You are on page 1of 14

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/276041107

VIH-1 Y Alzheimer: ¿una conexión real?

Article  in  Revista Complutense de Ciencias Veterinarias · September 2012


DOI: 10.5209/rev_RCCV.2012.v6.n2.41087

CITATIONS READS

0 400

3 authors, including:

Susana Alvarez Maria Angeles Muñoz-Fernández


Hospital General Universitario Gregorio Marañón Hospital General Universitario Gregorio Marañón
49 PUBLICATIONS   1,365 CITATIONS    698 PUBLICATIONS   17,158 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

nanotechnology View project

HIV, lopinavir/ritonavir View project

All content following this page was uploaded by Maria Angeles Muñoz-Fernández on 17 July 2015.

The user has requested enhancement of the downloaded file.


Rev. Med. Virol. 2012; 22: 33–45.
Published online 23 September 2011 in Wiley Online Library
(wileyonlinelibrary.com)
Reviews in Medical Virology DOI: 10.1002/rmv.711
REVIEW
HIV-1 infection and neurocognitive impairment
in the current era
María del Palacio†, Susana Álvarez† and Mª Ángeles Muñoz-Fernández*
Laboratorio Inmunobiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain

S U M M A RY
Brain HIV-1-infection may result in a syndrome of profound cognitive, behavioral and motor impairment known as
AIDS dementia complex (ADC) in adults and HIV-related encephalopathy in children. Although the introduction of
highly active antiretroviral therapy (HAART) has prolonged and improved the lives of infected individuals, it is clear
that HAART does not provide complete protection against neurological damage in HIV/AIDS. HIV-1 associated de-
mentia is a complex phenomenon, which could be the result of several mechanisms caused by those players using dif-
ferent intracellular signaling pathways. Understanding the causes of neurodegeneration during HIV-1 infection and the
factors which certain individuals develop disease can provide researches on new therapeutic targets to positively affect
disease outcomes. Controlling CNS viral replication with HAART is an essential primary approach, but it should be
complemented with adjunctive CNS-directed therapeutics. Understanding the nature of HIV-1 infection within the
CNS as well as inflammatory responses will ultimately lead to the elimination of HIV-associated neurocognitive disor-
ders. Copyright © 2011 John Wiley & Sons, Ltd.
Received: 3 May 2011; Revised: 5 August 2011; Accepted: 9 August 2011

INTRODUCTION impairment (ANI), to minor neurocognitive disor-


Over 40 million people worldwide are infected by der (MND), to the more severe HIV-associated
HIV-1 (UNAIDS/WHO), and while HIV-1 is most dementia (HAD; also known AIDS dementia com-
well known for its devastating effects on the im- plex, [ADC], or HIV encephalopathy) [1].
mune system and the resulting AIDS, it can also The advent of the highly active antiretroviral ther-
cause several neurological disorders, collectively apy (HAART) has effectively reduced morbidity and
known as HIV-associated neurocognitive disorders mortality in HIV-1 infection. In the vast majority of
(HAND). HAND encompass a hierarchy of pro- antiretroviral-naïve HIV-1-infected patients intro-
gressively more severe patterns of CNS involve- duced to HAART, the plasma viral load decreases to
ment, ranging from asymptomatic neurocognitive undetectable levels, and the cluster of differentiation
4 (CD4) count increases to normal values. Concur-
*Corresponding author: Dr Mª Ángeles Muñoz-Fernández, Laboratorio rently, the incidence of opportunistic complications
Inmuno-Biología Molecular, Hospital General Universitario, Gregorio has decreased. HAART also dramatically decrease
Marañón, C/Doctor Esquerdo 46, 28007 Madrid, Spain.
E-mail: mmunoz.hgugm@salud.madrid.org the (CSF) viral load [2–4].

These authors contributed equally to this work. There are additional factors suggesting that
HAART may not provide sufficient protection to
Abbreviations used
ANI, asymptomatic neurocognitive impairment; ADC, AIDS demen- prevent the CNS complications of HIV infection. Al-
tia complex; ART, antiretroviral therapy; BBB, blood brain barrier; though HAART may arrest neurocognitive impair-
COX, cyclooxygenase; CT, computed tomography; HIVE, HIV-1 en- ment in some populations with HAND [5], a subset
cephalitis; HAD, HIV-associated dementia; HAART, highly active
antiretroviral therapy; HAND, HIV-associated neurocognitive disor- of patients is susceptible to develop a leukoencephalo-
ders; HDS, HIV dementia scale; LPS, lipopolysaccharide; LTR, long pathy, secondary to immune reconstitution in an HIV-
terminal repeat; MCMD, minor cognitive motor disorder; MCP-1, infected CNS [6]. Chronic neuroinflammation is also
monocyte chemoattractant protein 1; MRI, magnetic resonance imag-
ing; MRS, magnetic resonance spectroscopy; NFL, neurofilament pro- present even in HAART-treated patients [7]. These
tein; NMDA, N-methyl-D-aspartate; NO, nitric oxide; PAF, platelet findings suggest that HAART does not provide
activating factor; PET, positron emission tomography; PGE2, prosta- complete protection from HIV-induced inflam-
glandin E2; QUIN, quinolinic acid; SIV, simian immunodeficiency vi-
rus; SPECT, single photon emission computed tomography; TJ, tight matory responses in the CNS and subsequent neuro-
junctions; TLR, toll-like receptors. degeneration. Finally, as life expectancies among

Copyright © 2011 John Wiley & Sons, Ltd.


34 M. D. Palacio et al.

HIV/AIDS patients increase and as the population change in the last research terminology revision
ages, neurologic disorders associated with aging are proposed by Antinori et al. is the addition of the
becoming more prevalent. Indeed, the risk of stroke category of asymptomatic neurocognitive impair-
[8] and diabetes mellitus [9] is increasing among in ment (ANI) based on the observation that some
HIV/AIDS patients, because in part of the marked individuals have demonstrable (and usually mild)
metabolic abnormalities such as hyperglycemia and cognitive impairment demonstrated by formal neu-
hyperlipidemia associated with different combination ropsychological tests without any observed abnor-
antiretroviral therapy regimens coupled with advanc- mality in everyday functioning. ANI is defined by
ing age. performance, at least 1 SD below the mean of de-
Effective control of CNS viral replication through mographically adjusted normative scores in at least
HAART is the essential primary approach, but that two cognitive areas [1].
it should be complemented with adjunctive CNS- There is evidence for a change in HAD in the era
directed therapeutics. HIV-induced inflammation of HAART, apart from the potentially increased im-
and its consequences, expressed both systemically portance of confounding factors (hepatitis C, tes-
and within the CNS, are attractive targets that tosterona deficiency, effects of aging. . .)[13]. So,
could be approached through multiple pharmaco- there is an increased frequency of verbal memory
logic agents. impairment along with a more classical distribu-
tion of impairment (fine motor incoordination),
CLINICAL FEATURES the mesial temporal lobe abnormalities appear to
Before the widespread use of HAART in the devel- be more relevant in a completed prospective posi-
oped world, 20%–30% of individuals infected with tron emission tomography (PET)–CSF study, and
HIV-1 developed a range of cognitive and motor standard CSF markers of ADC, such as beta-2
symptoms, including impaired short-term memory, microglobulin and the viral load in the CSF, are
reduced concentration, and leg weakness that are no longer sensitive to the presence or severity of
collectively known as HAD. These symptoms often HAD [14].
occurred together with behavioral symptoms, such Highly active antiretroviral therapy has, thus,
as personality changes, apathy, and social with- raised the CD4 cell count which is commonly seen
drawal, and HAD could, in its more severe forms, in patients with HAD and has introduced two po-
lead to a nearly vegetative and mute state. The on- tentially new risk factors: nadir CD4 cell count
set of HAD is correlated with high plasma viral [15] and disease duration [14]. In the Cohen’s study
loads, which accounts for the strong effects of where they examine patterns of brain volume loss
HAART on reducing its incidence, although with in HIV-1 infected adults on HAART and demo-
the longer lifespan of patients with HIV-1, there is graphic and clinical factors contributing to brain
increasing prevalence of HAD [10,11]. Therefore, volume loss, the results suggest that the duration
with the use of HAART, a more subtle form of of a patient’s infection may influence certain brain
CNS dysfunction, minor cognitive motor disorder volumes, independent of other clinical factors, such
(MCMD), has either become more common or is as cognitive status, viral load, and immunological
more obvious because it does not progress to status. Similarly, nadir CD4 was a significant factor
HAD [10,11]. In this syndrome, loss of memory in half the models examined, primary in the cortical
and decreases in computational and other higher regions and the hippocampus, suggesting that a
cortical functions are much less pronounced. It story of advanced immune suppression may exert
has recently been estimated that ~10% of HIV-1 a negative effect on brain structure. The association
infected adults have HAD but that MCMD might between immune suppression and brain volume
be several times more common, involving perhaps has not yet been previously described in HIV-1 in-
as many as 30% of the HIV-1 infected population. fection and provides further rationale for initiating
Furthermore, the clinical presence of MCMD has antiretroviral treatment early in the course of infec-
been associated with pathological changes in the tion [16].
CNS that are characteristic of HIV-1 invasion HIV-1 infection is now a chronic condition and can
(known as HIV-1 encephalitis; HIVE), and MCMD exhibit interactions with other neurodegenerative
is associated with a worse overall prognosis for disorders. Notably, increased b-amyloid is found in
HIV-1 infected individuals [12]. The most notable the brain in HIV-1 infection, and patients with

Copyright © 2011 John Wiley & Sons, Ltd. Rev. Med. Virol. 2012; 22: 33–45.
DOI: 10.1002/rmv
NeuroAIDS in the current era 35

HAND show increased aggregation of b-amyloid lymphocytes, an increased number of microglia


within neurons [17]. The complexities in disease and macrophages correlates well with the severity
pathobiology for HAND are becoming even broader. of pre-mortem HAND [19,22]. Infection of the
CNS by HIV-1 can be detected and monitored by
NEUROPATHOGENESIS measurement of viral load in CSF. Several groups
The CNS is susceptible to infection by retroviruses have reported a positive correlation between CSF
of various species and by members of the lentivirus viral load and the observed degree of cognitive
family, in particular. The specific requirements for dysfunction in patients with HAND [23,24]. More-
entry to the brain and the many cell types in the over, CSF viral load appears to correlate with viral
CNS increase the complexity of virus—cell interac- load in brain measured by quantitative PCR
tions in the brain. In theory, five main cell types [24,25], and the highest concentrations of virus are
(astrocytes, oligodendrocytes, neurons, perivascu- observed in those subcortical structures most fre-
lar macrophages, and microglia) are susceptible to quently affected in patients with severe HAND/
retroviral infection, but of these five, the latter two HAD [25].
are the most commonly infected by HIV-1 [18]. In addition to initial neuroinvasion and infection
of perivascular macrophages and microglia, factors
Mechanisms of neuroinvasion of human associated with progressive HIV-1 infection in the
immunodeficiency virus-1 periphery, thus outside the brain, may be required
Human immunodeficiency virus-1 enters the brain to eventually trigger the development of HAND
early in the course of infection, presumably via and dementia [26]. One such factor could be an el-
infected macrophages and lymphocytes, and then evated number of circulating monocytes expressing
persists primarily in perivascular macrophages two markers of activated monocytes, CD16 and
and microglia [19,20]. Three pathways have been CD69. Another important player may be the blood
proposed for viral entry into the brain: (i) carriage brain barrier (BBB), which separates the CNS from
of HIV-1 by infected leukocytes (“Trojan horse” hy- the periphery and supposedly controls the traffic
pothesis); (ii) passage of cell-free virus into the of low-molecular-weight nutrients, peptides, pro-
brain; and (iii) release of virus into the brain by teins, and cells in and out of the brain (see for
infected endothelial cells [18] (Figure 1). This can BBB review [27]). Thus, the condition of the BBB
occur within 1–2 weeks after the virus enters into may potentially determine continuing or repeated
the systemic circulation [21]. In contrast to neuroinvasion during the course of HIV disease.

Figure 1. HIV-1 neuroinvasion. (1) According to the "Trojan Horse hypothesis" entry of HIV-1 into the brain takes place by the migration
of infected monocytes, which differentiate into perivascular macrophage. (2) Entrance of HIV-1 by transcytosis of brain microvascular en-
dothelial cells. (3) The direct entrance of the virus. Once the virus is in the brain, it infects productively macrophages and microglia

Copyright © 2011 John Wiley & Sons, Ltd. Rev. Med. Virol. 2012; 22: 33–45.
DOI: 10.1002/rmv
36 M. D. Palacio et al.

Tumor necrosis factor-a is known to increase the Other co-receptors for HIV-1 infection have also
permeability of the BBB to HIV-infected cells [28], been identified, which are known as chemokine
induce expression of cell adhesion molecules, and receptors.
upregulate monocyte chemoattractant protein-1 Chemokine receptors belong in the large cate-
(MCP-1) [29]. Following extravasation, infected gory of G-protein-coupled seven transmembrane-
monocytes/perivascular macrophages begin to spanning domain receptors and mediate leukocyte
actively produce and release HIV/SIV, as well trafficking and contribute intimately to the organi-
as a variety of proinflammatory mediators that zation of inflammatory responses of the immune
impacts the function of surrounding cells and system [37–40]. Chemokines and their receptors
enhances further monocyte recruitment [30–32]. are in fact indispensable for maintenance, matura-
This occurs concurrently with dysregulation of tion, and migration of hematopoietic and neural
tight junctions (TJ). In vivo, several studies have stem cells [40]. However, the most prominent patho-
shown that the loss of transmembrane TJ proteins logical function of certain chemokine receptors
and perijunctional proteins in microvessels is asso- seems to be the mediation of HIV-1 infection [37–39].
ciated with increased permeability of the BBB fol- Infection by HIV-1 of macrophages and lympho-
lowing HIV/SIV neuroinvasion [33]. Fiala et al. cytes in the periphery and microglia in the brain
have described changes in TJ protein zo-1 pattern- can occur after binding of the viral envelope pro-
ing in brains of patients with HIVE, with a com- tein gp120 to one of several possible chemokine
plete lack of zo-1 staining in cortical vessels of receptors in conjunction with CD4. Depending on
one patient [28]. the exact variant of gp120, different HIV-1 strains
Human immunodeficiency virus-1 infection may use CCR5, and CCR3, or CXCR4, or a combi-
compromises the structural integrity of the intesti- nation of these chemokine receptors to enter target
nal tract and can cause leakage of bacteria into the cells [39]. It is assumed that HIV-1 in the brain uses
blood stream. Such microbial translocation results CCR5 as the principal coreceptor for infecting tar-
in elevated plasma levels of bacterial lipopolysac- get cells [41], although R5, X4, and R5/X4 isolates
charide (LPS), and in HIV-infected/AIDS patients, all have been found in the CNS of HIV-1-infected
is associated with increased monocyte activation individuals [42]. Several in vitro studies strongly
and dementia [34,35]. Additionally, it has recently suggest that CXCR4 is prominently involved in
reported that HAND is associated with higher HIV-associated neuronal damage, whereas CCR5
systemic levels of plasma LPS. The hypothesis is may play a dual role by being able to either serve
that HIV-1 infected patients with microbial traslo- a toxic or protective role [43–45]. It has been de-
cation from the gut will have higher plasma LPS scribed the presence of very different HIV-1 isolates
and thus more systemic immune activation lead- in the brain despite no great differences in clinical,
ing to increased risk of transit of HIV-infected pathological, or immunological parameters, indi-
monocytes into end organs including the CNS. cating that there is not a particular phenotypic
Another study suggests that HIV-1 infection property of the HIV-1 virus that might explain its
increases the vulnerability of the BBB in response neurovirulence and/or neurotropism [46].
to LPS and facilitates the transmigration of pe- Neurons and astrocytes express CCR5 and
ripheral monocytes/macrophages [36]. These find- CXCR4 [47,48], however, it is unclear which recep-
ings support an important role for toll-like tors are involved in HIV-1 entry in these cells. Al-
receptors besides monocytes and macrophages in though most studies have found a lack of infection
HAD [36]. of neurons, there are some reports of HIV-1 DNA
and protein expression in these cells [49], and we
have previously shown that neuroblastoma cells
Cells target in brain infection are susceptible to HIV-1 infection [50,51]. The possi-
Cluster of differentiation 4 is a primary receptor bility of neuronal infection certainly would be an
for HIV-1 infection and is commonly found on important factor in neuropathology, and infected
peripheral blood mononuclear cells especially on neurons, like astrocytes, may provide a reservoir of
T-cells, monocytes, and macrophages. Microglia virus with the capacity for reactivation. Oligoden-
and perivascular macrophages are the only two cell drocytes are believed to be minimally involved in
types into the brain that express CD4 antigens. HIV-1 dementia [18].

Copyright © 2011 John Wiley & Sons, Ltd. Rev. Med. Virol. 2012; 22: 33–45.
DOI: 10.1002/rmv
NeuroAIDS in the current era 37

Studies of cultured human astrocytes have may cause alterations in neurotransmitter action
shown that HIV-1 infection is initially productive and causes leukoencephalopathy resulting in neu-
yet noncytopathic and ultimately is converted to a ronal apoptosis [60]. TNF-a, platelet activating fac-
latent stage with little expression of viral protein. tor, prostaglandin E2 (PGE2), nitric oxide (NO), and
The frequency of astrocyte infection at perivascular quinolinic acid (QUIN) also cause neurotoxicity.
regions in severe HIVE (16%–19%) is similar to the NO is produced by microvascular endothelial cells,
frequency of lymphocyte infection in lymph nodes macrophages, and neurons that may result in N-
of patients with AIDS (15%–30%)[52,53]. Because methyl-D-aspartate type glutamate-associated neu-
approximately 70% of the cells in brain are astro- rotoxicity. Elevated levels of NO synthase have
cytes, they are a significant reservoir of latent been reported in the brain of HAD patients,
HIV-1 DNA, being a significant factor in HIV-1-me- whereas a 40-fold increase in expression of NO
diated neuropathogenesis. Although astrocyte dys- synthase has been described in neurons of drug ad-
function is just one of several pathways leading to dict HIV-1 adults [61,62]. TNF-a is produced by
neurodegeneration in AIDS, Churchill et al. demon- macrophages and microglia, and it mainly affects
strated that neurodegeneration resulting from as- oligodendrocytes, and it has been reported an ele-
trocyte dysfunction may be a consequence of vated level of TNF-a mRNA in HIV-1 adults with
astrocyte infection [54]. Under certain conditions, neurological complications [63].
latent HIV-1 infection of astrocytes can be reacti- Enhanced levels of PGE2, the major product of
vated following stimulation with TNF-a and IL-1b COX-2, have been detected in cerebrospinal fluid
[55,56]. Production of viral structural proteins in in patients with HAD, and correlate with severity
these seemingly latently infected cells could be of dementia [64]. Moreover, COX-2-positive macro-
stimulated by the addition of those cytokines in phages infiltrating the brain of patients with HIV-1
the persistent phase, but the stimulatory response encephalitis have been described [65], and viral
decreased with time after initial transfection. Stim- proteins as gp120 and Tat are able to induce COX-
ulation of fetal astrocytes with TNF-a led to an in- 2 and PGE2 synthesis in astrocytic and neuroblas-
crease of HIV-long terminal repeat (LTR) activity toma cells [66,67].
and increased binding of NF-kB to consensus It is, thus, apparent that the cellular responses to
kB binding sequence in the HIV-long terminal re- infection, and the effects of these responses on dif-
peat [57]. However, cytokine-stimulation resulted ferent cell populations in the brain, are the links be-
mainly in increase of the abundance of small early tween HIV-1 infection, HIV-1 encephalopathy, and
mRNAs [56]. HAD (Figure 2).

The inflammatory cascade Neuropathology


Although the low number of infected cells in the The hallmarks of HIV-1 associated neuropathology
brain cannot explain the extent of damage observed include the following: infiltration of macrophages;
in HIV-1 encephalopathy, it is widely accepted that the formation of microglial nodules and multinu-
viral proteins shed by infected cells, as well as a vari- cleated giant cells, suggestive of virus-induced fu-
ety of toxic products secreted by activated cells sion of microglia and/or macrophages, in central
(infected or uninfected), are the major factors in- white matter and deep gray matter; widespread re-
volved in the underlying neuropathology [58]. active astrogliosis, indicative of astrocyte activation
Excessive production of inflammatory biomole- and damage; the loss of specific neuron subpopula-
cules or mediators of inflammation produced by tions, particularly those involved in cognition (hip-
different cell types of CNS may induce neurotoxic- pocampus) and motor function (basal ganglia); the
ity. Monocytes, lymphocytes, and activated macro- loss of synaptic connections; and myelin pallor, or
phages after entering into CNS release various pro- the loss of myelin surrounding neuronal axons, in-
inflammatory, inflammatory cytokines, reactive ox- dicating damage to oligodendrocytes; and the pres-
ygen, and other biomolecules with high neurotoxic ence of HIV-1 DNA in the CSF [58,68]. Table 1
potential. These mediators individually, additively, shows potential complications associated with ex-
or synergistically disrupt normal functioning of aggerated neuroinflammation following peripheral
cells of CNS by inducing neurotoxicity [59]. This immune challenge.

Copyright © 2011 John Wiley & Sons, Ltd. Rev. Med. Virol. 2012; 22: 33–45.
DOI: 10.1002/rmv
38 M. D. Palacio et al.

Figure 2. Mechanism of neuropathogenesis. The two main components of this mechanism are the direct effect of the HIV-1 infection,
including HIV-1 proteins, and the indirect consequence of infection comprising the secretion of cytokines and neurotoxins. The
infected macrophages and microglia participate actively in the neurodegeneration shedding viral proteins and releasing significant
amount of cytokines and neurotoxins into the CNS. The alteration of astrocytes function results in an increase in the level of neuro-
toxicity in the brain. Neurotoxins released from several sources lead to neuronal injury

Some of the risk factor for all forms of HAND in- sex [69]. In early cohorts, studies before HAART,
clude: a high viral set point early in HIV infection, higher CSF levels of viral load were associated with
age (risk increases with increasing age), low CD4 progression to HAD, but this relationship is not
counts [15,16], anemia [26], low body mass index, seen in HAART-treated individuals. In contrast to
systemic symptoms, injection drug use and the fre- a-chemokines, b-chemokines are only expressed at
quency of coinfection with hepatitis C and female relatively low levels in the brain under normal con-
ditions. However, several b-chemokines are found
at increased concentrations in the CNS following
Table 1. Complications associated with HIV infection: CCL2, MIP-1a, MIP-1b, and
enhanced neuroinflammation. RANTES/CCL5 [70]. Furthermore, microglia acti-
vated by interferons and astrocytes activated by
Potential complications IL-1b and TNF-a express CCL2, which could also
contribute to the positive correlation between
Behavioral and physiological
CCL2 levels and increased risk of HAND [71–73].
Prolonged sickness
b-chemokines may contribute to neuronal toxicity
Prolonged depressive-like behavior
via existing pathways that are overstimulated by
Altered febrile response
higher than normal concentrations of these factors.
Exaggerated weight loss
Circulating HIV DNA concentrations, which
Cognitive
might be related to persistence of infection in cells
Impaired hippocampal-dependent memory
of the monocyte/macrophage lineage, are higher
Delirium
in individuals with HAND [74,75]. The identifica-
Neuronal
tion of a subpopulation of long-term survivors sug-
Dendritic atrophy
gested the possibility that host genetic factors
Decreased neurogenesis
influence HIV-1 pathogenesis. Multiple studies
Impaired long term potentiation
have confirmed the protective impact of CCR5Δ32
Axonal damage
on HIV-1 disease and have identified additional

Copyright © 2011 John Wiley & Sons, Ltd. Rev. Med. Virol. 2012; 22: 33–45.
DOI: 10.1002/rmv
NeuroAIDS in the current era 39

polymorphisms that alter disease progression. difficulties and whether these effects are active at
Apolipoprotein E is associated with severity of de- the encounter. It is anticipated that this dilemma
mentia, specifically among older HIV-1 infected will become increasingly important as the HIV-1-
individuals, making neurons more vulnerable to infected population ages and their vulnerability to
oxidative stress. neurodegenerative disease, particularly Alzheimer
Disease, increases [78]. Additionally, newer, atypi-
DIAGNOSTIC STUDIES cal CNS diseases have been described during the
Despite its basic characterization, more than two treatment era that may need to be distinguished
decades ago, the nosology and diagnosis of HAND from classic HAD, including CNS escape [79], im-
are difficult both in clinical practice and clinical mune reconstitution syndrome [6,80], and HIV
trials. The diagnosis relies on recognition of the leukoencephalopathy.
clinical syndrome and exclusion of alternative diag- The second, related clinical shortcoming centers
noses, rather than on specific laboratory-based on the problem of distinguishing ongoing disease
findings. The suggested revision in research termi- activity from static injury because of prior insults,
nology proposed by Antinori et al. adds quantita- caused either by HIV-1 or by other conditions.
tive standardization to evaluation, but does not There is a clear need for objective, laboratory-based
add greater specificity to diagnosis [1]. criteria for this condition, both in the clinic and in
Neuroimaging should be obtained in the process clinical trials, in order to predict risk, assess diagno-
of diagnostic evaluation in all patients with AIDS sis, and aid ongoing management. These consid-
and cognitive impairment. Certain findings on erations have rekindled interest in the use of CSF
computed tomography or magnetic resonance im- (or blood) biomarkers and biological measures
aging (MRI) are consistent with or even suggestive obtained by magnetic resonance methodologies
of HAD, including diffuse cerebral atrophy and [77]. Among the promising CSF indicators are mar-
subcortical or periventricular white matter changes kers of neural injury and local immune activation.
that appear hypodense on computed tomography The axonal neurofilament light chain protein and
or bright on T2 sequences on MRI. However, neu- the neuronal protein tau are frequently elevated in
roimaging in this setting is most useful as a means CSF in patients with HAD, may be detected before
to exclude other common neurological conditions the development of HAD, and normalize with
in individuals with AIDS, because neither atrophy treatment, whereas CSF immunoactivation mar-
nor white matter changes are sensitive or specific kers such as neopterin and MCP-1 (CCL2), and
for HAND [16,76]. Among other techniques, single CSF viral load can be used to measure the effects
photon emission computed tomography, PET, and of treatment on CNS intrathecal immunoactivation
magnetic resonance spectroscopy have been inves- and virus replication [81–83].
tigated as tools for both diagnosis and pathogenetic Various CSF markers of persistent intrathecal im-
studies of HAD [77]. mune activation or neuronal injury, such as neopterin,
Cerebrospinal fluid analysis is required in febrile or B2 microglobulin, MCP-1, and neurofilament protein,
encephalopathic patients to exclude opportunistic in- quinolinic acid (QUIN) also correlate with dementia
fections, but is generally not necessary in the more severity, but have not been fully validated [77].
typical nonfebrile, slowly progressive forms of HAND, Because of the obvious advantages of sampling
although it gets obtained important information. blood rather than CSF, it would be desirable to be
But patients now presenting with HAND fre- able to use blood markers for diagnosis and man-
quently have other comorbidities that both predis- agement. A number of studies have examined
pose to HAD and confound diagnosis. Thus, HIV- blood concentrations of the same type of markers
1-infected adults with a history of drug or alcohol as discussed for CSF, without suggesting clinical
abuse, psychiatric disease, head trauma, or nutri- utility. Cohen et al. demonstrate in a study that cy-
tional deficiencies are more frequently excluded tokine concentrations (beyond MCP-1 and MIP-
from effective treatment and are more often poorly 1b), in particular, the interleukins (IL-1b, IL-6, IL-
adherent to medications. Because of concomitant 8, IL-10, IL-16, IL-18..) were found to be strongly
neurologic dysfunction related to their risk for predictive of neurocognitive performance across a
HIV-1 infection, it is often difficult to determine range of tests of attention, executive functioning,
the effects of HIV-1 on their cognitive and motor and psychomotor speed [84].

Copyright © 2011 John Wiley & Sons, Ltd. Rev. Med. Virol. 2012; 22: 33–45.
DOI: 10.1002/rmv
40 M. D. Palacio et al.

Given the continued impact of HIV-1 associated There are several well-documented studies show-
cognitive dysfunction across the world and the ing that HAART has led to a very dramatic decline
evolving nature of neurological damage, it is even in the incidence of HAD in adult population.
more important to study the CSF in the context of In general, CSF HIV-1 infection responds very
HIV-1 infection. We need the equivalent of the excel- well to HAART [82,89], so that when plasma viral
lent surrogate marker plasma viral load to apply to load levels become undetectable, so do CSF levels.
the diagnosis, staging, and treatment of HAD [85]. However, the relative rates of viral decay in the
The three conditions comprising HAND (ANI, two compartments may differ in some patients,
MND, and HAD) may be classified using a variety with HIV-1 viral load concentrations falling more
of specific clinical and laboratory-based methods, slowly in the CSF than in plasma. Slower decay
depending upon the resources available in the setting has been noted in subjects with HAD and lower
where the patients are being evaluated. Nevertheless, CD4+ T-cell counts but without CSF pleocytosis
standardized procedures should be followed when- [90,91]. These observations can be interpreted as
ever possible, both to collect the needed information being consistent with a simple model of compart-
and to interpret that information to make three types mentalized CSF infection [92], with the lag in CSF
of determination: (i) the presence and severity of neu- viral response due either to slow cell turnover and
rocognitive impairment; (ii) the presence and severity consequent prolonged viral release by macro-
of functional decline; and (iii) the degree to which phages or to less potent drug concentrations within
cognitive impairment or functional decline are likely the CSF as a result of reduced drug entry.
to have been influenced by comorbid conditions or The CNS is a site that antiretroviral therapy
confounds (including HIV-related opportunistic (ART) often has difficulty reaching. Owing to its
CNS conditions, or unrelated developmental, psychi- physical structure, the presence of efflux pumps
atric, or neuromedical confounds). and its high expression of metabolizing enzymes,
A number of brief screening measures, including the BBB is an effective barrier against many antire-
the HIV dementia scale, mental alteration test, execu- troviral drugs [93,94]. In addition, (ART) are com-
tive interview, and HIV dementia assessment have monly protein bound within the plasma, further
been developed or adapted to screen for HAD. Over- limiting their access to the CNS [93,95]. Drugs eas-
all, there remains a clinical need for reliable and brief ily passing the BBB and affecting (macrophages in)
screening measures to detect and monitor HIV-re- the CNS are so called neuroactive drugs (neuro-
lated cognitive dysfunction as well as to quantify HAART). As with most antimicrobials, brain pene-
any response to therapeutic interventions [86–88]. tration is crucial to achieve the goal of maximal
suppression of HIV-1 replication, although it is un-
clear how well antiretroviral CSF concentrations
TREATMENT OF HUMAN actually reflect parenchymal concentrations. Sev-
IMMUNODEFICIENCY VIRUS-ASSOCIATED eral studies have shown an association between
NEUROCOGNITIVE DISORDERS WITH the use of neuroactive drugs (defined in different
ANTIRETROVIRALS. ADJUNCTIVE AGENTS ways) and good neurocognitive performance. The
A major obstacle in HIV-1 eradication is the ability cerebral penetration effectiveness index (CPE)
of the virus to remain latent in a subpopulation of ranking system has been proposed as a simple
the cells it infects. Latently infected cells can escape method for estimating the combined CNS effective-
the viral immune response and persist for long per- ness of antiretroviral regimens and has been shown
iods of time, despite the presence of successful to correlate with CSF viral load in a cohort study
HAART. Given the appropriate stimulus, latently [96]. But despite this, increased levels of intrathecal
infected cells can reactivate and start producing in- immune activation are found in a majority of sub-
fectious virions. The susceptibility of these cell jects successfully treated with antiretroviral ther-
populations to HIV-1, their life span, their prolifer- apy, although it is unclear if immune activation
ative capacity, and their ability to periodically pro- results from viral replication within the CNS or as
duce infectious virus subsequent to alterations in a response to other factors [97].
cellular physiology and/or immunologic controls Human immunodeficiency virus-associated neu-
are critical issues that determine the contribution rocognitive disorders may only reverse slowly; for
of these cells to viral persistence. example, in a study following individuals with

Copyright © 2011 John Wiley & Sons, Ltd. Rev. Med. Virol. 2012; 22: 33–45.
DOI: 10.1002/rmv
NeuroAIDS in the current era 41

HAND for up to 5 years after initiation of HAART NANOMEDICINE AND CENTRAL NERVOUS
>50% of individuals had not shown reversal of def- SYSTEM DRUG DELIVERY
icits [98]. Therefore, HAART alleviates HAND [99] Because HIV-1 can easily enter the CNS but antire-
but raise the question whether HAART is suffi- troviral drugs cannot, the brain essentially becomes
cient to make HAND disappear [100]. Augment- a sanctuary for HIV-1 [106,107], acting not only as a
ing the power of an already effective regimen by reservoir, making it difficult to completely eradi-
adding another potent drug, preferably one with cate the virus, but also increasing the chance of a vi-
a different mode of action should reduce the level ral mutation leading to drug resistance [108].
of viral load insofar as it originates from ongoing Owing to the importance of getting antiretroviral
rounds of viral replication. Thus, treatment inten- drugs into the brain, a number of nanotechnology-
sification with maraviroc or lopinavir/ritonavir based methods for crossing the BBB are being spe-
(good CNS penetration) for 4 weeks and 4 weeks cifically developed for this class of compounds.
with enfuvirtide (poor CNS penetration) has no ef- These methods include disruption of the BBB, de-
fect on residual CSF viral load levels or intrathecal velopment of nanoparticles with increased BBB
immune activation over the course of the study permeability, uptake by brain microvascular endo-
[101]. thelial cells via adsorptive-mediated transcytosis
Given that aberrant immune activation is likely to and cell-mediated delivery.
play a pivotal role in the CNS damage induced by A broad range of nanomedicines is being devel-
HIV-1, clinical trials have focused on adjunctive thera- oped to improve drug delivery for CNS disorders
pies targeted at attenuating CNS inflammation [102], [109,110]. BBB penetrance is dependent on nano-
neuroprotectants agents would be inhibitors of cell- particle size, shape, and protein and lipid coatings.
signaling pathways that are likely to be involved in These all affect drug uptake, release and ingress
neurodegeneration, inhibitors of excitotoxicity, and across the barrier. Recent reports support the no-
inhibitors of oxidative and nitrosative stress. tion that nanotechnology can serve to improve the
Adjuvant treatment strategies including neuro- delivery of antiretroviral drugs, called nanoART,
protection, immunomodulation, and symptom across the BBB and affect biodistribution and clini-
control have been reviewed by Turchan and collea- cal benefit for HIV-1 disease [111,112]. To test CNS-
gues [103]. Overall, none has shown clear and sub- penetrating ART, cell-based nanoparticle delivery
stantial clinical effects. The monoamine oxidase-B systems were developed in an effort to transport
inhibitor selegiline, administered using the selegi- ART across the BBB. Taking advantage of the Trojan
line transdermal system, was one of the recently horse principle, these nanoparticles are taken up by
tested antioxidative drugs for the treatment of monocytes and sheltered within the monocytes as
HIV-associated cognitive impairment psychiatric they are carried across the BBB. The drug is then re-
medications, such as serotonin reuptake inhibitors leased to act within the CNS [113], and for this,
(citalopram, paroxetine), lithium and valproic acid these cells make great candidates for cell-mediated
(both glucagon synthase kinase-3b inhibitors) have drug delivery to the CNS. Other laboratories have
been tested as adjunct treatments of HAND and conjugated nanoparticles with Tat, which has an af-
found to have limited beneficial effect [104]. Mino- finity for nuclear transport mechanisms [114]. This
cycline, a tetracycline-type antibiotic, has emerged results in a nanoparticle that has high CNS penetra-
as a potential inhibitor of microglial activation bility while still bypassing efflux transporters to
and inhibits the reactive oxygen species production prolong exposure within the CNS.
and expression of proinflammatory cytokines by
reactive microglia, indicating that minocycline is CONCLUSIONS
not only neuroprotective but also exhibits inhibi- Given the rising prevalence of HIV/AIDS in the in-
tory effect on microglial activation [105]. Future dustrialized and developing world, it is likely that
trials might include using humanized, monoclonal new HIV-associated neurologic syndromes will
antibodies against IL-1, IL-6, or TNF-a. Another ap- emerge as time progresses. NeuroAIDS is expected
proach might target QUIN, a neurotoxin present in to be a major upcoming health issue among long-
large quantities in the CSF and brains of patients term HIV seropositive survivors and AIDS
with AIDS dementia that is produced by activated patients. Today in developed countries, HAART
macrophages and microglia. has changed AIDS from a fatal disease to a more

Copyright © 2011 John Wiley & Sons, Ltd. Rev. Med. Virol. 2012; 22: 33–45.
DOI: 10.1002/rmv
42 M. D. Palacio et al.

manageable chronic infection. But even with ag- specifically targets the CNS and adjunctive therapies
gressive therapy, the virus cannot be completely now becoming available, eliminating virus (and
eradicated, and regions that remain relatively iso- virus-mediated activation) in the brain is a realistic
lated from the systemic circulation, such as the goal. Understanding the signals that lead to their
CNS, might provide a “sanctuary” for latent or passage into the brain and in attenuating viral
slowly replicating virus. growth and inflammatory responses within the
Reduced severity of disease has paralleled low- CNS will ultimately lead to the elimination of
ered viral replication and reduced overt neuropa- HAND.
thology. What remains are neuroinflammatory
responses heralded by low levels of viral replication,
disordered glial crosstalk and monocyte transmigra- CONFLICT OF INTEREST
tion into the CNS. With antiretroviral treatment that The authors have no competing interest.

REFERENCES
1. Antinori A, Arendt G, Becker JT, et al. infected patients. Neurology 2007; 68: antiretroviral therapy era. Journal of Neuro-
Updated research nosology for HIV- 1257–1261. virology 2010; 16: 25–32.
associated neurocognitive disorders. 9. Ledergerber B, Furrer H, Rickenbach M, 17. Achim CL, Adame A, Dumaop W, et al.
Neurology 2007; 69: 1789–1799. et al. Factors associated with the inci- Increased accumulation of intraneuronal
2. Price RW, Spudich S. Antiretroviral ther- dence of type 2 diabetes mellitus in HIV- amyloid beta in HIV-infected patients.
apy and central nervous system HIV type infected participants in the Swiss HIV Journal of Neuroimmune Pharmacology
1 infection. Journal of Infectious Diseases cohort study. Clinical Infectious Diseases 2009; 4: 190–199.
2008; 197(Suppl 3):S294–306. 2007; 45: 111–119. 18. Kramer-Hammerle S, Rothenaigner I,
3. McArthur JC, McDermott MP, McClernon 10. McArthur JC. HIV dementia: an evolving Wolff H, et al. Cells of the central nervous
D, et al. Attenuated central nervous sys- disease. Journal of Neuroimmunology 2004; system as targets and reservoirs of the hu-
tem infection in advanced HIV/AIDS 157: 3–10. man immunodeficiency virus. Virus Re-
with combination antiretroviral therapy. 11. Ellis RJ, Calero P,Stockin MD. HIV infec- search 2005; 111: 194–213.
Archives of Neurology 2004; 61: 1687–1696. tion and the central nervous system: a 19. Petito CK, Cho ES, Lemann W, et al. Neu-
4. Robertson KR, Robertson WT, Ford S, et al. primer. Neuropsychology Review 2009; 19: ropathology of acquired immunodefi-
Highly active antiretroviral therapy 144–151. ciency syndrome (AIDS): an autopsy
improves neurocognitive functioning. 12. Cherner M, Masliah E, Ellis RJ, et al. Neu- review. Journal of Neuropathology and Exper-
Journal of Acquired Immune Deficiency Syn- rocognitive dysfunction predicts postmor- imental Neurology 1986; 45: 635–646.
dromes 2004; 36: 562–566. tem findings of HIV encephalitis. 20. Koenig S, Gendelman HE, Orenstein JM,
5. Shanbhag MC, Rutstein RM, Zaoutis T, Neurology 2002; 59: 1563–1567. et al. Detection of AIDS virus in macro-
et al. Neurocognitive functioning in pedi- 13. Brew BJ, Crowe SM, Landay A, et al. Neu- phages in brain tissue from AIDS patients
atric human immunodeficiency virus in- rodegeneration and ageing in the HAART with encephalopathy. Science 1986; 233:
fection: effects of combined therapy. era. Journal of Neuroimmune Pharmacology 1089–1093.
Archives of Pediatrics & Adolescent Medicine 2009; 4: 163–174. 21. Gray F, Hurtrel M, Hurtrel B. Early central
2005; 159: 651–656. 14. Brew BJ. Evidence for a change in AIDS nervous system changes in human immu-
6. Rackstraw S, Meadway J, Bingham J, dementia complex in the era of highly ac- nodeficiency virus (HIV)-infection. Neuro-
et al. An emerging severe leukoencepha- tive antiretroviral therapy and the possi- pathology and Applied Neurobiology 1993;
lopathy: is it due to HIV disease or bility of new forms of AIDS dementia 19: 3–9.
highly active antiretroviral therapy? Inter- complex. AIDS 2004; 18(Suppl 1): S75–S78. 22. Glass JD, Fedor H, Wesselingh SL, et al.
national Journal of STD and AIDS 2006; 17: 15. Munoz-Moreno JA, Fumaz CR, Ferrer MJ, Immunocytochemical quantitation of hu-
205–207. et al. Nadir CD4 cell count predicts neuro- man immunodeficiency virus in the brain:
7. Anthony IC, Ramage SN, Carnie FW, et al. cognitive impairment in HIV-infected correlations with dementia. Annals of Neu-
Influence of HAART on HIV-related CNS patients. AIDS Research and Human Retro- rology 1995; 38: 755–762.
disease and neuroinflammation. Journal of viruses 2008; 24: 1301–1307. 23. Brew BJ, Pemberton L, Cunningham P,
Neuropathology and Experimental Neurology 16. Cohen RA, Harezlak J, Schifitto G, et al. et al. Levels of human immunodeficiency
2005; 64: 529–536. Effects of nadir CD4 count and duration virus type 1 RNA in cerebrospinal fluid
8. Ortiz G, Koch S, Romano JG, et al. of human immunodeficiency virus infec- correlate with AIDS dementia stage. Jour-
Mechanisms of ischemic stroke in HIV- tion on brain volumes in the highly active nal of Infectious Diseases 1997; 175: 963–966.

Copyright © 2011 John Wiley & Sons, Ltd. Rev. Med. Virol. 2012; 22: 33–45.
DOI: 10.1002/rmv
NeuroAIDS in the current era 43

24. McArthur JC, McClernon DR, Cronin MF, 36. Wang H, Sun J, Goldstein H. Human im- Sciences of the United States of America 2000;
et al. Relationship between human immu- munodeficiency virus type 1 infection 97: 8075–8080.
nodeficiency virus-associated dementia increases the in vivo capacity of peripheral 46. Alvarez S, Jimenez JL, Serramia MJ, et al.
and viral load in cerebrospinal fluid monocytes to cross the blood-brain barrier Lack of association of HIV-1 biological or
and brain. Annals of Neurology 1997; 42: into the brain and the in vivo sensitivity of molecular properties with neurotropism
689–698. the blood-brain barrier to disruption by li- for brain cells. Journal of Molecular Neuro-
25. Wiley CA, Soontornniyomkij V, Radhakrish- popolysaccharide. Journal of Virology 2008; science 2006; 29: 131–144.
nan L, et al. Distribution of brain HIV load 82: 7591–7600. 47. Asensio VC, Campbell IL. Chemokines in
in AIDS. Brain Pathology 1998; 8: 277–284. 37. Locati M, Murphy PM. Chemokines and the CNS: plurifunctional mediators in di-
26. Gartner S. HIV infection and dementia. chemokine receptors: biology and clinical verse states. Trends in Neurosciences 1999;
Science 2000; 287: 602–604. relevance in inflammation and AIDS. 22: 504–512.
27. Banks WA, Ercal N, Price TO. The blood- Annual Review of Medicine 1999; 50: 48. Miller RJ, Meucci O. AIDS and the brain: is
brain barrier in neuroAIDS. Current HIV 425–440. there a chemokine connection? Trends in
Research 2006; 4: 259–266. 38. Dragic T, Litwin V, Allaway GP, et al. HIV- Neurosciences 1999; 22: 471–479.
28. Fiala M, Looney DJ, Stins M, et al. TNF- 1 entry into CD4+ cells is mediated by the 49. Canto-Nogues C, Sanchez-Ramon S,
alpha opens a paracellular route for HIV-1 chemokine receptor CC-CKR-5. Nature Alvarez S, et al. HIV-1 infection of neu-
invasion across the blood-brain barrier. 1996; 381: 667–673. rons might account for progressive HIV-
Molecular Medicine 1997; 3: 553–564. 39. Oberlin E, Amara A, Bachelerie F, et al. 1-associated encephalopathy in children.
29. Weiss JM, Downie SA, Lyman WD, et al. The CXC chemokine SDF-1 is the ligand Journal of Molecular Neuroscience 2005; 27:
Astrocyte-derived monocyte-chemoattrac- for LESTR/fusin and prevents infection 79–89.
tant protein-1 directs the transmigration by T-cell-line-adapted HIV-1. Nature 1996; 50. Alvarez Losada S, Canto-Nogues C,
of leukocytes across a model of the human 382: 833–835. Munoz-Fernandez MA. A new possible
blood-brain barrier. Journal of Immunology 40. Tran PB, Miller RJ. Chemokine receptors: mechanism of human immunodefi-
1998; 161: 6896–6903. signposts to brain development and dis- ciency virus type 1 infection of neural
30. Orandle MS, MacLean AG, Sasseville VG, ease. Nature Reviews Neuroscience 2003; 4: cells. Neurobiology of Disease 2002; 11:
et al. Enhanced expression of proinflam- 444–455. 469–478.
matory cytokines in the central nervous 41. van der Meer P, Ulrich AM, Gonzalez- 51. Obregon E, Punzon C, Fernandez-Cruz E,
system is associated with neuroinvasion Scarano F, et al. Immunohistochemical et al. HIV-1 infection induces differ-
by simian immunodeficiency virus and analysis of CCR2, CCR3, CCR5, and entiation of immature neural cells through
the development of encephalitis. Journal CXCR4 in the human brain: potential autocrine tumor necrosis factor and nitric
of Virology 2002; 76: 5797–5802. mechanisms for HIV dementia. Experimen- oxide production. Virology 1999; 261:
31. Toborek M, Lee YW, Flora G, et al. tal and Molecular Pathology 2000; 69: 193–204.
Mechanisms of the blood-brain barrier dis- 192–201. 52. Embretson J, Zupancic M, Beneke J, et al.
ruption in HIV-1 infection. Cellular and Mo- 42. Gorry PR, Bristol G, Zack JA, et al. Macro- Analysis of human immunodeficiency
lecular Neurobiology 2005; 25: 181–199. phage tropism of human immunodefi- virus-infected tissues by amplification
32. Librizzi L, Mazzetti S, Pastori C, et al. Acti- ciency virus type 1 isolates from brain and in situ hybridization reveals latent
vation of cerebral endothelium is required and lymphoid tissues predicts neurotrop- and permissive infections at single-cell
for mononuclear cell recruitment in a ism independent of coreceptor specificity. resolution. Proceedings of the National
novel in vitro model of brain inflamma- Journal of Virology 2001; 75: 10073–10089. Academy of Sciences of the United States of
tion. Neuroscience 2006; 137: 1211–1219. 43. Kaul M, Ma Q, Medders KE, et al. HIV-1 America 1993; 90: 357–361.
33. Maclean AG, Belenchia GE, Bieniemy DN, coreceptors CCR5 and CXCR4 both medi- 53. Nuovo GJ, Becker J, Burk MW, et al. In
et al. Simian immunodeficiency virus dis- ate neuronal cell death but CCR5 paradox- situ detection of PCR-amplified HIV-1
rupts extended lengths of the blood--brain ically can also contribute to protection. Cell nucleic acids in lymph nodes and
barrier. Journal of Medical Primatology 2005; Death and Differentiation 2007; 14: 296–305. peripheral blood in patients with
34: 237–242. 44. Hesselgesser J, Taub D, Baskar P, et al. asymptomatic HIV-1 infection and ad-
34. Ancuta P, Kamat A, Kunstman KJ, et al. Neuronal apoptosis induced by HIV-1 vanced-stage AIDS. Journal of Acquired
Microbial translocation is associated with gp120 and the chemokine SDF-1 alpha is Immune Deficiency Syndromes 1994; 7:
increased monocyte activation and de- mediated by the chemokine receptor 916–923.
mentia in AIDS patients. PloS One 2008; CXCR4. Current Biology 1998; 8: 595–598. 54. Churchill MJ, Wesselingh SL, Cowley D,
3: e2516. 45. Meucci O, Fatatis A, Simen AA, et al. Ex- et al. Extensive astrocyte infection is prom-
35. Brenchley JM, Price DA, Douek DC. HIV pression of CX3CR1 chemokine receptors inent in human immunodeficiency virus-
disease: fallout from a mucosal catastro- on neurons and their role in neuronal sur- associated dementia. Annals of Neurology
phe? Nature Immunology 2006; 7: 235–239. vival. Proceedings of the National Academy of 2009; 66: 253–258.

Copyright © 2011 John Wiley & Sons, Ltd. Rev. Med. Virol. 2012; 22: 33–45.
DOI: 10.1002/rmv
44 M. D. Palacio et al.

55. Tornatore C, Nath A, Amemiya K, et al. 64. Griffin DE, Wesselingh SL, McArthur JC. subcortical brain injury in HIV infection.
Persistent human immunodeficiency virus Elevated central nervous system prosta- Neurology 2006; 66: 1255–1257.
type 1 infection in human fetal glial cells glandins in human immunodeficiency 74. Shiramizu B, Gartner S, Williams A, et al.
reactivated by T-cell factor(s) or by the virus-associated dementia. Annals of Neu- Circulating proviral HIV DNA and HIV-
cytokines tumor necrosis factor alpha and rology 1994; 35: 592–597. associated dementia. AIDS 2005; 19: 45–52.
interleukin-1 beta. Journal of Virology 1991; 65. Fiala M, Liu QN, Sayre J, et al. Cyclooxy- 75. Shiramizu B, Williams AE, Shikuma C,
65: 6094–6100. genase-2-positive macrophages infiltrate et al. Amount of HIV DNA in peripheral
56. Tornatore C, Meyers K, Atwood W, et al. the Alzheimer’s disease brain and damage blood mononuclear cells is proportional
Temporal patterns of human immunodefi- the blood-brain barrier. European Journal of to the severity of HIV-1-associated neuro-
ciency virus type 1 transcripts in human fetal Clinical Investigation 2002; 32: 360–371. cognitive disorders. The Journal of Neuro-
astrocytes. Journal of Virology 1994; 68: 93–102. 66. Blanco A, Alvarez S, Fresno M, et al. Extra- psychiatry and Clinical Neurosciences 2009;
57. Atwood WJ, Tornatore CS, Traub R, et al. cellular HIV-Tat induces cyclooxygenase-2 21: 68–74.
Stimulation of HIV type 1 gene expression in glial cells through activation of nuclear 76. Tucker KA, Robertson KR, Lin W, et al.
and induction of NF-kappa B (p50/p65)- factor of activated T cells. Journal of Immu- Neuroimaging in human immunodefi-
binding activity in tumor necrosis factor nology 2008; 180: 530–540. ciency virus infection. Journal of Neuroim-
alpha-treated human fetal glial cells. AIDS 67. Alvarez S, Blanco A, Fresno M, et al. Nu- munology 2004; 157: 153–162.
Research and Human Retroviruses 1994; clear factor-kappaB activation regulates 77. Price RW, Epstein LG, Becker JT, et al. Bio-
10:1207–1211. cyclooxygenase-2 induction in human markers of HIV-1 CNS infection and in-
58. Gendelman HE, Lipton SA, Tardieu M, astrocytes in response to CXCL12: role in jury. Neurology 2007; 69: 1781–1788.
et al. The neuropathogenesis of HIV-1 in- neuronal toxicity. Journal of Neurochemistry 78. Valcour V, Paul R. HIV infection and de-
fection. Journal of Leukocyte Biology 1994; 2010; 113: 772–783. mentia in older adults. Clinical Infectious
56: 389–398. 68. Neuenburg JK, Brodt HR, Herndier BG, Diseases 2006; 42: 1449–1454.
59. Munoz-Fernandez MA, Fresno M. The et al. HIV-related neuropathology, 1985 79. Wendel KA, McArthur JC. Acute menin-
role of tumour necrosis factor, interleukin to 1999: rising prevalence of HIV en- goencephalitis in chronic human immuno-
6, interferon-gamma and inducible nitric cephalopathy in the era of highly active deficiency virus (HIV) infection: putative
oxide synthase in the development and antiretroviral therapy. Journal of Acquired central nervous system escape of HIV rep-
pathology of the nervous system. Progress Immune Deficiency Syndromes 2002; 31: lication. Clinical Infectious Diseases 2003; 37:
in Neurobiology 1998; 56: 307–340. 171–177. 1107–1111.
60. Panek RB, Benveniste EN. Class II MHC 69. Jayadev S, Garden GA. Host and viral fac- 80. McCombe JA, Auer RN, Maingat FG, et al.
gene expression in microglia. Regulation tors influencing the pathogenesis of HIV- Neurologic immune reconstitution inflam-
by the cytokines IFN-gamma, TNF-alpha, associated neurocognitive disorders. Jour- matory syndrome in HIV/AIDS: outcome
and TGF-beta. Journal of Immunology 1995; nal of Neuroimmune Pharmacology 2009; 4: and epidemiology. Neurology 2009; 72:
154: 2846–2854. 175–189. 835–841.
61. Adamson DC, Wildemann B, Sasaki M, 70. Kelder W, McArthur JC, Nance-Sproson T, 81. Abdulle S, Hagberg L, Svennerholm B,
et al. Immunologic NO synthase: elevation et al. Beta-chemokines MCP-1 and RANTES et al. Continuing intrathecal immunoacti-
in severe AIDS dementia and induction by are selectively increased in cerebrospinal vation despite two years of effective anti-
HIV-1 gp41. Science 1996; 274: 1917–1921. fluid of patients with human immunodefi- retroviral therapy against HIV-1 infection.
62. Minagar A, Shapshak P, Fujimura R, et al. ciency virus-associated dementia. Annals of AIDS 2002; 16: 2145–2149.
The role of macrophage/microglia and Neurology 1998; 44: 831–835. 82. Spudich S, Lollo N, Liegler T, et al. Treat-
astrocytes in the pathogenesis of three 71. Sevigny JJ, Albert SM, McDermott MP, ment benefit on cerebrospinal fluid HIV-1
neurologic disorders: HIV-associated de- et al. An evaluation of neurocognitive sta- levels in the setting of systemic virological
mentia, Alzheimer disease, and multiple tus and markers of immune activation as suppression and failure. Journal of Infec-
sclerosis. Journal of Neurological Sciences predictors of time to death in advanced tious Diseases 2006; 194: 1686–1696.
2002; 202: 13–23. HIV infection. Archives of Neurology 2007; 83. Hagberg L, Cinque P, Gisslen M, et al. Ce-
63. Wesselingh SL, Takahashi K, Glass JD, 64: 97–102. rebrospinal fluid neopterin: an informative
et al. Cellular localization of tumor necro- 72. Sevigny JJ, Albert SM, McDermott MP, biomarker of central nervous system im-
sis factor mRNA in neurological tissue et al. Evaluation of HIV RNA and markers mune activation in HIV-1 infection. AIDS
from HIV-infected patients by combined of immune activation as predictors of HIV- Research and Therapy 2010; 7:15.
reverse transcriptase/polymerase chain associated dementia. Neurology 2004; 63: 84. Cohen RA, de la Monte S, Gongvatana A,
reaction in situ hybridization and immu- 2084–2090. et al. Plasma cytokine concentrations asso-
nohistochemistry. Journal of Neuroimmunol- 73. Ragin AB, Wu Y, Storey P, et al. Monocyte ciated with HIV/hepatitis C coinfection
ogy 1997; 74: 1–8. chemoattractant protein-1 correlates with are related to attention, executive and

Copyright © 2011 John Wiley & Sons, Ltd. Rev. Med. Virol. 2012; 22: 33–45.
DOI: 10.1002/rmv
NeuroAIDS in the current era 45

psychomotor functioning. Journal of Neu- pharmacotherapy of brain HIV-1 infection. 104. Ances BM, Letendre SL, Alexander T, et al.
roimmunology 2011; 233: 204–210. Glia 2008; 56: 1711–1735. Role of psychiatric medications as adjunct
85. McArthur JC, Letendre SL. Is the glass 95. Boffito M, Back DJ, Blaschke TF, et al. Pro- therapy in the treatment of HIV associated
three-quarters full or one-quarter empty? tein binding in antiretroviral therapies. neurocognitive disorders. International Re-
Journal of Infectious Diseases 2006; 194: AIDS Research and Human Retroviruses view of Psychiatry 2008; 20: 89–93.
1628–1631. 2003; 19: 825–835. 105. Dheen ST, Kaur C, Ling EA. Microglial ac-
86. Bottiggi KA, Chang JJ, Schmitt FA, et al. 96. Letendre S, Marquie-Beck J, Capparelli E, tivation and its implications in the brain
The HIV Dementia Scale: predictive power et al. Validation of the CNS penetration- diseases. Current Medicinal Chemistry
in mild dementia and HAART. Journal of effectiveness rank for quantifying antire- 2007; 14: 1189–1197.
Neurological Sciences 2007; 260: 11–15. troviral penetration into the central 106. Alexaki A, Liu Y, Wigdahl B. Cellular
87. Smith CA, van Gorp WG, Ryan ER, et al. nervous system. Archives of Neurology reservoirs of HIV-1 and their role in viral
Screening subtle HIV-related cognitive 2008; 65: 65–70. persistence. Current HIV Research 2008; 6:
dysfunction: the clinical utility of the HIV 97. Eden A, Price RW, Spudich S, et al. Im- 388–400.
dementia scale. Journal of Acquired Immune mune activation of the central nervous 107. Banks WA, Robinson SM, Nath A. Perme-
Deficiency Syndromes 2003; 33: 116–118. system is still present after >4 years of ef- ability of the blood-brain barrier to HIV-1
88. Woods SP, Moore DJ, Weber E, et al. Cog- fective highly active antiretroviral therapy. Tat. Experimental Neurology 2005; 193:
nitive neuropsychology of HIV-associated Journal of Infectious Diseases 2007; 196: 218–227.
neurocognitive disorders. Neuropsychology 1779–1783. 108. Bagasra O. A unified concept of HIV la-
Review 2009; 19: 152–168. 98. Tozzi V, Balestra P, Bellagamba R, et al. tency. Expert Opinion on Biological Therapy
89. Mellgren A, Antinori A, Cinque P, et al. Persistence of neuropsychologic deficits 2006; 6: 1135–1149.
Cerebrospinal fluid HIV-1 infection usu- despite long-term highly active antire- 109. Jimenez JL, Clemente MI, Weber ND, et al.
ally responds well to antiretroviral treat- troviral therapy in patients with HIV- Carbosilane dendrimers to transfect hu-
ment. Antiviral Therapy 2005; 10: 701–707. related neurocognitive impairment: man astrocytes with small interfering
90. Eggers C, Hertogs K, Sturenburg HJ, et al. prevalence and risk factors. Journal of RNA targeting human immunodeficiency
Delayed central nervous system virus sup- Acquired Immune Deficiency Syndromes virus. BioDrugs 2010; 24: 331–343.
pression during highly active antiretro- 2007; 45: 174–182. 110. Mintzer MA, Simanek EE. Nonviral vec-
viral therapy is associated with HIV 99. McCutchan JA, Wu JW, Robertson K, et al. tors for gene delivery. Chemical Reviews
encephalopathy, but not with viral drug HIV suppression by HAART preserves 2009; 109: 259–302.
resistance or poor central nervous sys- cognitive function in advanced, immune- 111. Nowacek A, Gendelman HE. NanoART,
tem drug penetration. AIDS 2003; 17: reconstituted AIDS patients. AIDS 2007; neuroAIDS and CNS drug delivery. Nano-
1897–1906. 21: 1109–1117. medicine (London, England) 2009; 4: 557–574.
91. Ellis RJ, Gamst AC, Capparelli E, et al. Ce- 100. Simioni S, Cavassini M, Annoni JM, et al. 112. Kuo YC, Chen HH. Effect of nanoparticu-
rebrospinal fluid HIV RNA originates Cognitive dysfunction in HIV patients de- late polybutylcyanoacrylate and methyl-
from both local CNS and systemic sources. spite long-standing suppression of vire- methacrylate-sulfopropylmethacrylate on
Neurology 2000; 54: 927–936. mia. AIDS 2010; 24: 1243–1250. the permeability of zidovudine and lami-
92. Schnell G, Spudich S, Harrington P, et al. 101. Yilmaz A, Verhofstede C, D’Avolio A, et al. vudine across the in vitro blood-brain bar-
Compartmentalized human immunodefi- Treatment intensification has no effect on rier. International Journal of Pharmaceutics
ciency virus type 1 originates from long- the HIV-1 central nervous system infection 2006; 327: 160–169.
lived cells in some subjects with HIV-1-as- in patients on suppressive antiretroviral 113. Dou H, Morehead J, Destache CJ, et al.
sociated dementia. PLoS Pathogens 2009; 5: therapy. Journal of Acquired Immune Defi- Laboratory investigations for the morpho-
e1000395. ciency Syndromes 2010; 55(5): 590–596. logic, pharmacokinetic, and anti-retroviral
93. McGee B, Smith N, Aweeka F. HIV phar- 102. Zink MC, Uhrlaub J, DeWitt J, et al. Neuro- properties of indinavir nanoparticles in
macology: barriers to the eradication of protective and anti-human immunodefi- human monocyte-derived macrophages.
HIV from the CNS. HIV Clinical Trials ciency virus activity of minocycline. Virology 2007; 358: 148–158.
2006; 7: 142–153. JAMA 2005; 293: 2003–2011. 114. Berry CC, de la Fuente JM, Mullin M, et al.
94. Ronaldson PT, Persidsky Y, Bendayan R. 103. Turchan J, Sacktor N, Wojna V, et al. Neu- Nuclear localization of HIV-1 tat functio-
Regulation of ABC membrane transpor- roprotective therapy for HIV dementia. nalized gold nanoparticles. IEEE Transac-
ters in glial cells: relevance to the Current HIV Research 2003; 1: 373–383. tions on Nanobioscience 2007; 6: 262–269.

Copyright © 2011 John Wiley & Sons, Ltd. Rev. Med. Virol. 2012; 22: 33–45.
DOI: 10.1002/rmv

View publication stats

You might also like