Sie sind auf Seite 1von 18

Downloaded from http://perspectivesinmedicine.cshlp.

org/ on October 18, 2013 - Published by Cold Spring Harbor Laboratory Press

HIV-1-Related Central Nervous System Disease: Current Issues in


Pathogenesis, Diagnosis, and Treatment
Serena Spudich and Francisco Gonzlez-Scarano

Cold Spring Harb Perspect Med 2012; doi: 10.1101/cshperspect.a007120 originally published online
February 28, 2012

Subject Collection HIV

HIV Pathogenesis: Dynamics and Genetics of HIV-1 Pathogenesis: The Virus


Viral Populations and Infected Cells Ronald Swanstrom and John Coffin
John Coffin and Ronald Swanstrom
Human Immunodeficiency Virus Vaccine Trials The T-Cell Response to HIV
Robert J. O'Connell, Jerome H. Kim, Lawrence Bruce Walker and Andrew McMichael
Corey, et al.
HIV Transmission HIV-1 Reverse Transcription
George M. Shaw and Eric Hunter Wei-Shau Hu and Stephen H. Hughes
Novel Cell and Gene Therapies for HIV HIV Pathogenesis: The Host
James A. Hoxie and Carl H. June A.A. Lackner, Michael M. Lederman and Benigno
Rodriguez
Behavioral and Biomedical Combination HIV: Cell Binding and Entry
Strategies for HIV Prevention Craig B. Wilen, John C. Tilton and Robert W. Doms
Linda-Gail Bekker, Chris Beyrer and Thomas C.
Quinn
HIV-1 Assembly, Budding, and Maturation Innate Immune Control of HIV
Wesley I. Sundquist and Hans-Georg Krusslich Mary Carrington and Galit Alter
HIV-1 Assembly, Budding, and Maturation HIV DNA Integration
Wesley I. Sundquist and Hans-Georg Krusslich Robert Craigie and Frederic D. Bushman
Lessons in Nonhuman Primate Models for AIDS HIV-1-Related Central Nervous System Disease:
Vaccine Research: From Minefields to Milestones Current Issues in Pathogenesis, Diagnosis, and
Jeffrey D. Lifson and Nancy L. Haigwood Treatment
Serena Spudich and Francisco Gonzlez-Scarano

For additional articles in this collection, see http://perspectivesinmedicine.cshlp.org/cgi/collection/

Copyright 2012 Cold Spring Harbor Laboratory Press; all rights reserved
HIV-1-Related Central Nervous System Disease:
Current Issues in Pathogenesis, Diagnosis,
and Treatment

Serena Spudich1 and Francisco Gonzalez-Scarano2


1
Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06520
2
Department of Neurology, The University of Texas School of Medicine at San Antonio,
San Antonio, Texas 78229
Correspondence: scarano@mail.med.upenn.edu

HIV-associated central nervous system (CNS) injury continues to be clinically significant in


the modern era of HIV infection and therapy. A substantial proportion of patients with
suppressed HIV infection on optimal antiretroviral therapy have impaired performance on
neuropsychological testing, suggesting persistence of neurological abnormalities despite
treatment and projected long-term survival. In the underresourced setting, limited accessibil-
ity to antiretroviral medications means that CNS complications of later-stage HIV infection
continue to be a major concern. This article reviews key recent advances in our understand-
www.perspectivesinmedicine.org

ing of the neuropathogenesis of HIV, focusing on basic and clinical studies that reveal viral
and host features associated with viral neuroinvasion, persistence, and immunopathogenesis
in the CNS, as well as issues related to monitoring and treatment of HIV-associated CNS
injury in the current era.

IV-1 infects the nervous system in virtually suppressive treatment, there appears to be a
H all patients with systemic infection and fre-
quently causes central nervous system (CNS)
continued prevalence of mild-moderate neuro-
cognitive impairment in a significant pro-
and peripheral nervous system (PNS) disorders. portion or even a majority of patients. This
Until the introduction of combination antire- disquieting finding, combined with the stagger-
troviral therapy (cART) in the mid-1990s, ing numbers of patients who continue to be
HIV-1-associated dementia (HAD) and related newly infected with HIV worldwide, and the
cognitive and motor disorders affected 20% limited availability of optimal antiretroviral
30% of patients with advanced immunosup- treatment in many of the persons affected with
pression or AIDS. The incidence of overt this condition, make understanding and effec-
HAD in countries where effective combination tively preventing HIV-1-related neurological
antiretroviral medications are widely available injury a continued key area of investigation. To
is now markedly diminished. However, in encompass this more complex range of disor-
the setting of chronic, apparently systemically ders seen in patients treated with cART, most

Editors: Frederic D. Bushman, Gary J. Nabel, and Ronald Swanstrom


Additional Perspectives on HIV available at www.perspectivesinmedicine.org
Copyright # 2012 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a007120
Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120

1
S. Spudich and F. Gonzalez-Scarano

investigators now refer to HIV-associated neu- of comorbidities and neurodegeneration that


rocognitive disorders (HAND) rather than occur with aging. Additionally, because cART
HAD as the principal primary CNS complica- appears to be beneficial in the amelioration
tion of HIV infection. and prevention of the most severe forms of
HAND, newfound attention has been focused
on the possible long-term cognitive benefits of
HISTORY
initiation of cART in early stages of HIV
A dementing illness characterized by attention infection.
and memory deficits, motor impairment, and
personality changes was recognized in a signifi-
cant proportion of patients with advanced KEY ADVANCES IN THE AREA
AIDS within the first years of the HIV epide-
Viral Entry and Maintenance of Infection
mic (Navia et al. 1986b). Further investigation
in the Nervous System
of this disorder revealed that these complica-
tions were a direct result of HIV-1 infection As with some other viruses that circulate in the
and attendant inflammation in the CNS. The bloodstream, HIV entry into the CNS is largely
neuropathology was characterized by diffuse mediated through blood lymphocytes and
brain atrophy with large ventricles, widespread monocytes that enter the perivascular spaces
low-grade inflammation with microglial nod- either in the course of their natural surveillance,
ules, perivascular lymphocyte cuffing, multi- or because they are attracted by chemokines
nucleated cells expressing HIV p24 and other to sites of inflammation. Viral strains isolated
antigens, and patchy demyelination and white from the brain are more commonly CCR5-
matter gliosis (Gabuzda et al. 1986; Navia tropic and replicate effectively in cultured mac-
www.perspectivesinmedicine.org

et al. 1986a). Although inexorably progressive rophages, suggesting that monocytes may pre-
to severe disability and death in the absence of dominate as Trojan horses in the process of
disease-modifying HIV therapy, the course of CNS entry, as described years ago for classically
this clinical disorder has been altered consider- described lentiviruses such as visna virus of
ably by treatment with antiretroviral therapy sheep (Haase 1986). Alternatively, HIV may be
and especially cART. Originally defined as the brought into the CNS by lymphocytes, which
AIDS-dementia complex (ADC) based on can harbor viruses that replicate in macro-
motor, cognitive, and behavioral symptoms phages (Collman et al. 1992), or conceivably
and signs, current research nosology defines a as free virions, where the means of entry would
broader spectrum now called HIV-associated be through endothelial cells. Regardless of the
neurocognitive disorder, with graded classifi- mechanism of entry, cells of the macrophage
cations based on abnormal performance on lineage are the only cells in the brain that are
neuropsychological testing, and the presence routinely found to harbor HIV antigens or
or absence of a patients perception of func- RNA by conventional methods such as immu-
tional limitation related to cognitive impair- nohistochemistry or in situ hybridization for
ment (Antinori et al. 2007). Changes in the viral RNA, although other cell types such as
severity of neurological disease in the current astrocytes may harbor HIV sequences without
era may also be accompanied by alterations robust expression of RNA (or proteins) (Wiley
in the underlying etiology of neurological et al. 1986). Detection of such infection requires
morbidity in the setting of long-term survival other methodology such as in situ polymerase
with HIV, including the consequences of possi- chain reaction (PCR) amplification or laser
ble ongoing low-grade viral replication and capture microdissection followed by PCR
inflammation within the CNS, cumulative (Churchill et al. 2009). Recent studies have
exposure to antiretroviral and other medica- also concentrated on determining whether a
tions, chronic systemic inflammation leading subset of monocytes is particularly important
to accelerated vascular disease, and the effects in either delivering virus to the CNS or in

2 Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120


Pathogenesis and Management of Neuro-AIDS

amplifying a local inflammatory process, are mostly derived from the circulating mono-
whether such a subset is increased or enhanced cytes, are highly infected in the brains of HIV-
by systemic inflammation mediated by circulat- infected persons or in macaques (rhesus or
ing bacterial products, and which cytokines and pig-tailed) that are infected experimentally
chemokines increase the recruitment of mono- with SIV (Kim et al. 2006); these may be labeled
cytes and lymphocytes into the CNS and by by CD163, a marker for this subtype that
extension are more likely to deliver virus into also appears to be increased in circulating
the brain (Fig. 1). monocytes in macaques with CNS infection
Although there is general agreement that (Borda et al. 2008). The life span of these peri-
the predominant infected cells are macrophage- vascular macrophages was previously thought
like, there is controversy regarding which of to be days or weeks; recent studies in rhesus
the several subtypes of CNS macrophages are macaques have indicated that they are proba-
harboring HIV (macrophage subtypes are re- bly longer-lived (Soulas et al. 2009). Multi-
viewed by Perry et al. 2010). Most investiga- nucleated giant and other CNS lesions contain-
tors agree that perivascular macrophages, which ing SIV in an experimental model or HIV in

Capillary lumen Infected, activated monocytes Infected, activated HIV virion


CD4+ T cells

Brain microvascular Tight


endothelial cell a junction d
www.perspectivesinmedicine.org

Basement b
membrane
Perivascular
macrophage ?
Astrocyte
e
b

Microglial cell
b

c Multinucleated giant cell

Fusion of infected
and uninfected cells
Brain parenchyma

Figure 1. Potential models for HIV neuroinvasion and infection of the central nervous system (CNS).
(a) HIV-infected monocytes with an activated phenotype may transport HIV into the nervous system via migra-
tion across the blood brain barrier. (b) Infected monocytes likely differentiate into perivascular tissue macro-
phages and proceed to produce HIV within the CNS. This macrophage infection and replication allows for
release of free virions and may facilitate infection of microglial cells. (c) Cell-to-cell fusion involving cells
that express CD4 and HIV coreceptors results in formation of multinucleated giant cells within the brain, a hall-
mark of HIV-related brain pathology. (d) Infected CD4 T lymphocytes may also serve as a mechanism of entry
of HIV into the brain. There is varied evidence regarding the relative contribution of CD4 T lymphocytes ver-
sus cells of the monocyte/macrophage in initiating and sustaining HIV infection within the CNS. (e) Although
astrocytes might harbor HIVand also contribute to HIV-related brain disease through mechanisms of astroglio-
sis induced by local chemokines and cytokines, astrocytes infection is not thought to support ongoing replica-
tion within the CNS. (Adapted from Gonzalez-Scarano et al. 2005; with permission, from Macmillan Publishers
Ltd. # 2005.)

Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120 3


S. Spudich and F. Gonzalez-Scarano

autopsy samples are composed of cells express- in CD14/CD69 positive cells in patients on
ing different macrophage markers, including cART with dementia in comparison to those
CD163, CD68, and CD387; some cells appear without (Kusdra et al. 2002). Similarly, in the
productively infected; others do not express rhesus macaque model of SIV encephalitis
viral antigens (Soulas et al. 2011). Nevertheless, (SIVE) increased monocyte turnover and ex-
although it has previously been suggested that pression of the CD163 marker are associated
because of their rapid turnover perivascular with brain penetration and encephalitic changes
macrophages could not contribute to the long- (Burdo et al. 2010).
term presence of HIV/SIV within the CNS, While these observations were developing,
which could then serve as a reservoir when sys- independent evidence that systemic inflamma-
temic infection was cured, these more recent tion driven by depletion in the gut immunological
discoveries would propose that indeed this peri- system and consequent microbial translocation
vascular population could harbor virus for long had a role in HIV pathogenesis arose from
periods of time. Parenthetically, the astrocytes several areas, including animal models and
found to be infected using sensitive methodol- human observations (see Lackner et al. 2011
ogies are often close to the perivascular spaces. for details). This led to a series of discoveries
Microglia, which are parenchymal, or deep- suggesting that microbial translocation and con-
er within the brain, are known to be long- comitant immunological activation are as-
lived and are replaced only infrequently during sociated with the presence of neurological
an individuals lifetime. Recent studies in mice complications in HIV infections (Ancuta et al.
have indicated that they may represent a sepa- 2008). Furthermore, this finding may explain
rate ontogeny within bone marrow cells (Gin- why there has been a strong correlation between
houx et al. 2010), although hematopoietic inflammatory activity, as characterized by the
www.perspectivesinmedicine.org

circulating cells could potentially also give presence of macrophages expressing activation
rise to morphologically appearing microglial markers in the CNS, and the development of
cells, albeit a small minority. However, there is HAND or HAD. This correlation may be as
some controversy as to whether true microglia strong as that of the presence of viral proteins
are commonly infected by HIV, and some inves- and other evidence of HIV replication.
tigators believe that they are never infected. A model that incorporates current concepts
Multinucleated giant cells, the pathological of systemic pathogenesis and the CNS-specific
hallmark of HIV in the CNS, arise from macro- observations would then propose that infection
phage-type cells, and although their frequent of the CNS is driven by systemic activation of
perivascular location would suggest they are monocytesat least partly owing to microbial
from that perivascular macrophage popula- translocation from a depleted gut immune sys-
tion, there are no markers that can reliably temwhich then are more likely to invade the
confirm this. Furthermore, the life span of mul- brain perivascular space. As some of these cells
tinucleated giant cells is completely unknown, are infected, they bring in virus that spreads
although one can speculate based on their pres- locally and sets up a nidus of replication in-
ence in pathological specimens that they last at dependent from the systemic circulation (see
least days. next section).
Studies performed well over a decade ago Concomitantly, the perivascular inflamma-
first proposed that a subset of monocytes, tion results in the secretion of cytokines and
characterized by expression of the markers chemokines that in turn amplify the reaction,
CD14 and CD69, were particularly prominent attracting in addition other circulating mono-
in patients with HIV neurological complica- cytes and infected CD4 T lymphocytes that
tions and specifically HAD (Pulliam et al. can also add to the CNS viral burden (Xing
1997). More recently, investigators from the et al. 2009). Chief among the chemokines asso-
same group related these original findings to a ciated with HIV infection of the CNS is MCP1
less impressive but still measurable increase (CCL2), which is present in easily measurable

4 Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120


Pathogenesis and Management of Neuro-AIDS

concentrations in the cerebrospinal fluid (CSF) differentiate between genetic divergence owing
and is associated with dementia, but also IP10 to a founder effect and adaptive evolution;
and others (see next sections). even fewer have estimated the timing of diver-
gence in the CNS. Most investigators who have
compared the rate of nonsynonymous to synon-
CNS Compartmentalization
ymous changes have concluded that evolution
Studies in acute HIV infection have shown that in pol, env, or nef in brain isolates is adaptive
virus is present in the CSF at early points during (Huang et al. 2002; Thomas et al. 2007b; Gray
HIV infection, including in some patients with et al. 2011). What is less clear is which specific
primary infection (Schnell et al. 2010); whether pressures are driving the adaptation. Theoreti-
such an early seeding forms the basis for inde- cally, enhanced replication in macrophages
pendent replication in the brain, or whether it elaborated belowresponse to antiretrovirals,
is cleared and virus penetrates at other points or the peculiarities of the immune response
in the course of the infection, has been the within the CNS could be playing a role.
subject of many excellent studies without a clear Chief among the potential selective pres-
consensus (Caragounis et al. 2008; Harrington sures are the requirement for robust replication
et al. 2009). It is likely that different scenarios in macrophages. Macrophages express CD4 at
take place depending on the host, the route of lower levels than CD4 lymphocytes (Lee et al.
infection, and possibly the individual isolates. 1999). Accordingly, HIV isolates from the
Nevertheless, although the details may vary, CNS tend to have an increased capacity to use
long-term HIV infection leads to genetically reduced levels of CD4 for entry and infection
isolated populations in the CNS, as evidenced (Peters et al. 2004; Martn-Garcia et al. 2006;
by studies using pol, env, nef, and other genes Thomas et al. 2007a); however, many of these
www.perspectivesinmedicine.org

(Thomas et al. 2007; Brown et al. 2011; Cowley isolates have been obtained postmortem,
et al. 2011; Gray et al. 2011). The most recent and could reflect an end-stage phenotype.
studies (Brown et al. 2011) have used single Similarly, the CNS is an environment with a rel-
genome analysis (SGA) to overcome potential atively low penetration of antibodies, and con-
PCR artifacts and confirmed the conclusions figurations that might promote neutralization
derived with bulk amplification before the are better tolerated under these circumstances
development of SGA. Additionally, studies in of immunological privilege. In fact, isolates
experimental infection of rhesus macaques from the brain have been shown to be sensitive
with a cloned SIV isolate suggested that virus to neutralizing sera, and particularly to a mono-
is not only compartmentalized in the CNS, clonal antibody (b12) that overlaps the CD4
but that different regions have potentially differ- binding site (van Marle et al. 2002; Martn-
ent env genotypes, setting the stage for inde- Garcia et al 2005; Dunfee et al. 2007).
pendent entry events, some potentially early in Antiretroviral use and its penetration into
the course of infection, some perhaps much the CNS is another potential selective mecha-
later (Chen et al. 2006; Reeve et al. 2010). nism for HIV strains in this regard, and a
Important yet still unresolved questions are few studies have shown discordance between
whether the genetic compartmentalization the resistance phenotypes in the blood and
observed in the CNS is the result of a founder CSF-derived strains (Haas 2004; Smit et al.
effect with concomitant independent diver- 2004). However, the turnover of virus-infected
gence, or whether there are specific selective cells can be different between the plasma and
pressures that promote selection, and what CSF compartments (Schnell et al. 2009) consis-
those selective pressures might be. Most likely tent with the predominantly infected cell type
the end result observed in cross-sectional anal- in each compartmentlymphocytes in the
yses such as the ones cited previously are attrib- circulation, longer-lived macrophages in the
utable to a combination of factors. There have CNSand discordance in the sensitivity to
been comparatively few studies designed to antiretrovirals may be attributable to viral

Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120 5


S. Spudich and F. Gonzalez-Scarano

genetic information being obtained as a snap- proposed that a common end pathway of toxic-
shot rather than representing a true biological ity is through excitation of N-methyl-D-aspar-
phenomenon (Haas 2004). For example, resist- tate (NMDA)-subtype glutamate receptors,
ance genotypes detected in the circulation at which has the potential for mediating apopto-
one point may potentially not appear in the sis. Evidence supporting this model includes
CSF until later because of slower replication experiments that show that decreasing glu-
cycles. tamate secretion by infected macrophages is
partially protective in an in vitro model of
macrophage-mediated neurotoxicity (ODon-
Mechanisms of CNS Injury
nell et al. 2006), that toxicity associated with
The pathophysiology of HAD and HAND must viral proteins is dependent on expression of
eventually involve neurons, the principal effec- these receptors, and that the areas that are
tor cells of the nervous system. To understand most affected are associated with a concentra-
the apparent paradox of cognitive and motor tion of NMDA receptors.
symptoms in what is principally a macrophage An intriguing recent finding, seemingly un-
and microglial infection, investigators have related to HIV, comes from the work of Shau-
developed a number of in vitro and in vivo Kwaun Chen and colleagues. They described a
models, many using combinations of infected mouse mutant in the Hoxb8 gene that has a
monocyte-derived macrophages with mamma- phenotype of excessive grooming (Chen et al.
lian (often rat, but also human) neurons. A 2010). However, in the relevant CNS regions,
somewhat simplistic summary of the vast liter- this gene is normally expressed in microglia
ature on this subject divides the putative neuro- only, and the phenotype could be rescued by
toxic molecules into those that are the direct bone marrow transplantation. This article raises
www.perspectivesinmedicine.org

result of virus in the extracellular fluid, and the possibility that microglia can affect complex
those that propose that neurotoxicity is the behaviors even in the absence of neurodegener-
end result of macrophage and microglial reac- ation, although it is still quite possible that the
tion to a chronic infection with HIV. effects are attributable to aberrant secretion
Among the viral proteins that have been of cytokines. As such, it opens the door for a
implicated in neurotoxicity are gp120, Tat, mechanism of HIVeffect, potentially reversible,
Vpu, and Vpr. Of these, gp120 and Tat have that does not depend on the classic findings of
received the widest attention, and unques- neuronal dropout, and could occur without
tionably they can cause neurotoxicity in vitro. such histopathological changes.
More complicated is the question of whether
the concentrations of proteins that can be
Strain-Specific Neuropathogenesis
achieved in the in vivo extracellular fluid ever
approach the concentrations required to affect A number of studies have related specific HIV
neurons. For gp120, this may be the case, but sequences, primarily in gp120, to the develop-
extracellular concentrations of Tat in the nano- ment of HAND. Among these, those that as-
gram range are difficult to visualize. sociated either tropism in macrophages/mi-
A second pathway of neurotoxicity involves croglia with specific genotypes are the most
the production of potentially neurotoxic fac- worthy of note. For example, position 306 influ-
tors in association with macrophage infection. enced M tropism and CCR5 binding in a sub-
Among the factors implicated in neurotoxic- set of brain-derived isolates (Dunfee et al. 2007).
ity are quinolinic acid, tumor necrosis fac- Similarly, a variant at position 283 (N283) was
tor, platelet activating factor, and arachidonic associated with brain infection and also had en-
acid metabolites. Some of these have been hanced tropism for macrophages. These collec-
detected in the CSF, and related to neurocogni- tive findings suggest that changes that enhance
tive functioning, whereas others have been pri- M tropism are associated with brain infec-
marily tested in vitro. Many investigators have tion, but also that specific mutations are often

6 Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120


Pathogenesis and Management of Neuro-AIDS

context-dependent and that a universal brain 1997; Robertson et al. 1998). However, the
signature is unlikely. CSF HIV RNA may arise from sources other
Similarly, in view of the considerable burden than the CNS, and in addition to the brain,
of HIV infection in Africa as well as other devel- may also reflect virus in the systemic circulation
oping countries, many investigators have exam- that has been transported or leaked to the
ined the prevalence of neurological disorders nervous system. Importantly, genetic compart-
in individuals who are infected with HIV clades mentalization of virus and detection of diver-
other than the clade B that is predominant gent viral quasispecies between these tissues
in the developed world. Those studies are diffi- indicates that CSF HIV is not entirely a spillover
cult, because neurological and psychiatric care from that present in blood (see previous sec-
is suboptimal in developing countries, and tions for references).
because most if not all of the more sophisticated With the recognition that markers of cellu-
instruments that are used to determine CNS lar activation and inflammation were useful
involvement depend on cultural context. Never- indicators of disease activity in systemic HIV
theless, it is clear that HIV strains from clades infection, attention turned to the utility of fol-
other than B are associated with HAND. For lowing such measures in the CNS compart-
example, Mahadevan et al. (2007) studied the ment. Soluble CSF markers of macrophage
brains of patients with clade C infection and activation (neopterin), chemokines stimulating
found evidence of p24 antigen expression in ingress of macrophages and lymphocytes across
macrophage-like cells in patients who had the blood brain barrier (CCL2/MCP1 and
opportunistic infections such as toxoplasmosis. CXCL10/IP10), and molecules involved at var-
The pattern was similar to that of clade B CNS ious stages in the pathways for cell turnover and
infiltration, but there were no multinucleated activation within the nervous system compart-
www.perspectivesinmedicine.org

giant cells. In addition, Sacktor and coworkers ment are used to monitor processes that are
in Uganda (Sacktor et al. 2009) showed that thought to serve as the substrate for neuropa-
dementia occurs in patients with clade D in- thology in HAND (for review, see Cinque
fection, possibly in greater proportion than in et al. 2007). In one small study, moderately ele-
patients with clade A infection in the same vated CSF neopterin predicted subsequent pro-
region. gression to HAD (Brew et al. 1996). However,
although such markers have been correlated to
disease activity, they have not been clinically
Biomarkers of CNS Disease
used for diagnosis or monitoring of Neuro-
As the brain and spinal cord are relatively inac- AIDS owing to lack of specificity for active neu-
cessible for assessment, surrogate biomarkers of rological disease in the setting of the immune
CNS disease may provide some insight into activation characterizing HIV infection (Gisslen
ongoing processes relevant to HIV infection. et al. 2009). Recently, attention has turned to
However, a major problem has been identifying plasma markers related to immunopathogene-
markers that are specific enough and also meas- sis of systemic HIV, including soluble CD14
urable with assays routinely available. and lipopolysaccharide (Ancuta et al. 2008;
Measurement of HIV RNA in the CSF is the Sun et al. 2010). Direct markers of neurological
most practical means of assessing CNS viral injury assayed in CSF, including neurofilament
load. CSF HIV RNA is ubiquitous during light chain protein (NFL), tau protein, and pre-
chronic untreated HIV infection, with levels cursors and products of amyloid protein (amy-
that trend as the levels in blood but typically loid precursor proteins and Ab1-42) may be
are 10-fold lower in absolute terms (Spudich more valuable as measures of active neurode-
et al. 2005; Marra et al. 2007). In untreated pa- generation or injury (Hagberg et al. 2000; Gis-
tients, HIV RNA levels may be higher in patients slen et al. 2007, 2009; Clifford et al. 2009).
with active neurological disease as compared Imaging of the brain has been extensively
with asymptomatic individuals (Brew et al. investigated and used; overt HAD may be

Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120 7


S. Spudich and F. Gonzalez-Scarano

characterized by cerebral atrophy with or with- (Ances et al. 2008, 2010). Detection of abnor-
out periventricular white matter hyperinten- malities in neuroasymptomatic patients under-
sities, which are diffuse, largely symmetric, scores the potential utility of functional MRI as
and not characterized by edema or mass effect assessment before development of overt neuro-
(Price et al. 1991). However, these findings are logical disease. Similar pathology of impaired
neither specific for nor ubiquitous in HAD, blood flow may be detected by simpler cerebral
especially in its earlier stages, and, conversely, perfusion imaging, which may additionally
brain atrophy is noted in many neuroasympto- have a role in the assessment and monitoring
matic HIV-infected patients. Magnetic reso- of HIV-related CNS disease (Ances et al. 2009).
nance spectroscopy (MRS), which detects cel-
lular and biochemical processes based on
Beneficial Effects of Antiretroviral Therapy
diffusion of molecules through cerebral tissues,
in HIV-Associated CNS Disease
has yielded more specific insight into the in-
flammatory and neuronal processes occurring Combination antiretroviral therapy has had a
in the nervous system throughout the course dramatic beneficial impact on the incidence
of HIV infection. Overt HAD is associated and prevalence of severe forms of HAND or
with reduced relative levels of N-acetylaspartate, HAD. The Euro-SIDA cohort study clearly
indicating decreased neuronal function, and showed a decline in incidence of severe demen-
elevated levels of choline, associated with tia (then termed AIDS-dementia complex, or
brain inflammation and membrane turnover ADC), related to introduction of protease
(Meyerhoff et al. 1994; Chang 1995). Similar, inhibitors and use of cART (dArminio Mon-
although less severe, patterns are seen in asymp- forte et al. 2004). More recent evidence from
tomatic, untreated HIV infection, indicating the CHARTER study indicates a greatly reduced
www.perspectivesinmedicine.org

that MRS may be a valuable preclinical marker prevalence (2% overall) of severe HAD in a
of active CNS disease (Meyerhoff et al. 1999). cohort of HIV-infected individuals in the
Although changes in cerebral metabolites may current era (recruited between 2003 and 2007)
indicate regional inflammation and neuronal (Heaton et al. 2010). This improvement in the
injury, more subtle and potentially more neuro- prevalence and incidence of severe HAND
pathologically relevant information may be with cARTreflects the generally beneficial effect
obtained by systematic evaluation of white of initiation of cART on neurocognitive per-
matter tracts or white matter morphometry in formance witnessed in studies of initiation of
the brain by diffusion tensor imaging (DTI). antiretroviral therapy (Marra et al. 2003; Rob-
Some early work in this area suggests that in ertson et al. 2004; Cysique et al. 2006).
neuroasymptomatic HIV, there are reductions What are the biological underpinnings of
in major white matter tracts in a number of this improvement? Blood and CSF viral burden
brain regions (Pomara et al. 2001; Thurnher are clearly reduced by cART (Marra et al. 2003;
et al. 2005; Chang et al. 2008). More global Spudich et al. 2005, 2006), and the initiation of
sophisticated brain morphometry measure- cART is associated with sequential reduction in
ment may be used to detect focal atrophy of HIV RNA levels in both compartments over
gray or white matter structures (Wang et al. time (Ellis et al. 2000). CNS inflammation, the
2009). Functional magnetic resonance imaging putative substrate of ongoing CNS injury in
(fMRI), which takes advantage of the fact that the setting of HIV, is also partly ameliorated
hemodynamics in the brain are closely linked by cART. Treatment is associated with reduced
to neural activity, uses techniques that measure levels of markers of intrathecal inflammation,
cerebral blood flow and blood oxygen level such as cellular markers of T-cell activation,
dependence (BOLD) signals. Early studies in CSF white blood cell (WBC), CSF neopterin,
this area show reduced baseline cerebral blood and b-2 microglobulin (Yilmaz et al. 2004; Spu-
flow and increased functional demand in the dich et al. 2006; Sinclair et al. 2008). Finally,
brain parenchyma in HIV-infected patients markers of active neural injury in the CSF,

8 Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120


Pathogenesis and Management of Neuro-AIDS

including CSF NFL and tau protein are reduced lower, but not negligible, frequency of impair-
in the setting of cARTand have been observed to ment (Table 1) (Heaton et al. 2010).
decay over time in subjects initiating therapy The dramatic change in the severity and per-
(Mellgren et al. 2007). haps phenotype of the clinical disorders associ-
ated with HIV suggests that the etiology of
Neuro-AIDS may have altered during this
Persistent Evidence of HAND despite
time. A small number of studies have focused
Antiretroviral Therapy
on brain pathology in the era since the intro-
HAND is a clinical diagnosis, currently defined duction of cART. Examination of 589 brains
based on abnormal cognitive and motor per- obtained mainly from subjects on antiretroviral
formance on neuropsychological tests accord- therapy available through the National Neuro-
ing to criteria that denote three levels of AIDS Tissue Consortium (NNTC) revealed a
HIV-associated neurological disease: asympto- significant reduction in the proportion of
matic neurocognitive impairment (ANI), mild subjects with typical HIV-related brain pathol-
neurocognitive disorder (MND), and HAD ogy (including encephalitis, microglial nodules,
(Antinori et al. 2007, see Table 1). A number and leukoencephalopathy) compared with
of recent studies have documented persistence the pre-antiretroviral era (Everall et al. 2009;
of neurocognitive abnormalities, predomi- Heaton et al. 2010). However, 78% overall had
nantly along the milder spectrum of HAND, neuropathological abnormalities including vas-
in the setting of cART, with a prevalence rang- cular pathologies, Alzheimer type II gliosis, and
ing between 18% and 52% in varied settings other infectious and noninfectious pathologies.
(Robertson et al. 2007; Heaton et al. 2010; Cy- Premorbid HAND diagnosis (in 82%) in this
sique and Brew 2011). Studies including subjects cohort did not correlate with HIV-related brain
www.perspectivesinmedicine.org

with comorbidities that potentially confound pathology.


the diagnosis of HAND, including current Although classical HIV-related brain path-
or past substance abuse, mental health dis- ology may no longer be the only substrate for
orders, head trauma, low education level, and HAND, HIV-driven mechanisms may still be
coinfection with hepatitis, find a higher preva- important CNS abnormalities in the setting of
lence (up to 83% HAND in the highest comor- cART. Real-time PCR quantification of HIV
bidity group in CHARTER), whereas those RNA in autopsy specimens from the NNTC
excluding subjects with significant relevant has revealed detectable HIV-1 RNA in brain in
comorbidities and with low CD4 nadirs had a majority of cART-treated subjects (Kumar

Table 1. Diagnostic research criteria for HAND


Diagnostic entity Cognitive performance Functional status
Normal Normal Normal
Asymptomatic Acquired impairment in at least two No perceived impact on daily function
neurocognitive cognitive domains (,1 SD)
impairment (ANI)
Mild neurocognitive Acquired impairment in at least two Perceived interference with daily
disorder (MND) cognitive domains (,1 SD) function to at least a mild degree
(work inefficiency, reduced mental
acuity)
HIV-associated dementia Acquired impairment in at least two Marked impact on daily function
(HAD) domains, typically in multiple domains
with at least two domains with severe
impairment (,2 SD)
Adapted from Antinori et al. 2007; with permission, from Wolters Kluwer Health # 2007.

Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120 9


S. Spudich and F. Gonzalez-Scarano

et al. 2007), and abnormal levels of microglial readily cross the bloodbrain barrier. Based on
activation in the CNS were found to persist structural composition and effect on CSF HIV
in neuroasymptomatic subjects with successful RNA, antiretrovirals have been ranked accord-
plasma viral suppression on cART in another ing to their estimated CNS penetration effec-
recent neuropathological study (Anthony 2005). tiveness (CPE), with a combination of drugs
These studies are corroborated by evidence in a regimen assigned a combined CPE score
that intrathecal inflammation as detected in based on the sum of their individual rankings
CSF persists in the setting of long-term, sys- (Table 2) (Letendre et al. 2008). Given that treat-
temically effective cART (Eden et al. 2007; ment with cART benefits the nervous system in
Yilmaz et al. 2008). Although even in the set- terms of both the detection and magnitude of
ting of therapy that fails to successfully sup- HIV infection and attendant inflammation
press plasma HIV RNA (Spudich et al. 2006), within the CNS, more potent activity of antire-
cART usually successfully suppresses CSF HIV troviral medications within the CNS might pro-
RNA below detectable levels, viral escape vide additional benefit in the setting of
in the CSF may occur in up to 10% of individ- treatment of HAND. Despite predominant con-
uals on current regimens (Eden et al. 2010), currence between numerous studies that regi-
and in rare cases dramatic CSF escape has mens with higher CPE scores tend to lead to
been associated with clinically progressive more successful suppression of HIV RNA levels
neurological disease (Canestri et al. 2010). in the CNS (Letendre et al. 2008; Marra et al.
2009), the evidence that enhanced CPE scores
are related to improved neurocognitive out-
CNS Penetration of cART
comes in subjects with HIV infection is less
The CNS is separated from the systemic circula- definitive. Whereas some observational studies
www.perspectivesinmedicine.org

tion by blood brain and blood CSF barriers, show a cognitive benefit of initiation of regi-
thus potentially allowing for a sanctuary of mens with higher CPE scores in the setting of
infection that is only partially reached by HIV-related CNS disease (Letendre et al. 2004;
some antiretroviral medications which, owing Tozzi et al. 2009), others show poorer neuro-
to molecular size or hydrophilicity, do not cognitive performance in subjects treated with

Table 2. Antiretroviral central nervous system penetration effectiveness (CPE) scoring system used in recent
clinical studies
Increasing CNS penetration ! 0 0.5 1
Nucleoside reverse transcriptase inhibitors Didanosine Emtricitabine Abacavir
Tenofovir Lamivudine Zidovudine
Zalcitabine Stavudine
Adefovir
Nonnucleoside reverse transcriptase inhibitors Efavirenz Delavirdine
Nevirapine
Protease inhibitors Nelfinavir Amprenavir Amprenavir/r
Ritonavir Atazanavir Atazanavir/r
Saquinavir Fosamprenavir Fosamprenavir/r
Saquinavir/r Indinavir Indinavir/r
Tipranavir/r Lopinavir/r
Entry inhibitors Enfuvirtide Maraviroc
T-1249 Vicriviroc
Integrase inhibitors Raltegravir
Elvitegravir
Adapted from Smurzynski et al. 2011; with permission, from Wolters Kluwer Health # 2011.

10 Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120


Pathogenesis and Management of Neuro-AIDS

100 p = 0.0004 p = 0.0001 persist despite high CPE regimens. A recent study
did find a modest neurocognitive benefit of
higher CPE scores (although only between
p = 0.72 patients with more than three drugs in their reg-
75
Avg. % difference imens and higher or lower CPE scores) in the
context of randomized assignment of antiretrovi-
ral regimens (Smurzynski et al. 2011); addi-
50 tional randomized studies are warranted to
directly examine this issue.

25
New Research Areas
The Relationship among Aging, New
0 Comorbidities, and HAND
PI NI MCMD/I HAD
In resource-rich regions where life expectancy
Figure 2. Heterduplex tracking assay (HTA) was used after HIV diagnosis in a young adult is esti-
to compare V1/V2 and V4/V5 env populations in mated to be approximately 12 years below that
blood plasma (BP) and CSF from subjects with
of a noninfected individual (Lohse et al. 2007),
distinct stages of HIV infection and neurological
status (PI, primary infection; NI, not impaired; longer duration of survival with chronic
MCMD/I, minor cognitive and motor disorder; HIV-1 infection may change the scope and
HAD, HIV-associated dementia). The % difference etiology of disorders affecting the CNS. A great
for BP/CSF HTA band patterns was determined for deal of recent attention has been paid to
www.perspectivesinmedicine.org

each patient and the data compiled. A higher percent- the question of whether aging and HIV-1 will
age difference indicates more discordant BP/CSF have a synergistic effect on neurodegeneration
viral genetic populations for the particular region of
within the brain, accelerating injury triggered
env analyzed. The mean of V1/V2 and V4/V5 percent
difference results was determined for each patient to earlier by neuropathological processes associ-
reflect global env compartmentalization between BP ated with HIV. Systemic immunologic changes
and CSF, and results were compiled for comparison noted in HIV infection are in part characterized
between the different disease categories. p values by markers of immunosenesence, and chronic
shown were determined by Wilcoxon rank-sum test. infection with HIV is associated with the earlier
(Adapted from Harrington et al. 2009; with permis- onset of cancers and vascular disorders seen in
sion, from Wolters Kluwer Health # 2009.)
HIV-uninfected persons at more advanced age
(Desai and Landay 2010). However, the evi-
dence that aging and HIV have an enhanced
regimens with higher CPE scores (Marra et al. combined deleterious effect in the brain is
2009). A variety of issues may lead to the mixed. A large cohort study enrolling subjects
discrepant results of such studies. High CPE with HIV and advanced age, the Hawaii aging
regimens may be especially important in condi- cohort, found a higher prevalence of cognitive
tions of robust brain and meningeal infection deficits in older HIV-infected patients, related
with HIV, which typically occur in later stages to CD4 nadir, insulin resistance, and presence
of chronic infection, when targeted therapy to of Apo E alleles (Valcour et al. 2004), and a re-
CNS tissues may be key for control of compart- cent neuroimaging study found that HIV and
mentalized viral replication (Fig. 2). Further- aging caused independent reductions in cere-
more, retrospective, observational studies may bral blood flow (Ances et al. 2010). However,
be biased; for example, subjects with more pro- the Multicenter AIDS Cohort Study (MACS)
found neurological deficits may be started on found no difference between older HIV indi-
regimens with higher CNS penetration. Such viduals and older HIV2 individuals in rate of
deficits may not be entirely reversible and thus change in performance on longitudinal testing,

Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120 11


S. Spudich and F. Gonzalez-Scarano

suggesting the absence of an acceleration of less than three months after HIV transmission
neurological disease with aging (Becker et al. revealed elevations in CSF WBC counts, neo-
2009). Two additional recent studies using well- pterin, and CXCL10 equal to those in subjects
matched HIV-uninfected comparison groups in with chronic HIV infection (Fig. 3) (Spudich
analyses of age effects on neurocognitive per- et al. 2011).
formance showed no combined effects of HIV Furthermore, neuroimaging studies in re-
and age on cognitive function (Cysique et al. cently HIV-infected humans reveal lower n-
2011; Valcour et al. 2011). Other conditions acetylacetate in the frontal cortex during early
are emerging in the setting of long-term infection, suggestive of neuronal dysfunction
survival with chronic HIV infection that may or injury during this early period (Lentz et al.
influence the integrity of the CNS and alter 2009). Follow-up longitudinal studies in this
the substrate for cognitive and neurological group reveal dynamic patterns of cerebral me-
impairment in HIV. In particular, cardiovascu- tabolites over the first year of infection (Lentz
lar risk factors such as hyperlipidemia, hy- et al. 2011). Given that early HIV infection
pertension, carotid intima-media thickness, is characterized by neuroinflammation and
and past history of cardiovascular disease have evolving metabolite changes within the CNS,
been identified recently as associated with it is possible that either antiretroviral or anti-
reduced neurocognitive performance in HIV- inflammatory treatment initiated during early
infected subjects (Becker et al. 2009; Foley stages of infection may ameliorate injury sus-
et al. 2010; Wright et al 2010). It is unclear tained in the CNS during the early years of
whether these risk factors are non-HIV-related infection before immune systemic immuno-
cofactors that influence cognitive outcomes, suppression. Finally, the fact that CSF compart-
HIV- or cART-related systemic effects that mentalization of HIV species may begin within
www.perspectivesinmedicine.org

parallel processes of CNS injury, or conditions the first year of HIV infection in some individ-
that are directly involved in the mechanisms uals (Schnell et al. 2010) suggests that treatment
of HAND in the current era. Further efforts and eradication efforts may need to consider the
to investigate the association between vascular CNS as a potential independent site of replica-
changes and clinical, pathological, and imag- tion and mutation, beginning in the early stages
ing changes in the setting of HIV are warranted of HIV.
to expand possible treatment approaches to
HAND.
CONCLUSIONS
There has been substantial progress in the rec-
Importance of Acute/Early Infection
ognition and treatment of the most severe forms
Although severe HAD clearly is a condition of CNS HIV infection, and although the mech-
associated with long-term chronic HIV infec- anisms leading to neurological dysfunction are
tion and immunosuppression, recent evidence still under investigation, the central role of mac-
that acute and early HIVare crucial for systemic rophages and microglia is well established. Less
disease pathogenesis has raised questions about well understood are the less severe forms of CNS
whether early stages of HIV might also be disease now seen in the developed countries
important for neuropathogenesis. It has long where the use of cART is commonand the
been known that HIV may enter the nervous role of virus, inflammation, and CNS pene-
system within the first weeks after initial trance of antiretrovirals are areas of potential
systemic infection (Schacker et al. 1996; Pilcher new discoveries. Similarly, there has not been
et al. 2001). It is now clear that acute infection any successful adjuvant therapy: that is, one
also initiates a cascade of neuroinflammation, designed to treat the CNS specifically rather
providing conditions for inflammation-medi- than the virus. Such adjuvant treatment may
ated injury within the CNS. Analysis of CSF be important in preventing or ameliorating
from 96 antiretroviral nave subjects at a median HAND in the setting of cART.

12 Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120


Pathogenesis and Management of Neuro-AIDS

A Plasma HIV RNA level B CSF HIV RNA level


8 8
r 2 = 0.047 r 2 = 0.03
7 p = 0.05 7 p = 0.12
6 6

log 10 c/mL

log 10 c/mL
5 5
4 4
3 3
2 2
1 1
0 100 200 300 0 100 200 300

C CSF WBC D CSF neopterin


150 100
100 r 2 = 0.0128 r 2 = 0.10
50 p = 0.21 p = 0.0097
80
40
cells/mm3

60

nmol/L
30
40
20

10 20

0 0
www.perspectivesinmedicine.org

0 100 200 300 0 100 200 300


Days post HIV transmission Days post HIV transmission

Figure 3. Baseline levels of markers of HIV infection and inflammation in a primary infection cohort are shown
according to estimated days after transmission at blood and CSF sampling. Simple linear regression between the
number of days post-estimated HIV transmission and (A) blood plasma HIV RNA levels, (B) CSF HIV RNA
levels, (C ) CSF WBC counts, and (D) CSF neopterin levels. Regression lines (solid) and 95% confidence inter-
vals (dotted) are indicated; upper limit of normal values for CSF WBC and CSF neopterin are indicated on each
graph by a dotted horizontal line. (Adapted from Spudich et al. 2011; with permission, from Oxford University
Press # 2011.)

ACKNOWLEDGMENTS blood oxygen level-dependent amplitude in human


immunodeficiency virus-seropositive patients. J Neuro-
We thank Dr. Anna Aldovini for her helpful virol 14: 418 424.
review of this article. We also thank the National Ances BM, Sisti D, Vaida F, Liang CL, Leontiev O, Perthen
JE, Buxton RB, Benson D, Smith DM, Little SJ, et al.
Institutes of Health for funding support
2009. Resting cerebral blood flow: A potential biomarker
(P50NS027405 and R01MH081772). of the effects of HIV in the brain. Neurology 73: 702 708.
Ances BM, Vaida F, Yeh MJ, Liang CL, Buxton RB, Letendre
S, McCutchan JA, Ellis RJ.2010. HIV infection and aging
independently affect brain function as measured by func-
REFERENCES tional magnetic resonance imaging. J Infect Dis 201:
 Reference is also in this collection. 336 340.
Anthony IC, Ramage SN, Carnie FW, Simmonds P, Bell JE.
Ancuta P, Kamat A, Kunstman KJ, Kim EY, Autissier P, Wur- 2005. Influence of HAART on HIV-related CNS disease
cel A, Zaman T, Stone D, Mefford M, Morgello S, et al. and neuroinflammation. J Neuropathol Exp Neurol 64:
2008. Microbial translocation is associated with increased 529 536.
monocyte activation and dementia in AIDS patients. Antinori A, Arendt G, Becker JT, Brew BJ, Byrd DA, Cherner
PLoS One 3: e2516. doi: 10.1371/journal.pone.0002516. M, Clifford DB, Cinque P, Epstein LG, Goodkin K, et al.
Ances BM, Roc AC, Korczykowski M, Wolf RL, Kolson DL. 2007. Updated research nosology for HIV-associated
2008. Combination antiretroviral therapy modulates the neurocognitive disorders. Neurology 69: 17891799.

Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120 13


S. Spudich and F. Gonzalez-Scarano

Becker JT, Kingsley L, Mullen J, Cohen B, Martin E, Miller Cinque P, Brew BJ, Gisslen M, Hagberg L, Price RW. 2007.
EN, Ragin A, Sacktor N, Selnes OA, Visscher BR. 2009. Cerebrospinal fluid markers in central nervous system
Vascular risk factors, HIV serostatus, and cognitive HIV infection and AIDS dementia complex. Handb
dysfunction in gay and bisexual men. Neurology 73: Clin Neurol 85: 261 300.
1292 1299. Clifford DB, Fagan AM, Holtzman DM, Morris JC, Teshome
Borda JT, Alvarez X, Mohan M, Hasegawa A, Bernardino A, M, Shah AR, Kauwe JS. 2009. CSF biomarkers of Alz-
Jean S, Aye P, Lackner AA. 2008. CD163, a marker of heimer disease in HIV-associated neurologic disease.
perivascular macrophages, is up-regulated by microglia Neurology 73: 19821987.
in simian immunodeficiency virus encephalitis after Collman R, Balliet JW, Gregory SA, Friedman H, Kolson DL,
haptoglobin-hemoglobin complex stimulation and is Nathanson N, Srinivasan A. 1992. An infectious molecu-
suggestive of breakdown of the blood-brain barrier. lar clone of an unusual macrophage-tropic and highly
Am J Pathol 172: 725 737. cytopathic strain of human immunodeficiency virus
Brew BJ, Dunbar N, Pemberton L, Kaldor J. 1996. Predictive type 1. J Virol 66: 75177521.
markers of AIDS dementia complex: CD4 cell count and Cowley D, Gray LR, Wesselingh SL, Gorry PR, Churchill MJ.
cerebrospinal fluid concentrations of b 2-microglobulin 2011. Genetic and functional heterogeneity of CNS-
and neopterin. J Infect Dis 174: 294 298. derived tat alleles from patients with HIV-associated
Brew BJ, Pemberton L, Cunningham P, Law MG. 1997. dementia. J Neurovirol 17: 7081.
Levels of human immunodeficiency virus type 1 RNA Cysique LA, Brew BJ. 2011. Prevalence of non-confounded
in cerebrospinal fluid correlate with AIDS dementia HIV-associated neurocognitive impairment in the con-
stage. J Infect Dis 175: 963 966. text of plasma HIV RNA suppression. J Neurovirol 17:
Brown RJ, Peters PJ, Caron C, Gonzalez-Perez MP, Stones L, 176 183.
Ankghuambom C, Pondei K, McClure CP, Alemnji G, Cysique LA, Maruff P, Brew BJ. 2006. Variable benefit in neu-
Taylor S, et al. 2011. Inter-compartment recombination ropsychological function in HIV-infected HAART-
of HIV-1 contributes to env intra-host diversity and treated patients. Neurology 66: 14471450.
modulates viral tropism and senstivity to entry inhibi-
tors. J Virol 85: 60246037. Cysique LA, Maruff P, Bain MP, Wright E, Brew BJ. 2011.
HIV and age do not substantially interact in HIV-associ-
Burdo TH, Soulas C, Orzechowski K, Button J, Krishnan A,
ated neurocognitive impairment. J Neuropsych Clin Neu-
Sugimoto C, Alvarez X, Kuroda MJ, Williams KC. 2010.
rosci 23: 8389.
Increased monocyted turnover from bone marrow corre-
www.perspectivesinmedicine.org

lates with severity of SIV encephalitis and CD163 levels dArminio Monforte A, Cinque P, Mocroft A, Goebel FD,
in plasma. PLoS Pathog 6: e1000842. doi: 10.1371/ Antunes F, Katlama C, Justesen US, Vella S, Kirk O,
journal.ppat.1000842. Lundgren J. 2004. Changing incidence of central nervous
system diseases in the EuroSIDA cohort. Ann Neurol 55:
Canestri A, Lescure FX, Jaureguiberry S, Moulignier A,
320 328.
Amiel C, Marcelin AG, Peytavin G, Tubiana R, Pialoux
G, Katlama C. 2010. Discordance between cerebral spinal Desai S, Landay A. 2010. Early immune senescence in HIV
fluid and plasma HIV replication in patients with neuro- disease. Curr HIV/AIDS Rep 7: 4 10.
logical symptoms who are receiving suppressive antire- Dunfee RL, Thomas ER, Wang J, Kunstman K, Wolinsky
troviral therapy. Clin Infect Dis 50: 773 778. SM, Gabuzda D. 2007. Loss of the N-linked glycosylation
Caragounis EC, Gisslen M, Lindh M, Nordborg C, Wester- site at position 386 in the HIVenvelope V4 region enhan-
gren S, Hagberg L, Svennerholm B. 2008. Comparison ces macrophage tropism and is associated with dementia.
of HIV-1 pol and env sequences of blood, CSF, brain Virology 367: 222234.
and spleen isolates collected ante-mortem and post- Dunfee RL, Thomas ER, Gabuzda D. 2009. Enhanced mac-
mortem. Acta Neurol Scand 117: 108116. rophage tropism of HIV in brain and lymphoid tissues is
Chang L. 1995. In vivo magnetic resonance spectroscopy in associated with sensitivity to the broadly neutralizing
HIV and HIV-related brain diseases. Rev Neurosci 6: CD4 binding site antibody b12. Retrovirology 6: 69.
365378. Eden A, Price RW, Spudich S, Fuchs D, Hagberg L, Gisslen
Chang L, Wong V, Nakama H, Watters M, Ramones D, M. 2007. Immune activation of the central nervous sys-
Miller EN, Cloak C, Ernst T. 2008. Greater than tem is still present after .4 years of effective highly active
age-related changes in brain diffusion of HIV patients antiretroviral therapy. J Infect Dis 196: 17791783.
after 1 year. J Neuroimmune Pharmacol 3: 265 274. Eden A, Fuchs D, Hagberg L, Nilsson S, Spudich S, Svenner-
Chen MF, Westmoreland S, Ryzhova EV, Martn-Garca J, holm B, Price RW, Gisslen M. 2010. HIV-1 viral escape
Soldan SS, Lackner A, Gonzalez-Scarano F. 2006. Simian in cerebrospinal fluid of subjects on suppressive antire-
immunodeficiency virus envelope compartmentalizes in troviral treatment. J Infect Dis 202: 1819 1825.
brain regions independent of neuropathology. J Neuro- Ellis RJ, Gamst AC, Capparelli E, Spector SA, Hsia K, Wolf-
virol 12: 73 89. son T, Abramson I, Grant I, McCutchan JA. 2000. Cere-
Chen SK, Tvrdik P, Peden E, Cho S, Wu S, Spangrude G, brospinal fluid HIV RNA originates from both local
Capecchi MR. 2010. Hematopoietic origin of pathologi- CNS and systemic sources. Neurology 54: 927 936.
cal grooming in Hoxb8 mutant mice. Cell 141: 775 785. Everall I, Vaida F, Khanlou N, Lazzaretto D, Achim C, Leten-
Churchill MJ, Wesslingh SL, Cowley D, Pardo CA, McArthur dre S, Moore D, Ellis R, Cherne M, Gelman B, et al. 2009.
JC, Brew BJ, Gorry PR. 2009. Extensive astrocyte infec- Cliniconeuropathologic correlates of human immunode-
tion is prominent in HIV-associated dementia. Ann Neu- ficiency virus in the era of antiretroviral therapy. J Neuro-
rology 66: 253258. virol 15: 1 11.

14 Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120


Pathogenesis and Management of Neuro-AIDS

Foley J, Ettenhofer M, Wright MJ, Siddiqi I, Choi M, Kim W-K, Alvarez X, Fisher J, Bronfin B, Westmoreland W,
Thames AD, Mason K, Castellon S, Hinkin CH. Neuro- McLaurin J, Williams K. 2006. CD163 identifies perivas-
cognitive functioning in HIV-1 infection: Effects of cere- cular macrophages in normal and viral encephalitic
brovascular risk factors and age. Clin Neuropsychol 24: brains and potential precursors to perivascular macro-
265285. phages in blood. Am J Pathol 168: 822834.
Gabuzda DH, Ho DD, de la Monte SM, Hirsch MS, Rota TR, Kumar AM, Borodowsky I, Fernandez B, Gonzalez L, Kumar
Sobel RA. 1986. Immunohistochemical identification of M. 2007. Human immunodeficiency virus type 1 RNA
HTLV-III antigen in brains of patients with AIDS. Ann levels in different regions of human brain: Quantification
Neurol 20: 289295. using real-time reverse transcriptase-polymerase chain
Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, reaction. J Neurovirol 13: 210 224.
Mehler MF, Conway SJ, Ng LG, Stanley ER, Merad M, Kusdra L, McGuire D, Pulliam L. 2002. Changes in mono-
et al. 2010. Fate mapping reveals that adult microglia cyte/macrophage neurotoxicity in the era of HAART:
derive from primitive macrophages. Science 330: Implications for HIV-associated dementia. AIDS 16:
841845. 31 38.
Gisslen M, Hagberg L, Brew BJ, Cinque P, Price RW, Ros-  Lackner AA, Lederman MM, Rodriguez B. 2011. HIV
engren L. 2007. Elevated cerebrospinal fluid neurofilament pathogenesisThe host. Cold Spring Harb Perspect Med
light protein concentrations predict the development of doi: 10.1101/cshperspect.a007005.
AIDS dementia complex. J Infect Dis 195: 17741778. Lee B, Sharron M, Montaner LJ, Weissman D, Doms RW.
Gisslen M, Hagberg L, Cinque P, Brew B, Price R. 2008. CSF 1999. Quantification of CD4, CCR5, and CXCR4 levels
markers in the management of CNS HIV infection and on lymphocyte subsets, dendritic cells, and differentially
the AIDS dementia complex. In The spectrum of neuro- conditioned monocyte-derived macrophages. Proc Natl
AIDS disorders: Pathophysiology, diagnosis, and treatment Acad Sci 96: 52155220.
(ed. Goodkin K, Shapshak P, Vrma A), pp. 173 179. Lentz MR, Kim WK, Lee V, Bazner S, Halpern EF, Venna N,
American Society for Microbiology, Washington, DC. Williams K, Rosenberg ES, Gonzalez RG. 2009. Changes
Gisslen M, Krut J, Andreasson U, Blennow K, Cinque P, in MRS neuronal markers and T cell phenotypes
Brew BJ, Spudich S, Hagberg L, Rosengren L, Price RW, observed during early HIV infection. Neurology 72:
et al. 2009. Amyloid and tau cerebrospinal fluid bio- 14651472.
markers in HIV infection. BMC Neurol 9: 63. Lentz MR, Kim WK, Kim H, Soulas C, Lee V, Venna N,
Gray LR, Gabuzda D, Cowley D, Ellett A, Chiavaroli L, Wes- Halpern EF, Rosenberg ES, Williams K, Gonzalez RG.
www.perspectivesinmedicine.org

selingh SL, Churchill MJ, Gorry PR. 2011. CD4 and MHC 2011. Alterations in brain metabolism during the first
class 1 down-modulation activities of nef alleles from year of HIV infection. J Neurovirol 17: 220 229
brain- and lymphoid tissue-derived primary HIV-1 iso- Letendre SL, McCutchan JA, Childers ME, Woods SP,
lates. J Neurovirol 17: 8291. Lazzaretto D, Heaton RK, Grant I, Ellis RJ. 2004. Enhanc-
Gonzalez-Scarano F, Martin-Garcia J. 2005. The neuropa- ing antiretroviral therapy for human immunodeficiency
thogenesis of AIDS. Nat Rev Immunol 5: 69 81. virus cognitive disorders. Ann Neurol 56: 416423.
Haas DW. 2004. Sequence heterogeneity and viral dynamics Letendre S, Marquie-Beck J, Capparelli E, Best B, Clifford D,
in cerebrospinal fluid and plasma during antiretroviral Collier AC, Gelman BB, McArthur JC, McCutchan JA,
therapy. J Neurovirol 10 Suppl 1: 33 37. Morgello S, et al. 2008. Validation of the CNS penetra-
Haase AT. 1986. Pathogenesis of lentivirus infections. Nature tion-effectiveness rank for quantifying antiretroviral pen-
322: 130 136. etration into the central nervous system. Arch Neurol 65:
Hagberg L, Fuchs D, Rosengren L, Gisslen M. 2000. Intrathe- 65 70.
cal immune activation is associated with cerebrospinal Lohse N, Hansen AB, Pedersen G, Kronborg G, Gerstoft J,
fluid markers of neuronal destruction in AIDS patients. Sorensen HT, Vaeth M, Obel N. 2007. Survival of persons
J Neuroimmunol 102: 5155. with and without HIV infection in Denmark, 1995
Harrington PR, Schnell G, Letendre SL, Ritola K, Robertson 2005. Ann Intern Med 146: 87 95.
K, Hall C, Burch CL, Jabara CB, Moore DT, Ellis RJ, et al. Mahadevan A, Shankar SK, Parthasarathy S, Ranga U,
2009. Cross-sectional characterization of HIV-1 env Chickabasaviah YT, Santosh V, Vasanthapuram R, Pardo
compartmentalization in cerebrospinal fluid over the CA, Nath A, Zink MC. 2007. Characterization of HIV
full disease course. AIDS 23: 907 915. infected cells in infiltrates associated with CNS oppor-
Heaton RK, Clifford DB, Franklin DR Jr, Woods SP, Ake C, tunistic infections in patients with HIV clade C infection.
Vaida F, Ellis RJ, Letendre SL, Marcotte TD, Atkinson JH, J Neuropathol Exp Neurol 66: 799 808.
et al. 2010. HIV-associated neurocognitive disorders per- Marra CM, Lockhart D, Zunt JR, Perrin M, Coombs RW,
sist in the era of potent antiretroviral therapy: CHARTER Collier AC. 2003. Changes in CSF and plasma HIV-1
study. Neurology 75: 20872096. RNA and cognition after starting potent antiretroviral
Huang KJ, Alter GM, Wooley DP. 2002. The reverse tran- therapy. Neurology 60: 13881390.
scriptase sequence of human immunodeficiency virus Marra CM, Maxwell CL, Collier AC, Robertson KR, Imrie
type 1 is under positive evolutionary selection within A. 2007. Interpreting cerebrospinal fluid pleocytosis in
the central nervous system. J Neurovirol 8: 281 94. HIV in the era of potent antiretroviral therapy. BMC
Hughes ES, Bell JE, Simmonds P. 1997. Investigation of the Infect Dis 7: 37.
dynamics of the spread of HIV to brain and other tissues Marra CM, Zhao Y, Clifford DB, Letendre S, Evans S, Henry
by evolutionary analysis of sequences from the p17gag K, Ellis RJ, Rodriguez B, Coombs RW, Schifitto G, et al.
and env genes. J Virol 71: 12721280. 2009. Impact of combination antiretroviral therapy on

Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120 15


S. Spudich and F. Gonzalez-Scarano

cerebrospinal fluid HIV RNA and neurocognitive per- Reeve AB, Pearce NC, Patel K, Augustus KV, Novembre FJ.
formance. AIDS 23: 1359 1366. 2010. Neuropathogenic SIVsmmFGb genetic diversity
Martn-Garca J, Cocklin S, Chaiken IM, Gonzalez-Scarano and selection-induced tissue-specific compartmentaliza-
F. 2005. Interaction with CD4 and antibodies to CD4- tion during chronic infection and temporal evolution of
induced epitopes of the envelope gp120 from a micro- viral genes in lymphoid tissues and regions of the central
glia-adapted human immunodeficiency virus type 1 nervous system. AIDS Res Hum Retroviruses 26: 663 679.
isolate. J Virol 79, 67036713. Robertson K, Fiskus S, Kapoor C, Robertson W, Schneider
Martn-Garca J, Cao W, Varela-Rohena A, Plassmeyer ML, G, Shepard R, Howe L, Silva S, Hall C. 1998. CSF, plasma
Gonzalez-Scarano F. 2006. HIV-1 tropism for the central viral load and HIV associated dementia. J Neurovirol 4:
nervous system: Brain-derived envelope glycoproteins 90 94.
with lower CD4-dependence and reduced sensitivity to Robertson KR, Robertson WT, Ford S, Watson D, Fiscus S,
a fusion inhibitor. Virology 346: 169 179. Harp AG, Hall CD. 2004. Highly active antiretroviral
Mellgren A, Price RW, Hagberg L, Rosengren L, Brew BJ, therapy improves neurocognitive functioning. J Acquir
Gisslen M. 2007. Antiretroviral treatment reduces Immune Defic Syndr 36: 562566.
increased CSF neurofilament protein (NFL) in HIV-1 Robertson KR, Smurzynski M, Parsons TD, Wu K, Bosch RJ,
infection. Neurology 69: 1536 1541. Wu J, McArthur JC, Collier AC, Evans SR, Ellis RJ. 2007.
Meyerhoff DJ, MacKay S, Poole N, Dillon WP, Weiner MW, The prevalence and incidence of neurocognitive impair-
Fein G. 1994. N-Acetylaspartate reductions measured by ment in the HAART era. AIDS 21: 19151921.
1H MRSI in cognitively impaired HIV-seropositive indi- Sacktor N, Nakasujja N, Skolasky RL, Rezapour M, Robert-
viduals. Magn Reson Imaging 12: 653 659. son K, Musisi S, Katabira E, Ronald A, Clifford DB,
Meyerhoff DJ, Bloomer C, Cardenas V, Norman D, Weiner Laeyendecker O, et al. 2009. HIV subtype D is associated
MW, Fein G. 1999. Elevated subcortical choline metabo- with dementia, compared with subtype A, in immuno-
lites in cognitively and clinically asymptomatic HIV suppressed individuals at risk of cognitive impairment
patients. Neurology 52: 9951003. in Kampala, Uganda. Clinical Infect Dis 49: 780786.
Navia BA, Cho ES, Petito CK, Price RW. 1986a. The AIDS Shau-Kwaun C, Tvrdik P, Peden E, Cho S, Wu S, Spangrude
dementia complex: II. Neuropathology. Ann Neurol 19: G, Capecchi MR. 2010. Hematopoietic origin of patho-
525535. logical grooming in Hoxb8 mutant mice. Cell 141:
Navia BA, Jordan BD, Price RW. 1986b. The AIDS dementia 775 785.
www.perspectivesinmedicine.org

complex: I. Clinical features. Ann Neurol 19: 517 524. Schacker T, Collier AC, Hughes J, Shea T, Corey L. 1996.
ODonnell LA, Agrawal A, Jordan-Sciutto KL, Dichter MA, Clinical and epidemiologic features of primary HIV in-
Lynch DR, Kolson DL. 2006. Human immunodeficiency fection. Ann Intern Med 125: 257264.
virus (HIV)-induced neurotoxicity: Roles for the NMDA Schnell G, Spudich S, Harrington P, Price RW, Swanstrom R.
receptor subtypes. J Neurosci 26: 981 990. 2009. Compartmentalized HIV-1 originates from long-
Perry H, Nicoll JAR, Holmes C. 2010. Microglia in neurode- lived cells in some subjects with HIV-1-associated de-
generative disease. Nat Rev Neurol 6: 193 201. mentia. PLoS Pathog 5: e1000395. doi: 10.1371/journal.
Peters PJ, Bhattacharya J, Hibbitts S, Dittmar MT, Simmons ppat.1000395.
G, Bell J, Simmonds P, Clapham PR. 2004. Biological Schnell G, Price RW, Swanstrom R, Spudich S. 2010. Com-
analysis of human immunodeficiency virus type 1 R5 partmentalization and clonal amplification of HIV-1 var-
envelopes amplified from brain and lymph node tissues iants in the cerebrospinal fluid during primary infection.
of AIDS patients with neuropathology reveals two dis- J Virol 84: 23952407.
tinct tropism phenotypes and identifies envelopes in Shieh JTC, Martn J, Baltuch G, Malim MH, Gonzalez-
the brain that confer an enhanced tropism and fusigenic- Scarano F. 2000. Determinants of syncytia formation
ity for macrophages. J Virol 78: 69156926. in microglia by the human immunodeficiency virus
Pilcher CD, Shugars DC, Fiscus SA, Miller WC, Menezes P, type 1 (HIV-1): Role of the V1/V2 domains. J Virol
Giner J, Dean B, Robertson K, Hart CE, Lennox JL, et al. 74: 693701.
2001. HIV in body fluids during primary HIV infection: Sinclair E, Ronquillo R, Lollo N, Deeks SG, Hunt P, Yian-
Implications for pathogenesis, treatment and public noutsos CT, Spudich S, Price RW. 2008. Antiretroviral
health. Aids 15: 837 845. treatment effect on immune activation reduces cerebro-
Pillai SK, Pond SL, Liu Y, Good BM, Strain MC, Ellis RJ, spinal fluid HIV-1 infection. J Acquir Immune Defic Syndr
Letendre S, Smith DM, Gunthard HF, Grant I, et al. 47: 544 552.
2006. Genetic attributes of cerebrospinal fluid-derived Smit TK, Brew BJ, Tourtellotte W, Morgello S, Gelman BB,
HIV-1 env. Brain 129 (Pt 7): 18721883. Saksena NK. 2004. Independent evolution of human im-
Pomara N, Crandall DT, Choi SJ, Johnson G, Lim KO. 2001. munodeficiency virus (HIV) drug resistance mutations
White matter abnormalities in HIV-1 infection: A diffu- in diverse areas of the brain in HIV-infected patients,
sion tensor imaging study. Psychiatry Res 106: 15 24. with and without dementia, on antiretroviral treatment.
Price RW, Sidtis JJ, Brew BJ. 1991. AIDS dementia complex J Virol 78: 1013310148.
and HIV-1 infection: Aview from the clinic. Brain Pathol Smurzynski M, Wu K, Letendre S, Robertson K, Bosch RJ,
1: 155 162. Clifford DB, Evans S, Collier AC, Taylor M, Ellis R.
Pulliam L, Gascon R, Stubblebine M, McGuire D, McGrath 2011. Effects of central nervous system antiretroviral pen-
MS. 1997. Unique monocyte subset in patients with etration on cognitive functioning in the ALLRT cohort.
AIDS dementia. Lancet 349: 692 695. AIDS 25: 357 365.

16 Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120


Pathogenesis and Management of Neuro-AIDS

Soulas C, Donahue RE, Dunbar CE, Persons DA, Alvarez X, Tozzi V, Balestra P, Salvatori MF, Vlassi C, Liuzzi G, Giancola
Williams KC. 2009. Genetically modified CD34 hema- ML, Giulianelli M, Narciso P, Antinori A. 2009. Changes
topoietic stem cells contribute to turnover of brain in cognition during antiretroviral therapy: Comparison
perivascular macrophages in long-term repopulated pri- of 2 different ranking systems to measure antiretroviral
mates. Am J Pathol 174: 18081817. drug efficacy on HIV-associated neurocognitive disor-
Soulas C, Conerly C, Kim W-K, Burdo TH, Alvarez X, ders. J Acquir Immune Defic Syndr 52: 56 63.
Lackner AA, Williams KD. 2011. Recently infiltrating Valcour VGS, Cecilia M, Watters RM, Sacktor NC. 2004.
MAC387 monocytes/macrophages. A third macro- Cognitive impairment in older HIV-1-seropositive indi-
phage population involved in SIV and HIV encephalitic viduals: Prevalence and potential mechanisms. AIDS 18
lesion formation. Am. J Pathol 178: 21212135. Suppl 1: S79 S86.
Spudich S, Nilsson A, Lollo N, Liegler T, Petropoulos C, Valcour V, Paul R, Neuhaus J, Shikuma C. 2011. The effects
Deeks S, Paxinos E, Price R. 2005. Cerebrospinal fluid of age and HIVon neuropsychological performance. J Int
HIV infection and pleocytosis: Relation to systemic infec- Neuropsychol Soc 17: 190 195.
tion and antiretroviral treatment. BMC Infect Dis 5: 98. van Marle G, Rourke SB, Zhang K, Silva C, Ethier J, Gill MJ,
Spudich S, Lollo N, Liegler T, Deeks SG, Price RW. 2006. Power C. 2002. HIV dementia patients exhibit reduced
Treatment benefit on cerebrospinal fluid HIV-1 levels in viral neutralization and increased envelope sequence
the setting of systemic virological suppression and failure. diversity in blood and brain. AIDS 16: 1905 1914.
J Infect Dis 194: 1686 1696. Wang Y, Zhang J, Gutman B, Chan TF, Becker JT, Aizenstein
Spudich S, Gisslen M, Hagberg L, Lee E, Leigler T, Brew B, HJ, Lopez OL, Tamburo RJ, Toga AW, Thompson PM.
Fuchs D, Tambussi G, Cinque P, Hecht F, et al. 2011. Cen- 2009. Multivariate tensor-based morphometry on sur-
tral nervous system immune activation characterizes faces: Application to mapping ventricular abnormalities
primary HIV-1 infection even in subjects with minimal in HIV/AIDS. Neuroimage 49: 2141 2157.
cerebrospinal fluid viral burden. J Infect Dis 204: 753 Wiley CA, Schrier RD, Nelson JA, Lampert PW, Oldstone
760. MB. 1986. Cellular localization of human immunodefi-
Sun B, Abadjian L, Rempel H, Calosing C, Rothlind J, Pull- ciency virus infection within the brains of acquired
iam L. 2010. Peripheral biomarkers do not correlate with immune deficiency syndrome patients. Proc Natl Acad
cognitive impairment in highly active antiretroviral Sci 83: 7089 7093.
therapy-treated subjects with human immunodeficiency Wright EJ, Grund B, Robertson K, Brew BJ, Roediger M,
www.perspectivesinmedicine.org

virus type 1 infection. J Neurovirol 16: 115 124. Bain MP, Drummond F, Vjecha MJ, Hoy J, Miller C,
Thomas ER, Dunfee RL, Stanton J, Bogdan D, Taylor J, et al. Cardiovascular risk factors associated with lower
Kunstman K, Bell JE, Wolinsky SM, Gabuzda D. 2007a. baseline cognitive performance in HIV-positive persons.
Macrophage entry mediated by HIV Envs from brain Neurology 75: 864 873.
and lymphoid tissues is determined by the capacity to Xing HQ, Hayakawa H, Izumo K, Kubota R, Gelpi E, Budka
use low CD4 levels and overall efficiency of fusion. Virol- H, Izumo S. 2009. In vivo expression of proinflammatory
ogy 360: 105 119. cytokines in HIV encephalitis: An analysis of 11 autopsy
Thomas ER, Dunfee RL, Stanton J, Bogdan D, Kunstman K, cases. Neuropathology 29: 433 442.
Wolinsky SM, Gabuzda D. 2007b. High frequency of Yilmaz A, Stahle L, Hagberg L, Svennerholm B, Fuchs D,
defective vpu compared with tat and rev genes in brain Gisslen M. 2004. Cerebrospinal fluid and plasma HIV-1
from patients with HIV type 1-associated dementia. RNA levels and lopinavir concentrations following lopi-
AIDS Res Hum Retroviruses 23: 575580. navir/ritonavir regimen. Scand J Infect Dis 36: 823 828.
Thurnher MM, Castillo M, Stadler A, Rieger A, Schmid B, Yilmaz A, Price RW, Spudich S, Fuchs D, Hagberg L, Gisslen
Sundgren PC. 2005. Diffusion-tensor MR imaging of M. 2008. Persistent intrathecal immune activation in
the brain in human immunodeficiency virus-positive HIV-1-infected individuals on antiretroviral therapy.
patients. AJNR Am J Neuroradiol 26: 22752281. J Acquir Immune Defic Syndr 47: 168 173.

Cite this article as Cold Spring Harb Perspect Med 2012;2:a007120 17

Das könnte Ihnen auch gefallen