Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

HIV-associated neurocognitive disorder — pathogenesis and prospects for treatment

A Corrigendum to this article was published on 15 April 2016

This article has been updated

Key Points

  • Despite entering the era of combination antiretroviral therapy (CART), HIV-associated neurocognitive disorder (HAND) remains prevalent; however, less severe forms of HAND now predominate, and the most severe form, HIV-associated dementia, is rare

  • In individuals treated with CART, the risk of HAND increases with age and in the presence of cardiovascular disease risk factors

  • Latent HIV can persist in the brain even when systemic virological control is achieved with CART, thereby hampering efforts to eradicate HIV

  • Animal models of CNS HIV infection — such as macaques infected with simian immunodeficiency virus — develop severe HAND, viral encephalitis and neuronal apoptosis, and are central to understanding the immunopathogenesis of HIV-induced CNS damage

  • A growing body of work indicates that mild HAND can be modelled in immunocompetent mice infected with chimeric HIV (a model known as EcoHIV), and in chronically HIV-infected immunodeficient mice reconstituted with human immune systems

  • To date, clinical trials of HAND therapies have been unsuccessful, but further trials for the treatment of HAND are forthcoming, including a trial of intranasal insulin

Abstract

In the past two decades, several advancements have improved the care of HIV-infected individuals. Most importantly, the development and deployment of combination antiretroviral therapy (CART) has resulted in a dramatic decline in the rate of deaths from AIDS, so that people living with HIV today have nearly normal life expectancies if treated with CART. The term HIV-associated neurocognitive disorder (HAND) has been used to describe the spectrum of neurocognitive dysfunction associated with HIV infection. HIV can enter the CNS during early stages of infection, and persistent CNS HIV infection and inflammation probably contribute to the development of HAND. The brain can subsequently serve as a sanctuary for ongoing HIV replication, even when systemic viral suppression has been achieved. HAND can remain in patients treated with CART, and its effects on survival, quality of life and everyday functioning make it an important unresolved issue. In this Review, we describe the epidemiology of HAND, the evolving concepts of its neuropathogenesis, novel insights from animal models, and new approaches to treatment. We also discuss how inflammation is sustained in chronic HIV infection. Moreover, we suggest that adjunctive therapies — treatments targeting CNS inflammation and other metabolic processes, including glutamate homeostasis, lipid and energy metabolism — are needed to reverse or improve HAND-related neurological dysfunction.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Timeline of advances in neuro-AIDS research.
Figure 2: More-effective therapies have reduced the severity of HIV-associated the severity of HIV-associated neurocognitive disorders.
Figure 3: Neuropathogenic mechanisms that contribute to HIV-associated neurocognitive disorders.

Similar content being viewed by others

Change history

  • 15 April 2016

    The initially published version of this article included an incorrect grant number. The correct number of the NIH grant for David J. Volsky is MH104145. This error has been corrected in the HTML and PDF versions of the article.

References

  1. Fauci, A. S. & Marston, H. D. Ending the HIV-AIDS pandemic — follow the science. N. Engl. J. Med. 373, 2197–2199 (2015).

    Article  PubMed  Google Scholar 

  2. Maschke, M. et al. Incidence and prevalence of neurological disorders associated with HIV since the introduction of highly active antiretroviral therapy (HAART). J. Neurol. Neurosurg. Psychiatry 69, 376–380 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. DHHS Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV-1 infected adults and adolescents. AIDSinfo [online], (2015).

  4. The INSIGHT START Study Group. Initiation of antiretroviral therapy in early asymptomatic HIV infection. N. Engl. J. Med. 373, 795–807 (2015).

  5. Antinori, A. et al. Updated research nosology for HIV-associated neurocognitive disorders. Neurology 69, 1789–1799 (2007). This article updated the research nosology for HIV-associated neurocognitive disorders.

    Article  CAS  PubMed  Google Scholar 

  6. Heaton, R. K. et al. Neuropsychological impairment in human immunodeficiency virus-infection: implications for employment. HNRC Group. HIV Neurobehavioral Research Center. Psychosom. Med. 56, 8–17 (1994).

    Article  CAS  PubMed  Google Scholar 

  7. Heaton, R. et al. HIV-associated neurocognitive disorders (HAND) persist in the era of potent antiretroviral therapy: The CHARTER Study. Neurology 75, 2087–2096 (2010). This study reported that HIV-associated neurocognitive disorder remains prevalent even in individuals treated with combination antiretroviral therapy.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tozzi, V. et al. Persistence of neuropsychologic deficits despite long-term highly active antiretroviral therapy in patients with HIV-related neurocognitive impairment: prevalence and risk factors. J. Acquir. Immune Defic. Syndr. 45, 174–182 (2007).

    Article  PubMed  Google Scholar 

  9. Fois, A. F. & Brew, B. J. The potential of the CNS as a reservoir for HIV-1 infection: implications for HIV eradication. Curr. HIV/AIDS Rep. 12, 299–303 (2015).

    Article  PubMed  Google Scholar 

  10. McArthur, J. C. & Brew, B. J. HIV-associated neurocognitive disorders: is there a hidden epidemic? AIDS 24, 1367–1370 (2010).

    Article  PubMed  Google Scholar 

  11. Grant, I. et al. Evidence for early central nervous system involvement in the acquired immunodeficiency syndrome (AIDS) and other human immunodeficiency virus (HIV) infections: studies with neuropsychologic testing and magnetic resonance imaging. Ann. Intern. Med. 107, 828–836 (1987).

    Article  CAS  PubMed  Google Scholar 

  12. Ellis, R. J. et al. Neurocognitive impairment is an independent risk factor for death in HIV infection. Arch. Neurol. 54, 416–424 (1997).

    Article  CAS  PubMed  Google Scholar 

  13. Becker, J. T. et al. Cohort profile: recruitment cohorts in the neuropsychological substudy of the Multicenter AIDS Cohort Study. Int. J. Epidemiol. 44, 1506–1516 (2015).

    Article  PubMed  Google Scholar 

  14. McArthur, J. C. et al. Dementia in AIDS patients: incidence and risk factors. Neurology 43, 2245–2252 (1993).

    Article  CAS  PubMed  Google Scholar 

  15. Sacktor, N. et al. Antiretroviral therapy improves cognitive impairment in HIV+ individuals in Sub-Saharan Africa. Neurology 67, 311–314 (2006).

    Article  CAS  PubMed  Google Scholar 

  16. Grant, I. et al. Asymptomatic HIV-associated neurocognitive impairment increases risk for symptomatic decline. Neurology 82, 2055–2062 (2014). This study revealed that asymptomatic neurocognitive impairment increases the risk of symptomatic HIV-associated neurocognitive disorder in the future.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ragin, A. B. et al. Brain alterations within the first 100 days of HIV infection. Ann. Clin. Transl. Neurol. 2, 12–21 (2015). This manuscript highlights neuroimaging and cerebrospinal fluid cytokine findings in primary HIV infection.

    Article  PubMed  Google Scholar 

  18. Vo, Q. T. et al. Neuropsychological test performance before and after HIV-1 seroconversion: the Multicenter AIDS Cohort Study. J. Neurovirol. 19, 24–31 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Janssen, R. S., Nwanyanwu, O. C., Selik, R. M. & Stehr-Green, J. K. Epidemiology of human immunodeficiency virus encephalopathy in the United States. Neurology 42, 1472–1476 (1992).

    Article  CAS  PubMed  Google Scholar 

  20. Heaton, R. et al. HIV-associated neurocognitive disorders before and during the era of combination antiretroviral therapy: differences in rates, nature, and predictors. J. Neurovirol. 17, 3–16 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Childs, E. et al. Plasma viral load and CD4 lymphocytes predict HIV-associated dementia and sensory neuropathy. Neurology 52, 607 (1999).

    Article  CAS  PubMed  Google Scholar 

  22. McArthur, J. C. et al. Relationship between human immunodeficiency virus — associated dementia and viral load in cerebrospinal fluid and brain. Ann. Neurol. 42, 689–698 (1997).

    Article  CAS  PubMed  Google Scholar 

  23. Molsberry, S. A. et al. Mixed membership trajectory models of cognitive impairment in the multicenter AIDS cohort study. AIDS 29, 713–721 (2015).

    Article  PubMed  Google Scholar 

  24. Ellis, R. J. et al. CD4 nadir is a predictor of HIV neurocognitive impairment in the era of combination antiretroviral therapy. AIDS 25, 1747–1751 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Sacktor, N. et al. Prevalence of HIV-associated neurocognitive disorders in the Multicenter AIDS Cohort Study. Neurology 86, 334–340 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wright, E. J. et al. Factors associated with neurocognitive test performance at baseline: a substudy of the INSIGHT Strategic Timing of AntiRetroviral Treatment (START) trial. HIV Med. 16 (Suppl. 1), 97–108 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Wright, E. J. et al. No difference between the effects of immediate versus delayed ART on neuropsychological test performance in HIV+ adults with CD4 counts above 500 cells/microliter: the Strategic Timing of Anti-Retroviral (START) Neurology Substudy. Presented at the 15th European AIDS Conference, Barcelona (2015).

  28. McCutchan, J. A. et al. Role of obesity, metabolic variables, and diabetes in HIV-associated neurocognitive disorder. Neurology 78, 485–492 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Becker, J. T. et al. Vascular risk factors, HIV serostatus, and cognitive dysfunction in gay and bisexual men. Neurology 73, 1292–1299 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Fabbiani, M. et al. Cardiovascular risk factors and carotid intima-media thickness are associated with lower cognitive performance in HIV-infected patients. HIV Med. 14, 136–144 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Fazeli, P. L. et al. Cognitive functioning in adults aging with HIV: a cross-sectional analysis of cognitive subtypes and influential factors. J. Clin. Res. HIV AIDS Prev. 1, 155–169 (2015).

    Google Scholar 

  32. Valcour, V. et al. Higher frequency of dementia in older HIV-1 individuals: the Hawaii Aging with HIV-1 Cohort. Neurology 63, 822–827 (2004). This study showed that older age more than doubles the risk of HIV-associated neurocognitive disorder.

    Article  CAS  PubMed  Google Scholar 

  33. Joska, J. A. et al. Characterization of HIV-associated neurocognitive disorders among individuals starting antiretroviral therapy in South Africa. AIDS Behav. 15, 1197–1203 (2011).

    Article  PubMed  Google Scholar 

  34. Joska, J. A. et al. Neuropsychological outcomes in adults commencing highly active anti-retroviral treatment in South Africa: a prospective study. BMC Infect. Dis. 12, 39 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Marquine, M. J. et al. The veterans aging cohort study index is associated with concurrent risk for neurocognitive impairment. J. Acquir. Immune Defic. Syndr. 65, 190–197 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Heaton, R. et al. Aging amplifies HIV neurocognitive impairment: the effects may be related to vascular and metabolic factors. J. Neurovirol. 18, S46 (2012).

    Google Scholar 

  37. Vivithanaporn, P. et al. Hepatitis C virus co-infection increases neurocognitive impairment severity and risk of death in treated HIV/AIDS. J. Neurol. Sci. 312, 45–51 (2012).

    Article  PubMed  Google Scholar 

  38. Clifford, D. B. et al. Absence of neurocognitive effect of hepatitis C infection in HIV-coinfected people. Neurology 84, 241–250 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Weber, E. et al. Substance use is a risk factor for neurocognitive deficits and neuropsychiatric distress in acute and early HIV infection. J. Neurovirol. 19, 65–74 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Lawler, K. et al. Neurocognitive impairment among HIV-positive individuals in Botswana: a pilot study. J. Int. AIDS Soc. 13, 15 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Kwasa, J. et al. Lessons learned developing a diagnostic tool for HIV-associated dementia feasible to implement in resource-limited settings: pilot testing in Kenya. PLoS ONE 7, e32898 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Royal, W. 3rd et al. Clinical features and preliminary studies of virological correlates of neurocognitive impairment among HIV-infected individuals in Nigeria. J. Neurovirol. 18, 191–199 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Akolo, C. et al. Neurocognitive impairment associated with predominantly early stage HIV infection in Abuja, Nigeria. J. Neurovirol. 20, 380–387 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Kanmogne, G. D. et al. HIV-associated neurocognitive disorders in Sub-Saharan Africa: a pilot study in Cameroon. BMC Neurol. 10, 60 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Kelly, C. M. et al. HIV associated neurocognitive disorders (HAND) in Malawian adults and effect on adherence to combination anti-retroviral therapy: a cross sectional study. PLoS ONE 9, e98962 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wong, M. H. et al. Frequency of and risk factors for HIV dementia in an HIV clinic in Sub-Saharan Africa. Neurology 68, 350–355 (2007). This study found that HIV-associated neurocognitive disorder is common in sub-Saharan Africa.

    Article  CAS  PubMed  Google Scholar 

  47. Robertson, K. et al. Improved neuropsychological and neurological functioning across three antiretroviral regimens in diverse resource-limited settings: AIDS Clinical Trials Group study a5199, the International Neurological Study. Clin. Infect. Dis. 55, 868–876 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Heaton, R. K. et al. Neurocognitive change in the era of HIV combination antiretroviral therapy: the longitudinal CHARTER study. Clin. Infect. Dis. 60, 473–480 (2015). This longitudinal study highlights the changing epidemiology of HIV-associated neurocognitive disorder.

    Article  CAS  PubMed  Google Scholar 

  49. Masters, M. C. & Ances, B. M. Role of neuroimaging in HIV-associated neurocognitive disorders. Semin. Neurol. 34, 89–102 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Holt, J. L., Kraft-Terry, S. D. & Chang, L. Neuroimaging studies of the aging HIV-1-infected brain. J. Neurovirol. 18, 291–302 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ances, B. M. & Hammoud, D. A. Neuroimaging of HIV-associated neurocognitive disorders (HAND). Curr. Opin. HIV AIDS 9, 545–551 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  52. McIntosh, R. C., Rosselli, M., Uddin, L. Q. & Antoni, M. Neuropathological sequelae of Human Immunodeficiency Virus and apathy: a review of neuropsychological and neuroimaging studies. Neurosci. Biobehav. Rev. 55, 147–164 (2015).

    Article  PubMed  Google Scholar 

  53. Ragin, A. B. et al. Structural brain alterations can be detected early in HIV infection. Neurology 79, 2328–2334 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kelly, S. G. et al. Early suppressive antiretroviral therapy in HIV infection is associated with measurable changes in the corpus callosum. J. Neurovirol. 20, 514–520 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Li, S. et al. Matrix metalloproteinase levels in early HIV infection and relation to in vivo brain status. J. Neurovirol. 19, 452–460 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Burdo, T. H. et al. Soluble CD163 made by monocyte/macrophages is a novel marker of HIV activity in early and chronic infection prior to and after anti-retroviral therapy. J. Infect. Dis. 204, 154–163 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sailasuta, N. et al. Change in brain magnetic resonance spectroscopy after treatment during acute HIV infection. 7, e49272 (2012).

  58. Kore, I. et al. Neuropsychological impairment in acute HIV and the effect of immediate antiretroviral therapy. J. Acquir. Immune Defic. Syndr. 70, 393–399 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Chang, L. et al. Persistent brain abnormalities in antiretroviral-naive HIV patients 3 months after HAART. Antiviral Ther. 8, 17–26 (2003).

    CAS  Google Scholar 

  60. Hestad, K. et al. Regional brain atrophy in HIV-1 infection: association with specific neuropsychological test performance. Acta Neurol. Scand. 88, 112–118 (1993).

    Article  CAS  PubMed  Google Scholar 

  61. Jernigan, T. L. et al. Clinical factors related to brain structure in HIV: the CHARTER study. J. Neurovirol. 17, 248–257 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Cohen, R. A. et al. Effects of nadir CD4 count and duration of human immunodeficiency virus infection on brain volumes in the highly active antiretroviral therapy era. J. Neurovirol. 16, 25–32 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Paul, R. H. et al. Relative sensitivity of magnetic resonance spectroscopy and quantitative magnetic resonance imaging to cognitive function among nondemented individuals infected with HIV. J. Int. Neuropsychol. Soc. 14, 725–733 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Paul, R., Cohen, R., Navia, B. & Tashima, K. Relationships between cognition and structural neuroimaging findings in adults with human immunodeficiency virus type-1. Neurosci. Biobehav. Rev. 26, 353–359 (2002).

    Article  PubMed  Google Scholar 

  65. Lyons, J. L. et al. Plasma sCD14 is a biomarker associated with impaired neurocognitive test performance in attention and learning domains in HIV infection. J. Acquir. Immune Defic. Syndr. 57, 371–379 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Burdo, T. H. et al. Elevated sCD163 in plasma but not cerebrospinal fluid is a marker of neurocognitive impairment in HIV infection. AIDS 27, 1387–1395 (2013).

    Article  CAS  PubMed  Google Scholar 

  67. Kamat, A. et al. Monocyte activation markers in cerebrospinal fluid associated with impaired neurocognitive testing in advanced HIV infection. J. Acquir. Immune Defic. Syndr. 60, 234 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Sacktor, N. et al. Impact of minocycline on cerebrospinal fluid markers of oxidative stress, neuronal injury, and inflammation in HIV-seropositive individuals with cognitive impairment. J. Neurovirol. 20, 620–626 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Yuan, L. et al. Cytokines in CSF correlate with HIV-associated neurocognitive disorders in the post-HAART era in China. J. Neurovirol. 19, 144–149 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Mohamed, M. A. et al. Brain metabolism and cognitive impairment in HIV infection: a 3-T magnetic resonance spectroscopy study. Magn. Reson. Imaging 28, 1251–1257 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Towgood, K. J. et al. Regional cerebral blood flow and FDG uptake in asymptomatic HIV-1 men. Hum. Brain Mapp. 34, 2484–2493 (2013).

    Article  PubMed  Google Scholar 

  72. Haughey, N. J. et al. Perturbation of sphingolipid metabolism and ceramide production in HIV-dementia. Ann. Neurol. 55, 257–267 (2004).

    Article  CAS  PubMed  Google Scholar 

  73. Dickens, A. M. et al. Cerebrospinal fluid metabolomics implicate bioenergetic adaptation as a neural mechanism regulating shifts in cognitive states of HIV-infected patients. AIDS 29, 559–569 (2015). This study showed that changes in bioenergetics are associated with cognitive performance.

    CAS  PubMed  Google Scholar 

  74. Cassol, E., Misra, V., Dutta, A., Morgello, S. & Gabuzda, D. Cerebrospinal fluid metabolomics reveals altered waste clearance and accelerated aging in HIV patients with neurocognitive impairment. AIDS 28, 1579–1591 (2014).

    Article  CAS  PubMed  Google Scholar 

  75. Bandaru, V. V. R. et al. A lipid storage-like disorder contributes to cognitive decline in HIV-infected subjects. Neurology 81, 1492–1499 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Bandaru, V. V. et al. Associative and predictive biomarkers of dementia in HIV-1-infected patients. Neurology 68, 1481–1487 (2007).

    Article  CAS  PubMed  Google Scholar 

  77. Ernst, T., Jiang, C. S., Nakama, H., Buchthal, S. & Chang, L. Lower brain glutamate is associated with cognitive deficits in HIV patients: a new mechanism for HIV-associated neurocognitive disorder. J. Magn. Reson. Imaging 32, 1045–1053 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Gongvatana, A. et al. Progressive cerebral injury in the setting of chronic HIV infection and antiretroviral therapy. J. Neurovirol. 19, 209–218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Chang, L. et al. A multicenter in vivo proton-MRS study of HIV-associated dementia and its relationship to age. NeuroImage 23, 1336–1347 (2004).

    Article  CAS  PubMed  Google Scholar 

  80. Mielke, M. M., Bandaru, V. V., McArthur, J. C., Chu, M. & Haughey, N. J. Disturbance in cerebral spinal fluid sphingolipid content is associated with memory impairment in subjects infected with the human immunodeficiency virus. J. Neurovirol. 16, 445–456 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Suh, J. et al. Progressive increase in central nervous system immune activation in untreated primary HIV-1 infection. J. Neuroinflammation 11, 199 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. McGuire, J. L., Gill, A. J., Douglas, S. D. & Kolson, D. L. Central and peripheral markers of neurodegeneration and monocyte activation in HIV-associated neurocognitive disorders. J. Neurovirol. 21, 439–448 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Cassol, E. et al. Plasma metabolomics identifies lipid abnormalities linked to markers of inflammation, microbial translocation, and hepatic function in HIV patients receiving protease inhibitors. BMC Infect. Dis. 13, 203 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Archibald, S. et al. Brain morphometric correlates of metabolic variables in HIV: the CHARTER study. J. Neurovirol. 20, 603–611 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Seider, T. R. et al. Age exacerbates HIV-associated white matter abnormalities. J. Neurovirol. http://dx.doi.org/10.1007/s13365-015-0386-3 (2015).

  86. Chang, L. et al. Effects of APOE ε4, age, and HIV on glial metabolites and cognitive deficits. Neurology 82, 2213–2222 (2014). This article links glial dysfunction and changes seen on magnetic resonance spectroscopy to HIV-associated cognitive impairment and age.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Canizares, S., Cherner, M. & Ellis, R. J. HIV and aging: effects on the central nervous system. Semin. Neurol. 34, 27–34 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  88. Pfefferbaum, A. et al. Accelerated aging of selective brain structures in human immunodeficiency virus infection: a controlled, longitudinal magnetic resonance imaging study. Neurobiol. Aging 35, 1755–1768 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Thomas, J. B., Brier, M. R., Snyder, A. Z., Vaida, F. F. & Ances, B. M. Pathways to neurodegeneration: effects of HIV and aging on resting-state functional connectivity. Neurology 80, 1186–1193 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  90. Peluso, M. J. et al. Absence of cerebrospinal fluid signs of neuronal injury before and after immediate antiretroviral therapy in acute HIV infection. J. Infect. Dis. 212, 1759–1767 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  91. Peterson, J. et al. Cerebrospinal fluid (CSF) neuronal biomarkers across the spectrum of HIV infection: hierarchy of injury and detection. PLoS ONE 9, e116081 (2014). This study demonstrated change in markers of neuronal injury as HIV-associated neurocognitive disorderprogresses.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Abdulle, S. et al. CSF neurofilament protein (NFL) — a marker of active HIV-related neurodegeneration. J. Neurol. 254, 1026–1032 (2007).

    Article  CAS  PubMed  Google Scholar 

  93. Angel, T. E. et al. The cerebrospinal fluid proteome in HIV infection: change associated with disease severity. Clin. Proteomics 9, 3 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Jessen Krut, J. et al. Biomarker evidence of axonal injury in neuroasymptomatic HIV-1 patients. PLoS ONE 9, e88591 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Peluso, M. J. et al. Cerebrospinal fluid and neuroimaging biomarker abnormalities suggest early neurological injury in a subset of individuals during primary HIV infection. J. Infect. Dis. 207, 1703–1712 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Marcotte, T. D. et al. A concise panel of biomarkers identifies neurocognitive functioning changes in HIV-infected individuals. J. Neuroimmune Pharmacol. 8, 1123–1135 (2013).

    Article  PubMed  Google Scholar 

  97. Vago, L. et al. Pathological findings in the central nervous system of AIDS patients on assumed antiretroviral therapeutic regimens: retrospective study of 1597 autopsies. AIDS 16, 1925–1928 (2002). This pathological patient series study highlights the impact of combination antiretroviral therapy on reducing the frequency of the pathological effects of HIV infection.

    Article  CAS  PubMed  Google Scholar 

  98. Gelman, B. B. Neuropathology of HAND with suppressive antiretroviral therapy: encephalitis and neurodegeneration reconsidered. Curr. HIV/AIDS Rep. 12, 272–279 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  99. Navia, B. A., Cho, E. S., Petito, C. K. & Price, R. W. The AIDS dementia complex: II. Neuropathol. Ann. Neurol. 19, 525–535 (1986). This classic clinicopathological patient series provided the first detailed descriptions of HIV-associated dementia, termed AIDS dementia complex.

    Article  CAS  Google Scholar 

  100. Chang, L. et al. Greater than age-related changes in brain diffusion of HIV patients after 1 year. J. Neuroimmune Pharmacol. 3, 265–274 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  101. Stubbe-Drger, B. et al. Early microstructural white matter changes in patients with HIV: a diffusion tensor imaging study. BMC Neurol. 12, 23 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  102. Wright, P. W., Heaps, J. M., Shimony, J. S., Thomas, J. B. & Ances, B. M. The effects of HIV and combination antiretroviral therapy on white matter integrity. AIDS 26, 1501–1508 (2012).

    Article  CAS  PubMed  Google Scholar 

  103. Hoare, J. et al. Relationship between apolipoprotein E4 genotype and white matter integrity in HIV-positive young adults in South Africa. Eur. Arch. Psychiatry Clin. Neurosci. 263, 189–195 (2013).

    Article  PubMed  Google Scholar 

  104. Kamat, R. et al. Apathy is associated with white matter abnormalities in anterior, medial brain regions in persons with HIV infection. J. Clin. Exp. Neuropsychol. 36, 854–866 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  105. Lentz, M. R. et al. Diffusion tensor and volumetric magnetic resonance measures as biomarkers of brain damage in a small animal model of HIV. PLoS ONE 9, e105752 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Brown, A. Understanding the MIND phenotype: macrophage/microglia inflammation in neurocognitive disorders related to human immunodeficiency virus infection. Clin. Transl. Med. 4, 7 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  107. Gelman, I. H., Zhang, J., Hailman, E., Hanafusa, H. & Morse, S. S. Identification and evaluation of new primer sets for the detection of lentivirus proviral DNA. AIDS Res. Hum. Retroviruses 8, 1981–1989 (1992).

    Article  CAS  PubMed  Google Scholar 

  108. Fischer-Smith, T., Bell, C., Croul, S., Lewis, M. & Rappaport, J. Monocyte/macrophage trafficking in acquired immunodeficiency syndrome encephalitis: lessons from human and nonhuman primate studies. J. Neurovirol. 14, 318–326 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Schuenke, K. & Gelman, B. B. Human microglial cell isolation from adult autopsy brain: brain pH, regional variation, and infection with human immunodeficiency virus type 1. J. Neurovirol. 9, 346–357 (2003).

    Article  PubMed  Google Scholar 

  110. Churchill, M. J. et al. Extensive astrocyte infection is prominent in human immunodeficiency virus-associated dementia. Ann. Neurol. 66, 253–258 (2009).

    Article  PubMed  Google Scholar 

  111. Pu, H. et al. HIV-1 tat protein upregulates inflammatory mediators and induces monocyte invasion into the brain. Mol. Cell. Neurosci. 24, 224–237 (2003).

    Article  CAS  PubMed  Google Scholar 

  112. Chompre, G. et al. Astrocytic expression of HIV-1 Nef impairs spatial and recognition memory. Neurobiol. Dis. 49, 128–136 (2013).

    Article  CAS  PubMed  Google Scholar 

  113. van Marle, G. et al. Human immunodeficiency virus type 1 Nef protein mediates neural cell death: a neurotoxic role for IP-10. Virology 329, 302–318 (2004).

    Article  CAS  PubMed  Google Scholar 

  114. Nguyen, T. P., Soukup, V. M. & Gelman, B. B. Persistent hijacking of brain proteasomes in HIV-associated dementia. Am. J. Pathol. 176, 893–902 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Ferrington, D. A. & Gregerson, D. S. Immunoproteasomes: structure, function, and antigen presentation. Prog. Mol. Biol. Transl. Sci. 109, 75–112 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Ancuta, P. et al. Microbial translocation is associated with increased monocyte activation and dementia in AIDS patients. PLoS ONE 3, e2516 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Gonzalez, E. et al. The influence of CCL3L1 gene-containing segmental duplications on HIV-1/AIDS susceptibility. Science 307, 1434–1440 (2005).

    Article  CAS  PubMed  Google Scholar 

  118. Clements, J. E., Gama, L., Graham, D. R., Mankowski, J. L. & Zink, M. C. A simian immunodeficiency virus macaque model of highly active antiretroviral treatment: viral latency in the periphery and the central nervous system. Curr. Opin. HIV AIDS 6, 37–42 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  119. Dahl, V. et al. Low levels of HIV-1 RNA detected in the cerebrospinal fluid after up to 10 years of suppressive therapy are associated with local immune activation. AIDS 28, 2251–2258 (2014).

    Article  CAS  PubMed  Google Scholar 

  120. Hammond, E. R. et al. Persistent CSF but not plasma HIV RNA is associated with increased risk of new-onset moderate-to-severe depressive symptoms; a prospective cohort study. J. Neurovirol. http://dx.doi.org/10.1007/s13365-015-0416-1, (2016).

  121. Crowe, S., Zhu, T. & Muller, W. A. The contribution of monocyte infection and trafficking to viral persistence, and maintenance of the viral reservoir in HIV infection. J. Leukoc. Biol. 74, 635–641 (2003).

    Article  CAS  PubMed  Google Scholar 

  122. Caniglia, E. C. et al. Antiretroviral penetration into the CNS and incidence of AIDS-defining neurologic conditions. Neurology 83, 134–141 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  123. Gavegnano, C., Fromentin, E. & Schinazi, E. A. Lower levels of nucleoside analog triphosphates in primary human macrophages compared to human leukocytes could impair potency of antiretroviral drugs in human viral reservoirs. Glob. Antiviral J. 4 (Suppl 1), 70 (2008).

    Google Scholar 

  124. Churchill, M. J. et al. Use of laser capture microdissection to detect integrated HIV-1 DNA in macrophages and astrocytes from autopsy brain tissues. J. Neurovirol. 12, 146–152 (2006).

    Article  CAS  PubMed  Google Scholar 

  125. Borjabad, A. et al. Significant effects of antiretroviral therapy on global gene expression in brain tissues of patients with HIV-1-associated neurocognitive disorders. PLoS Pathog. 7, e1002213 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Andersen, A. B. et al. Cerebral FDG-PET scanning abnormalities in optimally treated HIV patients. J. Neuroinflammation 7, 13 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Gelman, B. B. et al. Potential role for white matter lysosome expansion in HIV-associated dementia. J. Acquir. Immune Def. Syndr. 39, 422–425 (2005).

    Article  CAS  Google Scholar 

  128. Chen, X., Hui, L., Geiger, N. H., Haughey, N. J. & Geiger, J. D. Endolysosome involvement in HIV-1 transactivator protein-induced neuronal amyloid beta production. Neurobiol. Aging 34, 2370–2378 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Achim, C. L. et al. Increased accumulation of intraneuronal amyloid β in HIV-infected patients. J. Neuroimmune Pharmacol. 4, 190–199 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  130. Hui, L., Chen, X., Haughey, N. J. & Geiger, J. D. Role of endolysosomes in HIV-1 Tat-induced neurotoxicity. ASN Neuro 4, 243–252 (2012).

    Article  CAS  PubMed  Google Scholar 

  131. Bae, M. et al. Activation of TRPML1 clears intraneuronal Aβ in preclinical models of HIV infection. J. Neurosci. 34, 11485–11503 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Jana, A. & Pahan, K. Human immunodeficiency virus type 1 gp120 induces apoptosis in human primary neurons through redox-regulated activation of neutral sphingomyelinase. J. Neurosci. 24, 9531–9540 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Figuera-Losada, M. et al. Cambinol, a novel inhibitor of neutral sphingomyelinase 2 shows neuroprotective properties. PLoS ONE 10, e0124481 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Shen, D. et al. Lipid storage disorders block lysosomal trafficking by inhibiting a TRP channel and lysosomal calcium release. Nat. Commun. 3, 731 (2012).

    Article  CAS  PubMed  Google Scholar 

  135. Potter, M. C., Figuera-Losada, M., Rojas, C. & Slusher, B. S. Targeting the glutamatergic system for the treatment of HIV-associated neurocognitive disorders. J. Neuroimmune Pharmacol. 8, 594–607 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  136. Vazquez-Santiago, F. J., Noel, R. J. Jr., Porter, J. T. & Rivera-Amill, V. Glutamate metabolism and HIV-associated neurocognitive disorders. J. Neurovirol. 20, 315–331 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Ferrarese, C. et al. Increased glutamate in CSF and plasma of patients with HIV dementia. Neurology 57, 671–675 (2001).

    Article  CAS  PubMed  Google Scholar 

  138. Vesce, S., Bezzi, P., Rossi, D., Meldolesi, J. & Volterra, A. HIV-1 gp120 glycoprotein affects the astrocyte control of extracellular glutamate by both inhibiting the uptake and stimulating the release of the amino acid. FEBS Lett. 411, 107–109 (1997).

    Article  CAS  PubMed  Google Scholar 

  139. Melendez, R. I., Roman, C., Capo-Velez, C. M. & Lasalde-Dominicci, J. A. Decreased glial and synaptic glutamate uptake in the striatum of HIV-1 gp120 transgenic mice. J. Neurovirol. http://dx.doi.org/10.1007/s13365-015-0403-6, (2015).

  140. Musante, V. et al. The HIV-1 viral protein Tat increases glutamate and decreases GABA exocytosis from human and mouse neocortical nerve endings. Cereb. Cortex 20, 1974–1984 (2010).

    Article  PubMed  Google Scholar 

  141. Haughey, N. J., Nath, A., Mattson, M. P., Slevin, J. T. & Geiger, J. D. HIV-1 Tat through phosphorylation of NMDA receptors potentiates glutamate excitotoxicity. J. Neurochem. 78, 457–467 (2001).

    Article  CAS  PubMed  Google Scholar 

  142. Gill, A. J., Kovacsics, C. E., Vance, P. J., Collman, R. G. & Kolson, D. L. Induction of heme oxygenase-1 deficiency and associated glutamate-mediated neurotoxicity is a highly conserved HIV phenotype of chronic macrophage infection that is resistant to antiretroviral therapy. J. Virol. 89, 10656–10667 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Lipton, S. A. Memantine prevents HIV coat protein-induced neuronal injury in vitro. Neurology 42, 1403–1405 (1992).

    Article  CAS  PubMed  Google Scholar 

  144. Muller, W. E., Schroder, H. C., Ushijima, H., Dapper, J. & Bormann, J. gp120 of HIV-1 induces apoptosis in rat cortical cell cultures: prevention by memantine. Eur. J. Pharmacol. 226, 209–214 (1992).

    Article  CAS  PubMed  Google Scholar 

  145. Anderson, E. R., Gendelman, H. E. & Xiong, H. Memantine protects hippocampal neuronal function in murine human immunodeficiency virus type 1 encephalitis. J. Neurosci. 24, 7194–7198 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Schifitto, G. et al. A placebo-controlled study of memantine for the treatment of human immunodeficiency virus-associated sensory neuropathy. J. Neurovirol. 12, 328–331 (2006).

    Article  CAS  PubMed  Google Scholar 

  147. Erdmann, N. et al. Glutamate production by HIV-1 infected human macrophage is blocked by the inhibition of glutaminase. J. Neurochem. 102, 539–549 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Erdmann, J. B. et al. Replicative intermediates of Maize streak virus found during leaf development. J. Gen. Virol. 91 (Pt 4), 1077–1081 (2010).

    Article  CAS  Google Scholar 

  149. Thomas, A. G., Bodner, A., Ghadge, G., Roos, R. P. & Slusher, B. S. GCP II inhibition rescues neurons from gp120IIIB-induced neurotoxicity. J. Neurovirol. 15, 449–457 (2009).

    Article  CAS  PubMed  Google Scholar 

  150. Gupta, S. et al. HIV-Tat elicits microglial glutamate release: role of NAPDH oxidase and the cystine-glutamate antiporter. Neurosci. Lett. 485, 233–236 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Shukla, S. M. & Sharma, S. K. Sinomenine inhibits microglial activation by Aβ and confers neuroprotection. J. Neuroinflammation 8, 117 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Thomas, A. G. et al. High-throughput assay development for cystine-glutamate antiporter (xc) highlights faster cystine uptake than glutamate release in glioma cells. PLoS ONE 10, e0127785 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Beck, S. E. et al. Paving the path to HIV neurotherapy: predicting SIV CNS disease. Eur. J. Pharmacol. 759, 303–312 (2015). This article reviews factors predicting simian immunodeficiency virus CNS disease, including neuroprotective host genes and biomarkers in blood and cerebrospinal fluid.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Sharer, L. R. et al. Comparison of simian immunodeficiency virus and human immunodeficiency virus encephalitides in the immature host. Ann. Neurol. 23, S108–S112 (1988).

    Article  PubMed  Google Scholar 

  155. Zink, M. C. et al. Simian immunodeficiency virus-infected macaques treated with highly active antiretroviral therapy have reduced central nervous system viral replication and inflammation but persistence of viral DNA. J. Infect. Dis. 202, 161–170 (2010). This simian immunodeficiency virus macaque model study demonstrated that the CNS was a reservoir for latent virus in macaques treated with combination antiretroviral therapy.

    Article  CAS  PubMed  Google Scholar 

  156. Zink, M. C. et al. High viral load in the cerebrospinal fluid and brain correlates with severity of simian immunodeficiency virus encephalitis. J. Virol. 73, 10480–10488 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Mankowski, J. L., Clements, J. E. & Zink, M. C. Searching for clues: tracking the pathogenesis of human immunodeficiency virus central nervous system disease by use of an accelerated, consistent simian immunodeficiency virus macaque model. J. Infect. Dis. 186, S199–S208 (2002).

    Article  PubMed  Google Scholar 

  158. Beck, S. E. et al. Macaque species susceptibility to simian immunodeficiency virus: increased incidence of SIV central nervous system disease in pigtailed macaques versus rhesus macaques. J. Neurovirol. 21, 148–158 (2015).

    Article  CAS  PubMed  Google Scholar 

  159. Brew, B. J., Pemberton, L., Cunningham, P. & Law, M. G. Levels of human immunodeficiency virus type 1 RNA in cerebrospinal fluid correlate with AIDS dementia stage. J. Infect. Dis. 175, 963–966 (1997).

    Article  CAS  PubMed  Google Scholar 

  160. Ellis, R. J. et al. Cerebrospinal fluid human immunodeficiency virus type 1 RNA levels are elevated in neurocognitively impaired individuals with acquired immunodeficiency syndrome. Ann. Neurol. 42, 679–688 (1997).

    Article  CAS  PubMed  Google Scholar 

  161. McArthur, J. C. et al. Relationship between human immunodeficiency virus-associated dementia and viral load in cerebrospinal fluid and brain. Ann. Neurol. 42, 689–698 (1997).

    Article  CAS  PubMed  Google Scholar 

  162. Mankowski, J. L., Queen, S. E., Tarwater, P. M., Fox, K. J. & Perry, V. H. Accumulation of β-amyloid precursor protein in axons correlates with CNS expression of SIV gp41. J. Neuropathol. Exp. Neurol. 61, 85–90 (2002).

    Article  CAS  PubMed  Google Scholar 

  163. Weed, M. R. et al. Central nervous system correlates of behavioral deficits following simian immunodeficiency virus infection. J. Neurovirol. 9, 452–464 (2003).

    Article  CAS  PubMed  Google Scholar 

  164. Schmitz, J. E. et al. Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science 283, 857–860 (1999).

    Article  CAS  PubMed  Google Scholar 

  165. Ratai, E. M. et al. CD8+ lymphocyte depletion without SIV infection does not produce metabolic changes or pathological abnormalities in the rhesus macaque brain. J. Med. Primatol. 40, 300–309 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Williams, K. & Burdo, T. H. Monocyte mobilization, activation markers, and unique macrophage populations in the brain: observations from SIV infected monkeys are informative with regard to pathogenic mechanisms of HIV infection in humans. J. Neuroimmune Pharmacol. 7, 363–371 (2012). This article provides an overview of the role of monocytes and macrophages in the CNS in an simian immunodeficiency virus model.

    Article  PubMed  Google Scholar 

  167. Graham, D. R. et al. Initiation of HAART during acute simian immunodeficiency virus infection rapidly controls virus replication in the CNS by enhancing immune activity and preserving protective immune responses. J. Neurovirol. 17, 120–130 (2011).

    Article  CAS  PubMed  Google Scholar 

  168. Marcondes, M. C. et al. Early antiretroviral treatment prevents the development of central nervous system abnormalities in simian immunodeficiency virus-infected rhesus monkeys. AIDS 23, 1187–1195 (2009).

    Article  CAS  PubMed  Google Scholar 

  169. Hatziioannou, T. & Evans, D. T. Animal models for HIV/AIDS research. Nat. Rev. Microbiol. 10, 852–867 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Dreyer, E. B., Kaiser, P. K., Offermann, J. T. & Lipton, S. A. HIV-1 coat protein neurotoxicity prevented by calcium channel antagonists. Science 248, 364–367 (1990).

    Article  CAS  PubMed  Google Scholar 

  171. Toggas, S. M. et al. Central nervous system damage produced by expression of the HIV-1 coat protein gp120 in transgenic mice. Nature 367, 188–193 (1994).

    Article  CAS  PubMed  Google Scholar 

  172. Lee, M. H. et al. Rescue of adult hippocampal neurogenesis in a mouse model of HIV neurologic disease. Neurobiol. Dis. 41, 678–687 (2011).

    Article  CAS  PubMed  Google Scholar 

  173. Okamoto, S. et al. HIV/gp120 decreases adult neural progenitor cell proliferation via checkpoint kinase-mediated cell-cycle withdrawal and G1 arrest. Cell Stem Cell 1, 230–236 (2007).

    Article  CAS  PubMed  Google Scholar 

  174. D'hooge, R., Franck, F., Mucke, L. & De Deyn, P. Age-related behavioural deficits in transgenic mice expressing the HIV-1 coat protein gp120. Eur. J. Neurosci. 11, 4398–4402 (1999).

    Article  CAS  PubMed  Google Scholar 

  175. Jaeger, L. B. & Nath, A. Modeling HIV-associated neurocognitive disorders in mice: new approaches in the changing face of HIV neuropathogenesis. Dis. Model. Mech. 5, 313–322 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Kim, B. O. et al. Neuropathologies in transgenic mice expressing human immunodeficiency virus type 1 Tat protein under the regulation of the astrocyte-specific glial fibrillary acidic protein promoter and doxycycline. Am. J. Pathol. 162, 1693–1707 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Fitting, S. et al. Interactive comorbidity between opioid drug abuse and HIV-1 Tat: chronic exposure augments spine loss and sublethal dendritic pathology in striatal neurons. Am. J. Pathol. 177, 1397–1410 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Fitting, S. et al. Synaptic dysfunction in the hippocampus accompanies learning and memory deficits in human immunodeficiency virus type-1 Tat transgenic mice. Biol. Psychiatry 73, 443–453 (2013). This article highlights the prospects of the use of Tat-transgenic mice in the study of HIV-associated neurocognitive disorder.

    Article  CAS  PubMed  Google Scholar 

  179. Paris, J. J., Singh, H. D., Carey, A. N. & McLaughlin, J. P. Exposure to HIV-1 Tat in brain impairs sensorimotor gating and activates microglia in limbic and extralimbic brain regions of male mice. Behav. Brain Res. 291, 209–218 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Carey, A. N., Sypek, E. I., Singh, H. D., Kaufman, M. J. & McLaughlin, J. P. Expression of HIV-Tat protein is associated with learning and memory deficits in the mouse. Behav. Brain Res. 229, 48–56 (2012).

    Article  CAS  PubMed  Google Scholar 

  181. Paris, J. J., Singh, H. D., Ganno, M. L., Jackson, P. & McLaughlin, J. P. Anxiety-like behavior of mice produced by conditional central expression of the HIV-1 regulatory protein, Tat. Psychopharmacology (Berl.) 231, 2349–2360 (2014).

    Article  CAS  Google Scholar 

  182. Mucke, L., Masliah, E. & Campbell, I. L. Transgenic models to assess the neuropathogenic potential of HIV-1 proteins and cytokines. Curr. Top. Microbiol. Immunol. 202, 187–205 (1995).

    CAS  PubMed  Google Scholar 

  183. Killian, M. S. & Levy, J. A. HIV/AIDS: 30 years of progress and future challenges. Eur. J. Immunol. 41, 3401–3411 (2011).

    Article  CAS  PubMed  Google Scholar 

  184. Lipton, S. A. Requirement for macrophages in neuronal injury induced by HIV envelope protein gp120. NeuroReport 3, 913–915 (1992).

    Article  CAS  PubMed  Google Scholar 

  185. Seay, K. et al. Mice transgenic for CD4-specific human CD4, CCR5 and cyclin T1 expression: a new model for investigating HIV-1 transmission and treatment efficacy. PLoS ONE 8, e63537 (2013). This study highlights the prospects of the use of transgenic mice with human HIV co-receptors to study HIV-associated neurocognitive disorder.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Hanna, Z. et al. Nef harbors a major determinant of pathogenicity for an AIDS-like disease induced by HIV-1 in transgenic mice. Cell 95, 163–175 (1998).

    Article  CAS  PubMed  Google Scholar 

  187. Goffinet, C. et al. Primary T-cells from human CD4/CCR5-transgenic rats support all early steps of HIV-1 replication including integration, but display impaired viral gene expression. Retrovirology 4, 53 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Kopp, J. B. et al. Progressive glomerulosclerosis and enhanced renal accumulation of basement membrane components in mice transgenic for human immunodeficiency virus type 1 genes. Proc. Natl Acad. Sci. USA 89, 1577–1581 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Lund, A. K., Lucero, J., Herbert, L., Liu, Y. & Naik, J. S. Human immunodeficiency virus transgenic rats exhibit pulmonary hypertension. Am. J. Physiol. Lung Cell. Mol. Physiol. 301, L315–L326 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Vigorito, M., Connaghan, K. P. & Chang, S. L. The HIV-1 transgenic rat model of neuroHIV. Brain Behav. Immun. 48, 336–349 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Li, M. D. et al. Transcriptome sequencing of gene expression in the brain of the HIV-1 transgenic rat. PLoS ONE 8, e59582 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Moran, L. M., Booze, R. M., Webb, K. M. & Mactutus, C. F. Neurobehavioral alterations in HIV-1 transgenic rats: evidence for dopaminergic dysfunction. Exp. Neurol. 239, 139–147 (2013).

    Article  CAS  PubMed  Google Scholar 

  193. Fields, J. A. et al. HIV alters neuronal mitochondrial fission/fusion in the brain during HIV-associated neurocognitive disorders. Neurobiol. Dis. 86, 154–169 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Li, W., Galey, D., Mattson, M. P. & Nath, A. Molecular and cellular mechanisms of neuronal cell death in HIV dementia. Neurotox. Res. 8, 119–134 (2005).

    Article  CAS  PubMed  Google Scholar 

  195. Goffinet, C., Allespach, I. & Keppler, O. T. HIV-susceptible transgenic rats allow rapid preclinical testing of antiviral compounds targeting virus entry or reverse transcription. Proc. Natl Acad. Sci. USA 104, 1015–1020 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Goffinet, C. et al. Antagonism of CD317 restriction of human immunodeficiency virus type 1 (HIV-1) particle release and depletion of CD317 are separable activities of HIV-1 Vpu. J. Virol. 84, 4089–4094 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Potash, M. et al. A mouse model for study of systemic HIV-1 infection, antiviral immune responses, and neuroinvasiveness. Proc. Natl Acad. Sci. USA 102, 3760–3765 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Devés, R. & Boyd, C. A. Transporters for cationic amino acids in animal cells: discovery, structure, and function. Physiol. Rev. 78, 487–545 (1998).

    Article  PubMed  Google Scholar 

  199. Hadas, E. et al. Transmission of chimeric HIV by mating in conventional mice: prevention by pre-exposure antiretroviral therapy and reduced susceptibility during estrus. Dis. Model. Mech. 6, 1292–1298 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Potash, M. J. et al. Transmission of chimeric HIV by mating in conventional mice: prevention by pre-exposure antiretroviral therapy and reduced susceptibility during estrus. Dis. Model. Mech. 7, 178–179 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. He, H. et al. Enhanced human immunodeficiency virus Type 1 expression and neuropathogenesis in knockout mice lacking Type I interferon responses. J. Neuropathol. Exp. Neurol. 73, 59–71 (2014). This article highlights the prospects of the use of the EcoHIV mouse model to study HIV-associated neurocognitive disorders.

    Article  CAS  PubMed  Google Scholar 

  202. Kelschenbach, J. L. et al. Mice chronically infected with chimeric HIV resist peripheral and brain superinfection: a model of protective immunity to HIV. J. Neuroimmune Pharmacol. 7, 380–387 (2012).

    Article  PubMed  Google Scholar 

  203. Marsden, M. D. & Zack, J. A. Studies of retroviral infection in humanized mice. Virology 479–480, 297–309 (2015).

    Article  CAS  PubMed  Google Scholar 

  204. Akkina, R. K. et al. Improvements and limitations of humanized mouse models for HIV research: NIH/NIAID 'Meet the Experts' 2015 Workshop Summary. AIDS Res. Hum. Retroviruses 32, 109–119 (2015).

    Article  Google Scholar 

  205. Gorantla, S. et al. Links between progressive HIV-1 infection of humanized mice and viral neuropathogenesis. Am. J. Pathol. 177, 2938–2949 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Dash, P. et al. Loss of neuronal integrity during progressive HIV-1 infection of humanized mice. J. Neurosci. 31, 3148–3157 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Boska, M. D. et al. Associations between brain microstructures, metabolites, and cognitive deficits during chronic HIV-1 infection of humanized mice. Mol. Neurodegener. 9, 58 (2014). This study highlights the prospects of the use of the NSG-hsCD34+ mouse model to study HIV-associated neurocognitive disorder.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Dash, P. K. et al. Long-acting nanoformulated antiretroviral therapy elicits potent antiretroviral and neuroprotective responses in HIV-1-infected humanized mice. AIDS 26, 2135–2144 (2012).

    Article  CAS  PubMed  Google Scholar 

  209. Rongvaux, A. et al. Human hemato-lymphoid system mice: current use and future potential for medicine. Annu. Rev. Immunol. 31, 635–674 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Arguello, P. A. & Gogos, J. A. Cognition in mouse models of schizophrenia susceptibility genes. Schizophr. Bull. 36, 289–300 (2010).

    Article  PubMed  Google Scholar 

  211. Snigdha, S. et al. A preclinical cognitive test battery to parallel the National Institute of Health Toolbox in humans: bridging the translational gap. Neurobiol. Aging 34, 1891–1901 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  212. Young, J. W., Amitai, N. & Geyer, M. A. Behavioral animal models to assess pro-cognitive treatments for schizophrenia. Handb. Exp. Pharmacol. 213, 39–79 (2012).

    Article  CAS  Google Scholar 

  213. Hill, A. L., Rosenbloom, D. I., Fu, F., Nowak, M. A. & Siliciano, R. F. Predicting the outcomes of treatment to eradicate the latent reservoir for HIV-1. Proc. Natl Acad. Sci. USA 111, 13475–13480 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Spudich, S., Lollo, N., Liegler, T., Deeks, S. G. & Price, R. W. Treatment benefit on cerebrospinal fluid HIV-1 levels in the setting of systemic virological suppression and failure. J. Infect. Dis. 194, 1686–1696 (2006).

    Article  CAS  PubMed  Google Scholar 

  215. Yilmaz, A., Svennerholm, B., Hagberg, L. & Gisslen, M. Cerebrospinal fluid viral loads reach less than 2 copies/ml in HIV-1-infected patients with effective antiretroviral therapy. Antivir. Ther. 11, 833–837 (2006).

    CAS  PubMed  Google Scholar 

  216. Letendre, S. L. et al. ING116070: a study of the pharmacokinetics and antiviral activity of dolutegravir in cerebrospinal fluid in HIV-1-infected, antiretroviral therapy-naive subjects. Clin. Infect. Dis. 59, 1032–1037 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Ellis, R. J. et al. Randomized trial of central nervous system-targeted antiretrovirals for HIV-associated neurocognitive disorder. Clin. Infect. Dis. 58, 1015–1022 (2014).

    Article  CAS  PubMed  Google Scholar 

  218. Etherton, M. R., Lyons, J. L. & Ard, K. L. HIV-associated neurocognitive disorders and antiretroviral therapy: current concepts and controversies. Curr. Infect. Dis. Rep. 17, 485 (2015).

    Article  PubMed  Google Scholar 

  219. Robertson, K., Liner, J. & Meeker, R. B. Antiretroviral neurotoxicity. J. Neurovirol. 18, 388–399 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Tovar-y-Romo, L. B. et al. Dendritic spine injury induced by the 8-hydroxy metabolite of efavirenz. J. Pharmacol. Exp. Ther. 343, 696–703 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Ma, Q. et al. Long-term efavirenz use is associated with worse neurocognitive functioning in HIV-infected patients. J. Neurovirol. http://dx.doi.org/10.1007/s13365-015-0382-7, (2015).

  222. Craft, S. et al. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial. Arch. Neurol. 69, 29–38 (2012).

    Article  PubMed  Google Scholar 

  223. Craft, S., Cholerton, B. & Baker, L. D. Insulin and Alzheimer's disease: untangling the web. J. Alzheimers Dis. 33, S263–S275 (2013).

    Article  CAS  PubMed  Google Scholar 

  224. Young, A. C. et al. Cerebral metabolite changes prior to and after antiretroviral therapy in primary HIV infection. Neurology 83, 1592–1600 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Gisslen, M. et al. Elevated cerebrospinal fluid neurofilament light protein concentrations predict the development of AIDS dementia complex. J. Infect. Dis. 195, 1774–1778 (2007).

    Article  CAS  PubMed  Google Scholar 

  226. Heyes, M. P. et al. Quinolinic acid in cerebrospinal fluid and serum in HIV-1 infection: relationship to clinical and neurological status. Ann. Neurol. 29, 202–209 (1991).

    Article  CAS  PubMed  Google Scholar 

  227. Sacktor, N. et al. Novel markers of oxidative stress in actively progressive HIV dementia. J. Neuroimmunol. 157, 176–184 (2004).

    Article  CAS  PubMed  Google Scholar 

  228. Turchan, J. et al. Oxidative stress in HIV demented patients and protection ex vivo with novel antioxidants. Neurology 60, 307–314 (2003).

    Article  CAS  PubMed  Google Scholar 

  229. Boven, L. A. et al. Increased peroxynitrite activity in AIDS dementia complex: implications for the neuropathogenesis of HIV-1 infection. J. Immunol. 162, 4319–4327 (1999).

    CAS  PubMed  Google Scholar 

  230. Li, W. et al. Nitrosative stress with HIV dementia causes decreased L-prostaglandin D synthase activity. Neurology 70, 1753–1762 (2008).

    Article  CAS  PubMed  Google Scholar 

  231. Sevigny, J. J. et al. Evaluation of HIV RNA and markers of immune activation as predictors of HIV-associated dementia. Neurology 63, 2084–2090 (2004).

    Article  CAS  PubMed  Google Scholar 

  232. Cinque, P. et al. Elevated cerebrospinal fluid levels of monocyte chemotactic protein-1 correlate with HIV-1 encephalitis and local viral replication. AIDS 12, 1327–1332 (1998).

    Article  CAS  PubMed  Google Scholar 

  233. Ryan, L. A. et al. Plasma levels of soluble CD14 and tumor necrosis factor-α type II receptor correlate with cognitive dysfunction during human immunodeficiency virus type 1 infection. J. Infect. Dis. 184, 699–706 (2001).

    Article  CAS  PubMed  Google Scholar 

  234. Hagberg, L. et al. Cerebrospinal fluid neopterin: an informative biomarker of central nervous system immune activation in HIV-1 infection. AIDS Res. Ther. 7, 15 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Burdo, T. H., Ellis, R. J. & Fox, H. S. Osteopontin is increased in HIV-associated dementia. J. Infect. Dis. 198, 715–722 (2008).

    Article  CAS  PubMed  Google Scholar 

  236. Erichsen, D. et al. Neuronal injury regulates fractalkine: relevance for HIV-1 associated dementia. J. Neuroimmunol. 138, 144–155 (2003).

    Article  CAS  PubMed  Google Scholar 

  237. Huang, Y. et al. Glutaminase dysregulation in HIV-1-infected human microglia mediates neurotoxicity: relevant to HIV-1-associated neurocognitive disorders. J. Neurosci. 31, 15195–15204 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Pemberton, L. A. & Brew, B. J. Cerebrospinal fluid S-100β and its relationship with AIDS dementia complex. J. Clin. Virol. 22, 249–253 (2001).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Victoria Maranto and Heather Thomas assisted with the preparation of this manuscript by collating references and assisting with preparation of figures. The authors of this manuscript have been supported by NIH grants: 2P30MH075673 (to J.C.M, B.S., N.H., A.B., N.S.); 1P01MH105280 (J.C.M, N.H., N.S.); 271201000036C-5-0-1, MH22005 and R21MH083465 (J.C.M., A.B.); 5R01MH099733, U01AI035042, NS081196 and MH107345 (N.S.); 5R01DA040390, 2R01MH077542, 1R01MH096636 and R03MH103985 (N.H.); DA037611, DA017618 and MH104145 (D.J.V.); AG034852, R21NS07062 and R03DA032470 (B.S.); R01MH083728 (M.P.); P50MH-094268 (M.H.), P01 MH070306 (J.M.); R01NS077869 and R01NS089482 (J.M.). J.C.M. and A.B. are supported by Johns Hopkins University Center for AIDS Research (P30AI094189). M.P. is supported by the Stanley Medical Research Institute, Chevy Chase, Maryland, USA.

Author information

Authors and Affiliations

Authors

Contributions

D.S. and A.M.D. contributed equally to this manuscript. All authors researched the literature for the article and contributed to discussion of the content. D.S., A.M.D., N.H., B.S., M.P., A.B., D.V. and J.C.M. wrote the manuscript. D.S., A.M.D., N.S., N.H., D.V., J.C.M. reviewed and/or edited the manuscript.

Corresponding author

Correspondence to Justin C. McArthur.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Glossary

Sterilizing cure

Elimination of all HIV-infected cells from the individual.

Asymptomatic neurocognitive impairment

(ANI). Cognitive impairment involving at least two cognitive domains that does not interfere with everyday functioning.

Mild neurocognitive disorder

(MND). Cognitive impairment involving at least two cognitive domains that produces at least mild interference in daily functioning.

CSF viral escape

Presence of detectable HIV in the cerebrospinal fluid (CSF) despite undetectable HIV RNA levels in the plasma.

HIV-associated dementia

(HAD). Marked cognitive impairment involving at least two cognitive domains that substantially interferes with daily functioning.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Saylor, D., Dickens, A., Sacktor, N. et al. HIV-associated neurocognitive disorder — pathogenesis and prospects for treatment. Nat Rev Neurol 12, 234–248 (2016). https://doi.org/10.1038/nrneurol.2016.27

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrneurol.2016.27

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing