S, Puri RK, Kumar A, Duplan H, Masson JM, Aggarwal BB (1997). Human immunodeficiency virus-1-tat protein induces the cell surface expression of endothelial leukocyte adhesion molecule-1, vascular cell adhesion molecule-1, and intercellular adhesion molecule-1 in human endothelial cells. Blood 90(4):1535-1544. Medline

(26) Epstein, LG, Gendelman, HE (1993). Human immunodeficiency virus type 1 infection of the nervous system: pathogenic mechanisms. Ann Neurol 33:429-436. Medline

(27) Rausch DM, Murray EA, Eiden LE (1999). The SIV-infected rhesus monkey model for HIV-associated dementia and implications for neurological diseases. J Leukoc Biol 65(4):466-74. Medline

(28) Horn TF, Huitron-Resendiz S, Weed MR, Henriksen SJ, Fox HS (1998). Early physiological abnormalities after simian immunodeficiency virus infection. Proc Natl Acad Sci U S A 95(25):15072-7. Medline

(29) Zheng J, Thylin MR, Ghorpade A, Xiong H, Persidsky Y, Cotter R, Niemann D, Che M, Zeng YC, Gelbard HA, Shepard RB, Swartz JM, Gendelman HE (1999). Intracellular CXCR4 signaling, neuronal apoptosis and neuropathogenic mechanisms of HIV-1-associated dementia. J Neuroimmunol 98(2):185-200. Medline

(30) Haughey NJ, Holden CP, Nath A, Geiger JD (1999). Involvement of inositol 1,4,5-trisphosphate-regulated stores of intracellular calcium in calcium dysregulation and neuron cell death caused by HIV-1 protein tat. J Neurochem 73(4):1363-74. Medline

(31) Chun TW, Fauci AS (1999). Latent reservoirs of HIV: obstacles to the eradication of virus. Proc Natl Acad Sci U S A 96(20):10958-61. Medline

(32) Hickey W (1999). Leukocyte traffic in the central nervous system: the participants and their roles. Semin Immunol 11(2):125-37. Medline

 
Top
NeuroAids is a project of Science OnLine
funded through a grant from the National Institute of Mental Health.
AAAS Logo Copyright ©1998 by AAAS Science Publications, Inc.