Elsevier

Neurobiology of Disease

Volume 45, Issue 2, February 2012, Pages 657-670
Neurobiology of Disease

HIV-1 Tat neurotoxicity: A model of acute and chronic exposure, and neuroprotection by gene delivery of antioxidant enzymes

https://doi.org/10.1016/j.nbd.2011.10.005Get rights and content

Abstract

HIV-associated neurocognitive disorder (HAND) is an increasingly common, progressive disease characterized by neuronal loss and progressively deteriorating CNS function. HIV-1 gene products, particularly gp120 and Tat elicit reactive oxygen species (ROS) that lead to oxidant injury and cause neuron apoptosis. Understanding of, and developing therapies for, HAND requires accessible models of the disease. We have devised experimental approaches to studying the acute and chronic effects of Tat on the CNS. We studied acute exposure by injecting recombinant Tat protein into the caudate-putamen (CP). Ongoing Tat expression, which more closely mimics HIV-1 infection of the brain, was studied by delivering Tat-expression over time using an SV40-derived gene delivery vector, SV(Tat). Both acute and chronic Tat exposure induced lipid peroxidation and neuronal apoptosis. Finally, prior administration of recombinant SV40 vectors carrying antioxidant enzymes, copper/zinc superoxide dismutase (SOD1) or glutathione peroxidase (GPx1), protected from Tat-induced apoptosis and oxidative injury. Thus, injection of recombinant HIV-1 Tat and the expression vector, SV(Tat), into the rat CP cause respectively acute or ongoing apoptosis and oxidative stress in neurons and may represent useful animal models for studying the pathogenesis and, potentially, treatment of HIV-1 Tat-related damage.

Highlights

► HIV-1 gene product Tat elicits reactive oxygen species and cause neuron apoptosis. ► We studied acute exposure by injecting recombinant Tat into the caudate-putamen. ► Ongoing Tat-induced neurotoxicity was studied using a SV40-derived vector, SV(Tat). ► Acute and chronic Tat injection induced lipid peroxidation and neuronal apoptosis. ► Gene delivery of antioxidant enzymes protected from Tat-induced lesions.

Introduction

HIV-1 enters the Central Nervous System (CNS) soon after it enters the body. There, it is largely impervious to highly active anti-retroviral therapeutic drugs (HAART). As survival with chronic HIV-1 infection improves, the number of people harboring the virus in their CNS increases. The prevalence of HIV-associated neurocognitive disorder (HAND) therefore continues to rise, and less fulminant forms of HAND such as minor neurocognitive/motor disorder (MCMD) have become more common than their more fulminant predecessors. HAND remains a significant independent risk factor for AIDS mortality (Major et al., 2000, Mattson et al., 2005, McArthur et al., 2005, Nath and Sacktor, 2006, Ances and Ellis, 2007, Antinori et al., 2007).

The pathogenesis of HAND largely reflects the neurotoxicity of HIV-1 proteins. Neurons themselves are rarely infected by HIV-1, and neuronal damage is felt to be mainly indirect. HIV-1 infects resident microglia, periventricular macrophages and some astrocytes (Ranki et al., 1995), leading to increased production of cytokines and to release of HIV-1 proteins, the most likely neurotoxins, among which are the envelope (Env) proteins and Tat (Kaul et al., 2001, van de Bovenkamp et al., 2002). The HIV-1 trans-acting protein Tat, an essential protein for viral replication, is a key mediator of neurotoxicity. Brain areas that are particularly susceptible to Tat toxicity include the CA3 region and the dentate gyrus of the hippocampus and the striatum. Tat is internalized by neurons primarily through lipoprotein related protein receptor (LRP) and by activation of NMDA receptor (Eugenin et al., 2003). It also interacts with several cell membrane receptors, including integrins, VEGF receptor in endothelial cells and possibly CXCR4 (Ghezzi et al., 2000).

Tat can directly depolarize neuron membranes, independently of Na+ flux (Magnuson et al., 1995) and may potentiate glutamate- and NMDA-triggered calcium fluxes and neurotoxicity (Magnuson et al., 1995). It promotes excitotoxic neuron apoptosis (Bonavia et al., 2001, Haughey et al., 2001) by activating endoplasmic reticulum pathways to release intracellular calcium ([Ca2 +]i) (Norman et al., 2008). Consequent dysregulation of calcium homeostasis (Kruman et al., 1998, Nath et al., 2000, Bonavia et al., 2001) leads to mitochondrial calcium uptake, caspase activation and, finally, neuronal death. Tat also increases levels lipid peroxidation (Haughey et al., 2004) by generating the reactive oxygen species (ROS) superoxide (O2) and hydrogen peroxide (H2O2). It activates inducible nitric oxide synthase (iNOS) to produce nitric oxide (NO), which binds superoxide anion to form the highly reactive peroxynitrite (ONOO) (Bonfoco et al., 1995). The latter may attack lipids, proteins and DNA, enhancing oxidant-related injury.

Tat neurotoxicity has been reported in cultured cells, but fewer studies have demonstrated its neurotoxic properties in vivo (Jones et al., 1998, Bansal et al., 2000, Askenov et al., 2001, Askenov et al., 2003). Tat-induced protein oxidation is well documented (Askenov et al., 2001, Askenov et al., 2003) but little is known about its effects on lipid peroxidation. We recently demonstrated that Tat activates multiple signaling pathways. In one of these, Tat-induced superoxide is an intermediate, while the other utilizes peroxide as a signal transducer (Agrawal et al., 2007). We show here that directly injecting Tat into the caudate putamen (CP) can induce neuron apoptosis and lipid peroxidation, and that Tat can be transported retrograde from the CP to the Substantia Nigra. We also tested the consequences of protracted exposure to Tat, by expressing Tat over time in CNS cells. Finally, prior gene delivery of the antioxidant enzymes Cu/Zn superoxide dismutase (SOD1) or glutathione peroxidase (GPx1) into the CP before injecting Tat protected against Tat-induced injury.

Section snippets

Animals

Female Sprague–Dawley rats (300–350 g) were purchased from Charles River Laboratories (Wilmington, MA). Protocols for injecting and euthanizing animals were approved by the Thomas Jefferson University Institutional Animal Care and Use Committee (IACUC), and are consistent with Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC) standards. Because estrogens can influence inflammation in the brain and attenuate HIV-1 gp120- and Tat-induced oxidative stress, experiments

Injection of Tat into the CP elicits apoptosis

After injection of 10 ng Tat into the CP, TUNEL assays were performed to detect apoptosis between 1 h and 8 weeks. TUNEL-positive cells were counted in the whole CP, at least in 5 different sections for each animal. There were extremely rare TUNEL-positive cells when the CP was injected with saline or rat IgG (negative controls). Numbers of TUNEL-positive cells peaked 2 days after injection of Tat (Figs. 1A, B). TUNEL staining was localized to the nuclei (Fig. 1C).

Tat-induced apoptotic cells are mainly neurons

Sections from the CP obtained 2 

Discussion

Oxidative stress plays a role in the development of HAND, and other neurodegenerative diseases (Beal, 1995, Smith et al., 1995, Cao et al., 1998, Mollace et al., 2001, Turchan et al., 2003). Oxidative stress in HIV-1 dementia has been documented by analyses of brain tissue, including increased levels of lipid peroxidation product (i.e., HNE) and the presence of oxidized proteins. Membrane-associated oxidative stress correlates with HIV-1 dementia pathogenesis and cognitive impairment (Mattson

Conclusion

Injection of Tat and SV(Tat) into the rat CP causes respectively acute and ongoing apoptosis and oxidative stress in neurons. These approaches to Tat administration may therefore represent useful animal models for studying the pathogenesis and treatment of HIV-1 Tat-related damage. Gene delivery of antioxidant enzymes by recombinant SV40-derived vectors protects against Tat-induced oxidative stress and neuronal apoptosis.

References (89)

  • X. Liu et al.

    Human immunodeficiency virus type 1 (HIV-1) Tat induces nitric-oxide synthase in human astroglia

    J. Biol. Chem.

    (2002)
  • J.P. Louboutin et al.

    HIV-1 gp120 neurotoxicity proximally and at a distance from the point of exposure: protection by rSV40 delivery of antioxidant enzymes

    Neurobiol. Dis.

    (2009)
  • J.P. Louboutin et al.

    Blood–brain barrier abnormalities caused by exposure to HIV-1 gp120 — protection by gene delivery of antioxidant enzymes

    Neurobiol. Dis.

    (2010)
  • J.P. Louboutin et al.

    HIV-1 gp120-induced neuroinflammation: relationship to neuron loss and protection by rSV40-delivered antioxidant enzymes

    Exp. Neurol.

    (2010)
  • J.C. McArthur et al.

    Neurological complications of HIV infection

    Lancet Neurol.

    (2005)
  • H.J. McKee et al.

    Immune responses against SIV envelope glycoprotein, using recombinant SV40 as a vaccine delivery vector

    Vaccine

    (2002)
  • V. Mollace et al.

    Oxidative stress and neuroAIDS: triggers, modulators and novel antioxidants

    Trends Neurosci.

    (2001)
  • A. Nath et al.

    Transient exposure to HIV-1 Tat protein results in cytokine production in macrophages and astrocytes: A hit and run phenomenon

    J. Biol. Chem.

    (1999)
  • J.L. Ridet et al.

    Lentivirus-mediated expression of glutathione peroxidase: neuroprotection in murine models of Parkinson's disease

    Neurobiol. Dis.

    (2006)
  • K. Rouger et al.

    X-linked vacuolated myopathy: TNF-alpha and IFN-gamma expression in muscle fibers with MHC class I on sarcolemma

    Am. J. Pathol.

    (2001)
  • B.V. Sauter et al.

    A replication-deficient rSV40 mediates liver-directed gene transfer and a long-term amelioration of jaundice in gunn rats

    Gastroenterology

    (2000)
  • M.A. Smith et al.

    Radical ageing in Alzheimer's disease

    Trends Neurosci.

    (1995)
  • S. Theodore et al.

    Methamphetamine and human immunodeficiency virus protein Tat synergize to destroy dopaminergic terminals in the rat striatum

    Neuroscience

    (2006)
  • B.Y. Zhou et al.

    Astrocyte activation and dysfunction and neuron death by HIV-1 tat expression in astrocytes

    Mol. Cell. Neurosci.

    (2004)
  • L. Agrawal et al.

    Antioxidant enzyme gene delivery to protect from HIV-1 gp120-induced neuronal apoptosis

    Gene Ther.

    (2006)
  • B.M. Ances et al.

    Dementia and neurocognitive disorders due to HIV-1 infection

    Semin. Neurol.

    (2007)
  • J.K. Andersen

    Oxidative stress in neurodegeneration: cause or consequence?

    Nat. Med.

    (2004)
  • A. Antinori et al.

    Updated research nosology for HIV-associated neurocognitive disorders

    Neurology

    (2007)
  • M.Y. Askenov et al.

    Oxidative damage induced by the injection of HIV-1 Tat protein in the rat striatum

    Neurosci. Lett.

    (2001)
  • M.Y. Askenov et al.

    Temporal relationship between HIV-1 Tat-induced neuronal degeneration, OX-42 immunoreactivity, reactive astrocytosis, and protein oxidation in the rat striatum

    Brain Res.

    (2003)
  • N. Avgeropoulos et al.

    SCID mice with HIV encephalitis develop behavioral abnormalities

    J. Acquir. Immune Defic. Syndr.

    (1998)
  • A. Bachis et al.

    Axonal transport of human immunodeficiency virus type 1 envelope protein glycoprotein 120 is found in association with neuronal apoptosis

    J. Neurosci.

    (2006)
  • A. Banerjee et al.

    HIV proteins (gp120 and Tat) and methamphetamine in oxidative stress-induced damage in the brain: potential role of the thiol antioxidant N-acetylcysteine amide

    Free Rad. Biol. Med.

    (2011)
  • M.F. Beal

    Aging, energy, and oxidative stress in neurodegenerative diseases

    Ann. Neurol.

    (1995)
  • E. Bonfoco et al.

    Apoptosis and necrosis: two distinct events induced, respectively, by mild and intense insults with N-methyl-d-aspartate or nitric oxide/superoxide in cortical cell cultures

    Proc. Natl. Acad. Sci. U.S.A.

    (1995)
  • A.J. Bruce-Keller et al.

    4-Hydroxynonenal, a product of lipid peroxidation, damages cholinergic neurons and impairs visuospatial memory in rats

    J. Neuropathol. Exp. Neurol.

    (1998)
  • A.J. Bruce-Keller et al.

    Proinflammatory and pro-oxidant properties of Tat in a microglial cell line: attenuation by 17b-estradiol

    J. Neurochem.

    (2001)
  • A.J. Bruce-Keller et al.

    Morphine causes rapid increases in glial activation and neuronal injury in the striatum of inducible HIV-1 Tat transgenic mice

    Glia

    (2008)
  • W. Cao et al.

    Oxygen free radicals involvement in ischemia and reperfusion of the brain injury to brain

    Neurosci. Lett.

    (1998)
  • K. Conant et al.

    Induction of monocyte chemoattractant protein-1 in HIV-1 Tat-stimulated astrocytes and elevation in AIDS dementia

    Proc Nath Acad Sci USA

    (1998)
  • P. Cordelier et al.

    Inhibition of HIV-1 in the central nervous system by IFN-alpha2 delivered by an SV40 vector

    J. Interferon Cytokine Res.

    (2003)
  • R.G. Cutler et al.

    Dysregulation of sphingolipids and sterol metabolism by ApoE4 in HIV dementia

    Neurology

    (2004)
  • E.A. Eugenin et al.

    MCP-1 (CCL2) protects human neurons and astrocytes from NMDA or HIV-tat-induced apoptosis

    J. Neurochem.

    (2003)
  • N.J. Haughey et al.

    HIV-1 tat through phosphorylation of NMDA receptors potentiates glutamate excitotoxicity

    J. Neurochem.

    (2001)
  • Cited by (62)

    • HIV-associated neurotoxicity and cognitive decline: Therapeutic implications

      2022, Pharmacology and Therapeutics
      Citation Excerpt :

      As our knowledge has advanced, it is clear that the development of HAND coincides with an increase in Tat and Tat-related toxicity. Direct intrastriatal injection of Tat protein-coupled with an SV40 vector resulted in a continuous generation of Tat protein in the rat striatum (Agrawal, Louboutin, Reyes, Van Bockstaele, & Strayer, 2012). One concern with other direct injection methods of Tat protein was the uncertainty surrounding the amount of Tat necessary to elicit toxicity.

    • Nanomedicines encountering HIV dementia: A guiding star for neurotherapeutics

      2022, Journal of Drug Delivery Science and Technology
    View all citing articles on Scopus

    This work was supported by NIH grant MH70287.

    View full text