Sie sind auf Seite 1von 11

Mitochondrion 4 (2004) 119–129


www.elsevier.com/locate/mito

Review

HIV-associated dementia, mitochondrial dysfunction,


and oxidative stress
Victor Valcour*, Bruce Shiramizu
NeuroAIDS Specialized Neuroscience Research Program, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
Received 11 March 2004; received in revised form 4 May 2004; accepted 6 May 2004

Abstract

Over the past several decades, researchers have made large advances in unraveling the pathogenesis of HIV-related
neurological disease leading to substantial benefits for affected individuals. Concomitant advances in HIV-treatment have
changed the landscape of HIV care, resulting in alterations in HIV neuroepidemiolgy and potentially the neuropathogenesis of
cognitive disorders. Specifically, widespread ARV medications use has heightened our awareness of mitochondrial toxicity,
oxidative stress, and metabolic abnormalities and stimulated more research into the related cognitive consequences. Specific
sources of oxidative stress among HIV-1-infected individuals to be discussed in this article include the direct effects of HIV-1,
chronic immune activation in response to HIV-1 and other pathogens, and co-morbid factors. Continued research in this area
could provide novel therapeutic targets.
q 2004 Elsevier B.V. and Mitochondria Research Society. All rights reserved.

Keywords: AIDS; Mitochondria; Oxidative stress

Beyond diseases with a clear mitochondrial basis mitochondrial dysfunction has been hypothesized for a
such as mitochondrial encephalopathy with lactic wide range of common neurodegenerative disorders
acidosis and stroke-like episodes (MELAS) and (Castellani et al., 2002; Dawson and Dawson, 2002;
myoclonus epilepsy with ragged red fibers (MERRF), Orrell and Schapira, 2002). A commonly referenced

Abbreviations: AACTG, adult AIDS clinical trials group; AD, Alzheimer’s disease; AIF, apoptosis-inducing factor; ARV, antiretroviral
medications; BBB, blood brain barrier; CNS, central nervous system; CSF, cerebrospinal fluid; FDRA, Friedreich’s Ataxia; GRID, gay related
immune deficiency; GSH, glutathione; HAD, HIV-associated dementia; HNE, 4-hydroxynonenal; MAO-B, monoamine oxidase B; MCMD,
minor cognitive motor disorder; MELAS, mitochondrial encephalopathy with lactic acidosis and stroke-like episodes; MERRF, myoclonus
epilepsy with ragged red fibers; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetra hydropyridine; NAC, N-Acetyl cysteine; NF-K beta, nuclear
transcription factor beta; NO, nitric oxide; NOS, nitric oxide synthase; NRTI, nucleoside reverse transcriptase inhibitors; PD, parkinson’s
disease; PET, positron emission tomography; ROS, reactive oxidative species; SOD, superoxide dismutase; TK, thymidine kinase.
* Corresponding author. Address: Office of Neurology and Aging Research, Sinclair 202, Leahi Hospital, 3675 Kilauea Avenue, Honolulu,
Hawaii 96816, USA. Tel.: C1-808-411-1582; fax: C1-808-735-8274.
E-mail address: vvalcour@hawaii.edu (V. Valcour).

1567-7249/$ - see front matter q 2004 Elsevier B.V. and Mitochondria Research Society. All rights reserved.
doi:10.1016/j.mito.2004.05.009
120 V. Valcour, B. Shiramizu / Mitochondrion 4 (2004) 119–129

mechanistic pathway involves accumulation of oxi- and oxidative stress may play important adjunct
dative stress, a process also associated with aging roles in HIV neuropathogenesis.
(Nicholls, 2002). In some cases (e.g. Alzheimer’s
disease (AD)), speculation concerning alternative
mechanistic pathways is common; however, consensus 1. The changing epidemiology of HAD and
among reported mechanisms is generally poor (Cas- neuronal susceptibility to mitochondrial
tellani et al., 2002). The variety of these propositions dysfunction
may be more reflective of the heterogeneity commonly
associated with mitochondrial disorders themselves Mortality from HIV-1 infection has declined
(Cohen and Gold, 2001). Tissue specificity, hetero- remarkably within developed countries over the past
plasmy of genetic material, threshold levels of genetic 10 years; yet little benefit has been realized concern-
abnormality or mitochondrial dysfunction required to ing the associated cognitive complications (McArthur
trigger disease, and incomplete penetrance of genetic et al., 2003). Much attention has been placed on direct
defects all contribute to a variable degree of pheno- HIV-1-related mechanisms and viral dynamics; how-
typic expression and implicit difficulty in diagnosing ever, existing evidence suggests that the direct actions
and researching clinical mitochondrial disorders of HIV-1 itself, by and large, are insufficient to fully
(Cohen and Gold, 2001; Wallace, 1999; Schon and explain the cognitive deficits associated with infection
Manfredi, 2003). (Mollace et al., 2001; Gartner, 2000; Albright et al.,
Interest in mitochondrial dysfunction relative to 2003). Support for this concept includes knowledge
HIV-1 infection has increased in recent years that the amount of HIV DNA in the brain does not
differ among demented compared to non-demented
paralleling a heightened awareness of antiretroviral
patients at post-mortem examination (Johnson et al.,
(ARV)-induced cellular toxicity (Lewis et al., 2001).
1996). Moreover, while HIV-1 is present in the brain
ARV medications and particularly dideoxynucleotide
early in infection, the onset of cognitive symptoms is
reverse transcriptase inhibitors (NRTI) are toxic to
variable and typically more strongly associated with
mitochondria partially through inhibition of mito-
advanced stages of immune suppression (McArthur
chondrial DNA polymerase gamma (Lewis et al.,
et al., 2003; Gartner, 2000).
2003). While our understanding of such toxicity is a
The neuroepidemiology of HIV-1-cognitive dis-
relatively recent phenomenon, HIV-associated risk
orders is changing in the face of widespread ARV use.
for mitochondrial damage, chiefly through oxidative Neurocognitive disease is often clinically less fulmi-
mechanisms, is not new and is inherently associated nate. The rate of mild impairment (minor cognitive
with acute and chronic viral infections (Peterhans, motor disorder (MCMD)) may be rising in proportion
1997; Schwarz, 1996; Akaike et al., 1990). Additional to that of HIV-associated dementia (HAD) (McArthur
specific aspects of HIV-1 itself and those of chronic et al., 2003). Consequently, a continued and vigilant
immune activation contribute further to reactive investigation into potential contributing factors
oxidative species (ROS) and disruption of energy associated with HIV-1 neurodegeneration is needed
homeostasis (Mollace et al., 2001; Toborek et al., and may yield important therapeutic targets to be
2003; Aksenov et al., 2003; Kruman et al., 1998). further interrogated.
Among HIV-1-infected individuals, accompanying A mitochondrial hypothesis is particularly intri-
oxidative risk factors, which include a high rate of guing. Multiple schemes including direct toxicity to
tobacco smoking, disproportionate representation of mitochondria resulting in acute energy depletion,
illicit drug use, and, more recently, an aging mutations and consequent DNA sequencing errors
seropositive population may further contribute to leading to depletion of important mitochondrial
increased oxidative stress among this vulnerable proteins, and cumulative oxidative stress contributing
population (Burns et al., 1996; Valcour et al., 2004). to cell membrane defects and signaling errors have
This paper reviews existing data relating to the been speculated and have received variable levels of
potential mitochondrial mechanisms associated with testing (Mollace et al., 2001; Turchan et al., 2003;
HIV and cognition. Mitochondrial dysfunction Haughey et al., 2004; Baier-Bitterlich et al., 1997;
V. Valcour, B. Shiramizu / Mitochondrion 4 (2004) 119–129 121

Hulgan et al., 2003; Olinski et al., 2002). For example, 2. Models of neurodegeneration and mitochondrial
the disruption of oxidative phosphorylation and the disorders
presence of ROS may induce mitochondrial-mediated
apoptosis through steric alteration of the mitochon- Mitochondrial disorders often include important
drial permeability transport pore resulting in loss of neurodegenerative components with central
neuronal cell populations (Schon and Manfredi, nervous system findings (Schon and Manfredi, 2003;
2003). Post-translational modifications of proteins Thyagarajan and Byrne, 2002). In some cases, such as
associated with ROS have been implicated in Friedreich’s Ataxia (FDRA), point mutations in
decreased blood brain barrier (BBB) defenses with mitochondrial DNA have been identified. Since the
HIV-1 infection (Mollace et al., 2001). resultant defect is one of mitochondrial dysfunction
Neurons, like muscle cells and especially cardiac with disruption to the electron transport chain and
muscle cells, are particularly vulnerable to the since many symptoms are similar to disease states
effects of mitochondrial damage due to their high in treated HIV-1-infected patients (Shikuma and
dependence on energy (Schon and Manfredi, 2003; Shiramizu, 2001), FRDA may provide some
Thyagarajan and Byrne, 2002; Facchinetti et al., insight into various potential mechanisms relating to
1998). Consequently, many well-recognized mito- HIV-associated mitochondrial dysfunction.
chondrial diseases have both myocardial and central FRDA is manifest by progressive limb ataxia,
nervous system consequences (Thyagarajan and axonal sensory neuropathy, and pyramidal signs
Byrne, 2002). Additionally, there are numerous (Cooper and Bradley, 2002). Non-neurological seque-
substrates prone to oxidation and consequent lae include skeletal abnormalities, cardiomyopathy,
and an increased incidence of diabetes mellitus. It is
dysfunction in the brain including catecholamines,
associated with an autosomal recessive inheritance
structural lipid components such as myelin, and
pattern involving a GAA trinucleotide expansion on
signaling molecules (Tong et al., 2002). Decreased
chromosome 9 leading to a marked reduction in
anti-oxidant defenses have also been described
frataxin and consequent insufficiency of oxidative
within the brain, potentially increasing susceptibility
phosphorylation (Cooper and Bradley, 2002). The
to ROS (Rego and Oliveira, 2003). Since NRTI-
resultant cellular state is one of marked decrease in
induced mitochondrial toxicity is related to a higher
reserve for oxidative stress possibly associated with
concentration of mitochondrial thymidine kinase
abnormalities in mitochondrial iron homeostasis and
(TK) as opposed to cytoplasmic TK, and since this the consequent development of free radicals via the
concentration is typically higher in cells with less Fenton reaction (Cooper and Bradley, 2002; Rotig
mitotic turnover, one could speculate that mitochon- et al., 1997; Puccio et al., 2001). Superoxide
dria in neurons may be particularly prone to their ill dismutase (SOD) deficiency in cultured FRDA cells
effects (Lewis et al., 2003). An increased concen- and both decreased myocardial hypertrophy and
tration of iron in selective cells within the brain, attenuation of oxidative markers with antioxidant
particularly dopaminergic cells within the substantia treatment (Idebenone) support an important role
nigra pars reticulata, may increase the risk further of oxidative stress in the pathogenesis of FRDA
by enhancing the development of more toxic (Rustin, 2003).
free radicals locally through the Fenton reaction Alzheimer’s disease is the most common form of
(Facchinetti et al., 1998). Such highly reactive states dementia in humans exhibiting an exponential rise in
associated with the presence of transition metals incidence associated with aging (Cummings et al.,
have been identified in the neurofibrillary tangles in 1998). While specific genetic determinants to early
Alzheimer’s disease and in Lewy bodies in Parkin- onset familial disease have been identified (e.g.
son’s disease (Smith et al., 1997; Castellani et al., amyloid precursor protein and the amyloid beta
2000). Cumulatively, these circumstances argue that processing proteins presenilin-1 and presenilin-2),
the central nervous system (CNS) may be particu- the etiology for the vast majority of cases remains
larly prone to the deleterious effects of mitochondria illusive (Lannfelt and Nordstedt, 2000). Existing data
dysfunction and ROS. support a possible secondary role for the effects of
122 V. Valcour, B. Shiramizu / Mitochondrion 4 (2004) 119–129

mitochondrial dysfunction and oxidative stress. This (implying enhanced risk for ROS) increased lipid
hypothesis underlies the study of antioxidant medi- peroxidation, and decreased glutathione levels within
cations including selegiline, Vitamin E, and poten- the substantia nigra of PD patients add further evidence
tially Ginkgo biloba, and somewhat relates to the that an oxidative mechanism may underlie this disease
investigation of anti-inflammatory agents as disease (Facchinetti et al., 1998; Dexter et al., 1992). While
modulators (Aisen, 2002; Andersen et al., 1995; Sano effects on dopamine homeostasis are the primary
et al., 1997; Le Bars et al., 1997). mechanism supporting the use of selegiline in the
Unlike FRDA, no underlying unifying mitochon- treatment of PD, the co-incident anti-oxidant qualities
drial deletions or mutations have been consistently may further enhance its benefit prompting a recent
associated with AD; nevertheless, abnormalities clinical trial for HIV-related cognitive deficits
associated with mitochondrial function are character- (Maruyama and Naoi, 1999).
istic findings in well-established disease (Castellani
et al., 2002; Swerdlow and Kish, 2002). Brain
bioenergetic studies reveal a decrease in brain 3. HIV-associated cognitive disorders-potential
metabolism in biopsy specimens (Sims et al., 1987) mechanisms of mitochondrial dysfunction
and with positron emission tomography (PET)
(Friedland et al., 1983; Foster et al., 1983) with PET 3.1. Oxidative stress
changes often preceding detectable cognitive abnorm-
alities (Haxby et al., 1987). Abnormalities in the Mitochondria efficiently maintain a proton gradient
Krebs cycle have been implicated; yet irregularities in across their membranes (Nicholls, 2002). Under
cytochrome C oxidase (Complex IV) are the most normal physiological conditions, only a small
commonly cited oxidative phosphorylation abnorm- inherent leak exists (Castellani et al., 2002; Dawson
alities in AD (Castellani et al., 2002). While these and Dawson, 2002; Orrell and Schapira, 2002), which
findings point to a mitochondrial contribution, it is not is augmented by sources of oxidative stress and
known if defects in mitochondrial DNA, per se, play a counter-balanced by endogenous antioxidants
significant role as anomalies in the respiratory chain (Facchinetti et al., 1998). This delicate balance of
proteins, including proteins encoded by nuclear DNA oxidant and anti-oxidant substrates is critical to
and a heightened role of exogenous oxidative stress cellular survival; imbalance, as seen in HIV-infected
could be involved (Castellani et al., 2002). A decrease patients and particularly among patients effectively
in mitochondria associated with increased mitochon- treated with ARV, results in oxidative stress (Fig. 1)
drial degradation and oxidative stress has been (Hulgan et al., 2003; Pace and Leaf, 1995).
identified among neurons in AD biopsy specimens The existence of endogenous ROS is inherent with
(Castellani et al., 2002). host responses to pathogens via neutrophils, activated
The associations among Parkinson’s Dementia macrophages, and through the process of phagocy-
(PD), mitochondrial dysfunction, and oxidative stress tosis (Jarstrand and Akerlund, 1994; Xiong et al.,
may be most strongly supported by existing research. 2000). Secreted cytokines may further induce oxi-
Owing to an animal model for PD utilizing 1-methyl-4- dative stress through induction of key enzymes such
phenyl-1,2,3,6-tetra hydropyridine (MPTP), a mito- as inducible nitric oxide synthatase (NOS) resulting in
chondrial hypothesis has been advanced (Dawson and additional ROS (nitric oxide (NO)) (Mollace et al.,
Dawson, 2002; Schon and Manfredi, 2003). 2001; Baier-Bitterlich et al., 1997). Nitric oxide is
A mitochondrial outer membrane protein, monoamine released by neurons in response to excitotoxic stimuli
oxidase B (MAO-B), converts MPTP to N-methyl-4- and with exposure to the HIV-1 glycoprotein GP120,
phenyl pyridine (MPP), which is a direct inhibitor possibly through indirect mechanisms (Mollace et al.,
of NADH ubiquinone oxidoreductase (Complex I). 1993). TNF alpha and interleukin-1-beta, both
Decreased immunoreactivity for complex implicated in HIV neuropathogenesis, have been
I subunits has been further identified in the neurons shown to induce NOS in astrocytes as well (Hori
of PD patients (Schon and Manfredi, 2003; et al., 1999). Increased cerebral spinal fluid (CSF)
Dawson and Dawson, 2003). Increased total iron oxidative stress markers (e.g.: 4-hydroxynonenal
V. Valcour, B. Shiramizu / Mitochondrion 4 (2004) 119–129 123

Fig. 1. Some sources of oxidant stress and consequences among HIV-1-infected adults.

(HNE)) have been identified in patients with HAD in further consequences for neurofilaments and neuronal
research conducted by Turchan et.al. (Turchan et al., structure (Beckman and Koppenol, 1996). ROS can
2003). Advancing an hypothesis implicating a mito- have a direct effect on complexes involved in
chondrial basis to the oxidative stress, this group oxidative phosphorylation, such as inhibition of
further identified progressively decreased mitochon- complex I resulting in decreased energy production
drial potential activity with advancing stages of HAD. (Nicholls, 2002). Oxidation of thiol-containing amino
The mechanisms by which ROS induce their acids is thought to play a role in the potential
detrimental effects are varied and, in some cases, disruption of receptor mediated cellular signaling
may be more applicable to cells within the brain. (Facchinetti et al., 1998).
Hydroxylation of various cellular moieties contributes Free radicals can damage DNA through a number
to cellular disruption. Lipid peroxidation, resulting in of mechanisms including direct alteration of base
disruption of membrane fluidity and increased pairs resulting in miscoding; some changes can be
permeability, is seen with HIV infection (Facchinetti pre-mutogenic (Olinski et al., 2002). Miscoding can
et al., 1998; Revillard et al., 1992). An interplay result in a decrease in critical proteins within neurons.
between changes in membrane permeability and the DNA analyses in HIV-infected lymphocytes demon-
induction of endothelial NOS associated with HIV strated heightened hydroxylation of guanine and
infection is proposed to diminish BBB defenses uridine residues when compared to lymphocytes
(Mollace et al., 2001). Peroxynitrite, a consequence from controls (Jaruga et al., 1999). Suppl.ementation
of increased nitric oxide free radical formation within with vitamins A, C, and E resulted in decreased
cells, has been co-localized to perivascular regions oxidative damage (Jaruga et al., 2002).
of HAD brains (Mollace et al., 2001) and associated Anti-oxidant defenses are impaired in HIV infec-
with loss of tight junctions (Boven et al., 2000). tion; however, the underlying mechanisms are not
Peroxyinitration of structural proteins can have clearly defined. Mitochondrial glutathione (GSH) is
124 V. Valcour, B. Shiramizu / Mitochondrion 4 (2004) 119–129

an important endogenous electron donor (anti-oxi- consequences (Ogedegbe et al., 2003). Apoptosis
dant) which is critical for efficient detoxification of may be a more critical issue associated with
hydrogen peroxide via enzymes found only in attenuated energy, oxidative stress and excitotoxicity.
mitochondria (Cadenas and Davies, 2000). In vitro In vitro models indicate a critical mitochondrial role
studies suggest marked sensitivity to decreased levels in signaling apoptosis through the actions of
of GSH resulting in oxidative stress (Jurma et al., the mitochondrial permeability transport pore
1997). Before widespread use of ARV, several studies (Nicholls, 2002). The opening of this membrane
indicated decreased GSH in several compartments pore results in loss of the electrochemical gradient and
including CSF among HIV-1-infected individuals release of a number of inner membrane constituents
(Buhl et al., 1989; Staal et al., 1992; Castagna et al., including cytochrome c, apoptosis-inducing factor
1995). Further GSH depletion has been associated (AIF), and caspases with the ultimate result of
with detoxification of ARV medications (Mollace signaling apoptosis (Nicholls, 2002; Wallace, 1999;
et al., 2001). GSH plasma levels correlate to HIV Rego and Oliveira, 2003). Excitotoxicity, such as that
disease progression and survival (Herzenberg et al., caused by glutamate stimulation of NMDA receptors
1997). Decreased activity of both catalase and SOD or oxidative stress can lead to apoptosis through this
have also been described (Pace and Leaf, 1995). mitochondrial mechanism (Rego and Oliveira, 2003;
The latter may be due to direct actions of the HIV Pace and Leaf, 1995). Enhanced apoptotic cell death
trans-activator protein, tat (Flores et al., 1993). is associated with exposure to TNF and hydrogen
The hypothesized consequences of these dimin- peroxide and somewhat attenuated with addition
ished antioxidant reserves are untested in vivo among of anti-oxidants in experiments conducted with
HIV-1-infected patients. A small study of limited HIV-1-infected cells (Malorni et al., 1993).
duration aimed at treating HIV-related neuropathy
through supplementation with micronutrients, includ-
ing potent anti-oxidants, resulted in an unexpected 3.3. Mitochondrial DNA deletions
beneficial change in absolute CD4 T-lymphocyte
counts, but no change in neuropathy symptoms Mitochondrial DNA deletions represent a theoreti-
(Kaiser et al., 2004). In vivo and in vitro pilot data cal mechanism of ARV toxicity in HIV patients.
support use of selegiline in HAD (Turchan et al., NRTI triphosphates inhibit mtDNA pol-gamma
2003; Sacktor et al., 2000). Consequently, a random- resulting in such deletions (Lewis, 2003). Mitochon-
ized placebo controlled intervention with transdermal drial DNA deletions underlie some encephalomyelo-
selegiline, an inhibitor of MAO-B, is underway pathies, such as Kearns Sayre Disease (Graeber and
through the Adult AIDS Clinical Trials Group Muller, 1998) and elevated quantities have been
(ACTG 5090, National Institutes of Health). Treat- reported in some neurodegenerative disorders
ment of HIV-1 infection with the antioxidant (Mawrin et al., 2004; Corral-Debrinski et al., 1994)
N-Acetyl cysteine (NAC) has been proposed to providing a precedent for HIV-associated neurologi-
decrease ROS and deactivate nuclear transcription cal diseases. Currently, there are limited data on the
factor beta (NF-K beta) (Kelly, 1998; De Rosa et al., frequency of such deletions associated with ARV
2000; Staal, 2000). treatment. While mtDNA deletions have been shown
in one case report (Bartley et al., 2001) and in sperm
3.2. Attenuated energy stores and activation samples of patients on ARV (White et al., 2001) they
of apoptosis have not been convincing reported in association with
disease states among ARV users, including data from
Energy depletion associated with mitochondrial our center relating to lipodystrophy (Walker and
DNA abnormalities has been postulated to underlie Venhoff, 2001; Shikuma et al., 2001). Nevertheless,
some aspects of metabolic complication in ARV- with prolonged exposure to ARV, cumulative oxi-
treated patients (Lewis et al., 2003). Elevated lactate dative stress, and aging of the HIV population,
levels, possibly representing altered energy homeo- threshold levels of mtDNA deletions could be met
stasis, have been described with unclear clinical resulting in clinically important consequences.
V. Valcour, B. Shiramizu / Mitochondrion 4 (2004) 119–129 125

4. Contributing factors associated with life-long environmental exposures


and ongoing physiological endogenous production
Over half of HIV-1-infected patients are past or eventually results in a threshold level of damage,
present smokers in comparison to 25–38% of the which, in turn, attenuates energy production and
general public (Wei et al., 2001; Mamary et al., 2002; disrupts cell structure (Tong et al., 2002). Decreased
Smola et al., 2001). Smoking is associated with mitochondrial function and increased mitochondrial
oxidative damage, representing a modifiable risk DNA rearrangements are seen with advancing age
factor among HIV-1-infected individuals (Wei et al., (Wallace, 1999). Age-related accumulation of oxi-
2001; Morrow et al., 1995; Reilly et al., 1996). In a dation-related mitochondrial damage may be
study of 3221 HIV-1-infected men and women enhanced in regions of high dopamine production in
enrolled in the Terry Beirn Community Programs the brain, including the caudate, putamen and
for Clinical Research on AIDS, current smokers were substantia nigra (Soong et al., 1992) The clinical
more likely than never smokers to develop AIDS presentation of HIV neurocognitive disease suggests
dementia complex after adjustment for CD4 count, that subcortical structures may be particularly vulner-
prior disease progression and use of ARV therapy able (Capaldini, 1998).
(Burns et al., 1996). These limited data require Co-existing medical conditions could further
confirmation with attention to nicotinic effects on contribute to oxidative stress. Treatment of HIV
attention and working memory (Rezvani and Levin, infection is associated with the development of
2001). While this epidemiological data by no means metabolic complications, many of which are
implies oxidative mechanisms, it is reasonable to associated with increased cardiovascular and cerebro-
speculate that an additive oxidative stress associated vascular risk (Valcour et al., 2004). Recent data
with multiple sources among individuals could occur support a role of oxidative stress in the pathogenesis
and that smoking status should be a considered of atherosclerotic disease (Lee et al., 2001). Early data
covariate in research efforts concerning these poten- support a heightened cardiovascular risk with HIV
tial mechanisms. infection and treatment; however, evidence for
Illicit drug use is a well-recognized risk factor for cerebrovascular complications is mixed (Bozzette
HIV-1-infection. Cocaine and methamphetamine, two et al., 2003; Cole et al., 2004; Friis-Moller et al.,
commonly abused drugs, have important effects on 2003). While speculative at this time, atherosclerotic
the homeostatic balance of oxidative species poten- cerebrovascular disease could modify the risk for
tially resulting in added risk for individuals with illicit HIV-related cognitive abnormalities (Valcour and
drug use and HIV (Devi and Chan, 1996; Shor-Posner HIV-associated, 2002). HIV-related immunodefi-
et al., 2002; Nath et al., 2002; Tang and Smit, 2000; ciency results in opportunistic infection. While
Lee et al., 2002). An increase in oxidative stress attenuated due to underlying immunodeficiency,
has been described with heavy alcohol use as well these infections trigger further immune activation
(Nath et al., 2002). and ROS. Non-opportunistic infections are
As a consequence to effective ARV treatment, it is potential contributors as well. Hepatitis C, which
increasingly common to encounter HIV-1-seroposi- occurs in 15–30% of HIV-infected individuals,
tive patients in their sixth and higher decade of life represents another trigger of the immune system
(Valcour et al., 2004) Aging has been associated with resulting in consequent oxidative stress (Sulkowski
increased cognitive dysfunction in a number of and Thomas, 2003).
studies (Valcour et al., 2002; Goodkin et al., 2001;
Chiesi et al., 1996). Mitochondrial abnormalities and
oxidative stress are thought to contribute to the 5. Concluding remarks
progressive senescence associated with aging among
HIV-seronegative individuals and could further In the early 1980s a cluster of Kaposi’s Sarcoma and
enhance neuronal damage among HIV-1-infected pneumocystic carinii pneumonia among gay men in
patients (Wallace, 1999; Tong et al., 2002). As Los Angeles marked the beginning of what would later
hypothesized, the accumulation of ROS damage be called Gay Related Immune Deficiency (GRID)
126 V. Valcour, B. Shiramizu / Mitochondrion 4 (2004) 119–129

and subsequently be recognized as the greatest Boven, L.A., Middel, J., Verhoef, J., De Groot, C.J., Nottet, H.S.,
pandemic of our time (MMWR Morb Mortal Wkly 2000. Monocyte infiltration is highly associated with loss
of the tight junction protein zonula occludens in HIV-1-
Rep. 31 (23) 1982). The first decade of research yielded
associated dementia. Neuropathol. Appl. Neurobiol. 26 (4),
tremendous advances in the treatment of opportunistic 356–360.
diseases, which subsequently improved life for Bozzette, S.A., Ake, C.F., Tam, H.K., Chang, S.W., Louis, T.A.,
afflicted individuals. The second decade of research 2003. Cardiovascular and cerebrovascular events in patients
provided potent treatments to combat the virus, treated for human immunodeficiency virus infection. N. Engl.
transforming the landscape of AIDS from a subacute J. Med. 348 (8), 702–710.
Buhl, R., Jaffe, H.A., Holroyd, K.J., Wells, F.B., Mastrangeli, A.,
illness with near certain death to a chronic disease with
Saltini, C., et al., 1989. Systemic glutathione deficiency in
hope for long-term survival for many individuals. symptom-free HIV-seropositive individuals. Lancet 2 (8675),
As we enter the third decade of research and 1294–1298.
continue to search for a cure, we must also focus our Burns, D.N., Hillman, D., Neaton, J.D., Sherer, R., Mitchell, T.,
efforts on combating the detrimental side effects to our Capps, L., et al., 1996. Cigarette smoking, bacterial pneumonia,
treatment armamentarium and the long-term conse- and other clinical outcomes in HIV-1 infection. Terry beirn
community programs for clinical research on AIDS. J. Acquir.
quences of chronic infection. Some effort must be
Immune Def. Syndr. Hum. Retrovirol. 13 (4), 374–383.
focused on mitochondrial mechanisms as a growing Cadenas, E., Davies, K.J.A., 2000. Mitochondrial free radical
volume of evidence supports a potential contribution to generation, oxidative stress, and aging1. Free Radic. Biol. Med.
the pathogenesis of associated disorders. Consequent 29 (3–4), 222–230.
discoveries may direct treatment options for this Capaldini, L., 1998. in: Sande, M.V.P. (Ed.), Psychosocial issues
vulnerable, chronically ill, and aging population. and psychiatric complications of HIV disease, 6th edition The
medical management of AIDS. WR Saunders Company,
Philadelphia, PA, pp. 249–252.
Castagna, A., Le Grazie, C., Accordini, A., Giulidori, P.,
Cavalli, G., Bottiglieri, T., et al., 1995. Cerebrospinal fluid S-
References adenosylmethionine (SAMe) and glutathione concentrations in
HIV infection: effect of parenteral treatment with SAMe.
Aisen, P.S., 2002. Anti-inflammatory agents in Alzheimer’s disease. Neurology 45 (9), 1678–1683.
Curr. Neurol. Neurosci. Rep. 2 (5), 405–409. Castellani, R.J., Siedlak, S.L., Perry, G., Smith, M.A., 2000.
Akaike, T., Ando, M., Oda, T., Doi, T., Ijiri, S., Araki, S., et al., Sequestration of iron by Lewy bodies in Parkinson’s disease.
1990. Dependence on O2- generation by xanthine oxidase of Acta Neuropathol. (Berl) 100 (2), 111–114.
pathogenesis of influenza virus infection in mice. J. Clin. Invest. Castellani, R., Hirai, K., Aliev, G., Drew, K.L., Nunomura, A.,
85 (3), 739–745. Takeda, A., et al., 2002. Role of mitochondrial dysfunction in
Aksenov, M.Y., Hasselrot, U., Wu, G., Nath, A., Anderson, C.,
Alzheimer’s disease. J. Neurosci. Res. 70 (3), 357–360.
Mactutus, C.F., et al., 2003. Temporal relationships between
Chiesi, A., Vella, S., Dally, L.G., Pedersen, C., Danner, S.,
HIV-1 Tat-induced neuronal degeneration, OX-42 immunor-
Johnson, A.M., et al., 1996. Epidemiology of AIDS dementia
eactivity, reactive astrocytosis, and protein oxidation in the rat
complex in Europe. AIDS in Europe Study Group. J. Acquir.
striatum. Brain Res. 987 (1), 1–9.
Immune Def. Syndr. Hum. Retrovirol. 11 (1), 39–44.
Albright, A.V., Soldan, S.S., Gonzalez-Scarano, F., 2003. Patho-
Cohen, B.H., Gold, D.R., 2001. Mitochondrial cytopathy in adults:
genesis of human immunodeficiency virus-induced neurological
what we know so far. Cleve Clin. J. Med. 68 (7), 625–626 (also
disease. J. Neurovirol. 9 (2), 222–227.
Andersen, K., Launer, L.J., Ott, A., Hoes, A.W., Breteler, M.M., see pages 629–642).
Hofman, A., 1995. Do nonsteroidal anti-inflammatory drugs Cole, J.W., Pinto, A.N., Hebel, J.R., Buchholz, D.W., Earley, C.J.,
decrease the risk for Alzheimer’s disease? The Rotterdam study. Johnson, C.J., et al., 2004. Acquired immunodeficiency
Neurology 45 (8), 1441–1445. syndrome and the risk of stroke. Stroke 35 (1), 51–56.
Baier-Bitterlich, G., Fuchs, D., Wachter, H., 1997. Chronic immune Cooper, J.M., Bradley, J.L., 2002. Friedreich’s ataxia. Int. Rev.
stimulation, oxidative stress, and apoptosis in HIV infection. Neurobiol. 53, 147–173.
Biochem. Pharmacol. 53 (6), 755–763. Corral-Debrinski, M., Horton, T., Lott, M.T., Shoffner, J.M.,
Bartley, P.B., Westacott, L., Boots, R.J., Lawson, M., Potter, J.M., McKee, A.C., Beal, M.F., et al., 1994. Marked changes in
Hyland, V.J., et al., 2001. Large hepatic mitochondrial DNA mitochondrial DNA deletion levels in Alzheimer brains.
deletions associated with L-lactic acidosis and highly active Genomics 23 (2), 471–476.
antiretroviral therapy. Aids 15 (3), 419–420. Cummings, J.L., Vinters, H.V., Cole, G.M., Khachaturian, Z.S.,
Beckman, J.S., Koppenol, W.H., 1996. Nitric oxide, superoxide, 1998. Alzheimer’s disease: etiologies, pathophysiology, cogni-
and peroxynitrite: the good, the bad, and ugly. Am. J. Physiol. tive reserve, and treatment opportunities. Neurology 51
271 (5 Pt 1), C1424–C1437. (Suppl. 1), S65–S17.
V. Valcour, B. Shiramizu / Mitochondrion 4 (2004) 119–129 127

Dawson, T.M., Dawson, V.L., 2002. Neuroprotective and neuror- Herzenberg, L.A., De Rosa, S.C., Dubs, J.G., Roederer, M.,
estorative strategies for Parkinson’s disease. Nat. Neurosci. 5, Anderson, M.T., Ela, S.W., et al., 1997. Glutathione deficiency
1058–1061. is associated with impaired survival in HIV disease. Proc. Natl
Dawson, T.M., Dawson, V.L., 2003. Molecular pathways of Acad. Sci. USA 94 (5), 1967–1972.
neurodegeneration in Parkinson’s disease. Science 302 (5646), Hori, K., Burd, P.R., Furuke, K., Kutza, J., Weih, K.A.,
819–822. Clouse, K.A., 1999. Human immunodeficiency virus-1-infected
De Rosa, S.C., Zaretsky, M.D., Dubs, J.G., Roederer, M., macrophages induce inducible nitric oxide synthase and nitric
Anderson, M., Green, A., et al., 2000. N-acetylcysteine oxide (NO) production in astrocytes: astrocytic NO as a possible
replenishes glutathione in HIV infection. Eur. J. Clin. Invest. mediator of neural damage in acquired immunodeficiency
30 (10), 915–929. syndrome. Blood 93 (6), 1843–1850.
Devi, B.G., Chan, A.W., 1996. Cocaine-induced peroxidative stress Hulgan, T., Morrow, J., D’Aquila, R.T., Raffanti, S., Morgan, M.,
in rat liver: antioxidant enzymes and mitochondria. Rebeiro, P., et al., 2003. Oxidant stress is increased during
J. Pharmacol. Exp. Ther. 279 (1), 359–366. treatment of human immunodeficiency virus infection. Clin.
Dexter, D.T., Jenner, P., Schapira, A.H., Marsden, C.D., 1992. Infect. Dis. 37 (12), 1711–1717.
Alterations in levels of iron, ferritin, and other trace metals in Jarstrand, C., Akerlund, B., 1994. Oxygen radical release by
neurodegenerative diseases affecting the basal ganglia. The neutrophils of HIV-infected patients. Chem. Biol. Interact. 91
royal kings and queens Parkinson’s disease research group. (2–3), 141–146.
Annu. Neurol. 32, S94–100. Jaruga, P., Jaruga, B., Olczak, A., Halota, W., Olinski, R.,
Facchinetti, F., Dawson, V.L., Dawson, T.M., 1998. Free radicals as Oxidative, D.N.A., 1999. base damage in lymphocytes of
mediators of neuronal injury. Cell Mol. Neurobiol. 18 (6), 667– HIV-infected drug users. Free Radic. Res. 31 (3), 197–200.
682. Jaruga, P., Jaruga, B., Gackowski, D., Olczak, A., Halota, W.,
Flores, S.C., Marecki, J.C., Harper, K.P., Bose, S.K., Nelson, S.K., Pawlowska, M., et al., 2002. Suppl.ementation with antioxidant
McCord, J.M., 1993. Tat protein of human immunodeficiency vitamins prevents oxidative modification of DNA in lympho-
virus type 1 represses expression of manganese superoxide cytes of HIV-infected patients. Free Radic. Biol. Med. 32 (5),
dismutase in HeLa cells. Proc. Natl Acad. Sci. USA 90 (16), 414–420.
7632–7636. Johnson, R.T., Glass, J.D., McArthur, J.C., Chesebro, B.W., 1996.
Foster, N.L., Chase, T.N., Fedio, P., Patronas, N.J., Brooks, R.A., Di Quantitation of human immunodeficiency virus in brains of
Chiro, G., 1983. Alzheimer’s disease: focal cortical changes demented and nondemented patients with acquired immunode-
shown by positron emission tomography. Neurology 33 (8), ficiency syndrome. Annu. Neurol. 39 (3), 392–395.
961–965. Jurma, O.P., Hom, D.G., Andersen, J.K., 1997. Decreased
Friedland, R.P., Budinger, T.F., Ganz, E., Yano, Y., Mathis, C.A., glutathione results in calcium-mediated cell death in PC12.
Koss, B., et al., 1983. Regional cerebral metabolic alterations in Free Radic. Biol. Med. 23 (7), 1055–1066.
dementia of the Alzheimer type: positron emission tomography Kaiser, J.O., Snatos, J., Leoung, G., Brown, G., Mass, S., Baum, M.,
with [18F]fluorodeoxyglucose. J. Comput. Assist. Tomogr. 7 (4), Poster, M., 2004. Broad-spectrum micronutrient supplemen-
590–598. tation in HIV-infected patients with dideoxynucleoside-related
Friis-Moller, N., Sabin, C.A., Weber, R., d’sArminio Monforte, A., peripheral neuropathy: a prospective, double-blind, placebo-
El-Sadr, W.M., Reiss, P., et al., 2003. Combination antiretro- controlled tria In: 11th Conference of Retroviruses and
viral therapy and the risk of myocardial infarction. N. Engl. Opportunistic Infections; 2004; San Francisco, CA 2004.
J. Med. 349 (21), 1993–2003. Kelly, G.S., 1998. Clinical applications of N-acetylcysteine. Altern.
Gartner, S., 2000. HIV infection and dementia. Science 287 (5453), Med. Rev. 3 (2), 114–127.
602–604. Kruman, I.I., Nath, A., Mattson, M.P., 1998. HIV-1 protein Tat
Goodkin, K., Wilkie, F.L., Concha, M., Hinkin, C.H., Symes, S., induces apoptosis of hippocampal neurons by a mechanism
Baldewicz, T.T., et al., 2001. Aging and neuro-AIDS conditions involving caspase activation, calcium overload, and oxidative
and the changing spectrum of HIV-1-associated morbidity and stress. Exp. Neurol. 154 (2), 276–288.
mortality. J. Clin. Epidemiol. 54 (Suppl. 1), S35–S43. Lannfelt, L., Nordstedt, C., 2000. Genetics of Alzheimer’s disease–
Graeber, M.B., Muller, U., 1998. Recent developments in the routes to the pathophysiology. J. Neural. Transm. Suppl. 59,
molecular genetics of mitochondrial disorders. J. Neurol. Sci. 155–161.
153 (2), 251–263. Le Bars, P.L., Katz, M.M., Berman, N., Itil, T.M., Freedman, A.M.,
Haughey, N.J., Cutler, R.G., Tamara, A., McArthur, J.C., Schatzberg, A.F., 1997. A placebo-controlled, double-blind,
Vargas, D.L., Pardo, C.A., et al., 2004. Perturbation of randomized trial of an extract of Ginkgo biloba for dementia.
sphingolipid metabolism and ceramide production in HIV- North American EGb study group. JAMA 278 (16), 1327–1332.
dementia. Annu. Neurol. 55 (2), 257–267. Lee, S.H., Blair, I.A., 2001. Oxidative DNA damage and cardiovas-
Haxby, J.V., Grady, C.L., Friedland, R.P., Rapoport, S.I., 1987. cular disease. Trends Cardiovasc. Med. 11 (3–4), 148–155.
Neocortical metabolic abnormalities precede nonmemory Lee, Y.W., Son, K.W., Flora, G., Hennig, B., Nath, A., Toborek, M.,
cognitive impairments in early dementia of the Alzheimer 2002. Methamphetamine activates DNA binding of specific
type: longitudinal confirmation. J. Neural Transm. Suppl. 24, redox-responsive transcription factors in mouse brain.
49–53. J. Neurosci. Res. 70 (1), 82–89.
128 V. Valcour, B. Shiramizu / Mitochondrion 4 (2004) 119–129

Lewis, W., 2003. Mitochondrial dysfunction and nucleoside reverse assessment of the role in carcinogenesis, atherosclerosis, and
transcriptase inhibitor therapy: experimental clarifications and acquired immunodeficiency syndrome. Free Radic. Biol. Med.
persistent clinical questions. Antiviral Res. 58 (3), 189–197. 33 (2), 192–200.
Lewis, W., Copeland, W.C., Day, B.J., 2001. Mitochondrial dna Orrell, R.W., Schapira, A.H., 2002. Mitochondria and amyotrophic
depletion, oxidative stress, and mutation: mechanisms of lateral sclerosis. Int. Rev. Neurobiol. 53, 411–426.
dysfunction from nucleoside reverse transcriptase inhibitors. Pace, G.W., Leaf, C.D., 1995. The role of oxidative stress in HIV
Lab Invest. 81 (6), 777–790. disease. Free Radic. Biol. Med. 19 (4), 523–528.
Lewis, W., Day, B.J., Copeland, W.C., 2003. Mitochondrial toxicity Peterhans, E., 1997. Oxidants and antioxidants in viral diseases:
of NRTI antiviral drugs: an integrated cellular perspective. Nat. disease mechanisms and metabolic regulation. J. Nutr. 127
Rev. Drug Discov. 2 (10), 812–822. (Suppl. 5), 962S–965S.
Malorni, W., Rivabene, R., Santini, M.T., Donelli, G., 1993. Puccio, H., Simon, D., Cossee, M., Criqui-Filipe, P., Tiziano, F.,
N-acetylcysteine inhibits apoptosis and decreases viral particles Melki, J., et al., 2001. Mouse models for Friedreich ataxia
in HIV-chronically infected U937 cells. FEBS Lett. 327 (1), 75– exhibit cardiomyopathy, sensory nerve defect and Fe–S enzyme
78. deficiency followed by intramitochondrial iron deposits. Nat.
Mamary, E.M., Bahrs, D., Martinez, S., 2002. Cigarette smoking Genet. 27 (2), 181–186.
and the desire to quit among individuals living with HIV. AIDS Rego, A.C., Oliveira, C.R., 2003. Mitochondrial dysfunction and
Patient Care STDS 16 (1), 39–42. reactive oxygen species in excitotoxicity and apoptosis:
Maruyama, W., Naoi, M., 1999. Neuroprotection by (-)-deprenyl implications for the pathogenesis of neurodegenerative diseases.
and related compounds. Mech. Ageing Dev. 111 (2–3), 189– Neurochem. Res. 28 (10), 1563–1574.
200. Reilly, M., Delanty, N., Lawson, J.A., FitzGerald, G.A., 1996.
Mawrin, C., Kirches, E., Krause, G., Schneider-Stock, R., Modulation of oxidant stress in vivo in chronic cigarette
Bogerts, B., Vorwerk, C.K., et al., 2004. Region-specific smokers. Circulation 94 (1), 19–25.
analysis of mitochondrial DNA deletions in neurodegenerative Revillard, J.P., Vincent, C.M., Favier, A.E., Richard, M.J.,
disorders in humans. Neurosci. Lett. 357 (2), 111–114. Zittoun, M., Kazatchkine, M.D., 1992. Lipid peroxidation in
McArthur, J.C., Haughey, N., Gartner, S., Conant, K., Pardo, C., human immunodeficiency virus infection. J. Acquir. Immune
Nath, A., et al., 2003. Human immunodeficiency virus- Def. Syndr. 5 (6), 637–638.
associated dementia: an evolving disease. J. Neurovirol. 9 (2), Rezvani, A.H., Levin, E.D., 2001. Cognitive effects of nicotine.
205–221. Biol. Psychiatr. 49 (3), 258–267.
Mollace, V., Colasanti, M., Rodino, P., Massoud, R., Lauro, G.M., Rotig, A., de Lonlay, P., Chretien, D., Foury, F., Koenig, M., Sidi, D.,
Nistico, G., 1993. Cytokine-induced nitric oxide generation by et al., 1997. Aconitase and mitochondrial iron–sulphur protein
cultured astrocytoma cells involves Ca(CC)-calmodulin- deficiency in Friedreich ataxia. Nat. Genet. 17 (2), 215–217.
independent NO-synthase. Biochem. Biophys. Res. Commun. Rustin, P., 2003. The use of antioxidants in Friedreich’s ataxia
191 (2), 327–334. treatment. Expert Opin. Invest. Drugs 12 (4), 569–575.
Mollace, V., Nottet, H.S., Clayette, P., Turco, M.C., Muscoli, C., Sacktor, N., Schifitto, G., McDermott, M.P., Marder, K.,
Salvemini, D., et al., 2001. Oxidative stress and neuroAIDS: McArthur, J.C., Kieburtz, K., 2000. Transdermal selegiline in
triggers, modulators and novel antioxidants. Trends Neurosci. HIV-associated cognitive impairment: pilot, placebo-controlled
24 (7), 411–416. study. Neurology 54 (1), 233–235.
Morrow, J.D., Frei, B., Longmire, A.W., Gaziano, J.M., Sano, M., Ernesto, C., Thomas, R.G., Klauber, M.R., Schafer, K.,
Lynch, S.M., Shyr, Y., et al., 1995. Increase in circulating Grundman, M., et al., 1997. A controlled trial of selegiline,
products of lipid peroxidation (F2-isoprostanes) in smokers. alpha-tocopherol, or both as treatment for Alzheimer’s disease.
Smoking as a cause of oxidative damage. N. Engl J. Med. 332 The Alzheimer’s disease cooperative study. N. Engl J. Med. 336
(18), 1198–1203. (17), 1216–1222.
Nath, A., Hauser, K.F., Wojna, V., Booze, R.M., Maragos, W., Schon, E.A., Manfredi, G., 2003. Neuronal degeneration and
Prendergast, M., et al., 2002. Molecular basis for interactions of mitochondrial dysfunction. J. Clin. Invest. 111 (3), 303–312.
HIV and drugs of abuse. J. Acquir. Immune Def. Syndr. 31 Schwarz, K.B., 1996. Oxidative stress during viral infection: a
(Suppl. 2), S62–S69. review. Free Radic. Biol. Med. 21 (5), 641–649.
Nicholls, D., 2002. Mitochondrial bioenergetics, aging, and aging- Shikuma, C.M., Shiramizu, B., 2001. Mitochondrial toxicity
related disease. Sci. Aging Knowledge Environ. 2002 (31), associated with nucleoside reverse transcriptase inhibitor
pe12. therapy. Curr. Infect. Dis. Rep. 3 (6), 553–560.
Nicholls, D.G., 2002. Mitochondrial function and dysfunction in the Shikuma, C.M., Hu, N., Milne, C., Yost, F., Waslien, C.,
cell: its relevance to aging and aging-related disease. Int. Shimizu, S., et al., 2001. Mitochondrial decrease in subcu-
J. Biochem. Cell Biol. 34 (11), 1372–1381. taneous adipose tissue of HIV-infected individuals with
Ogedegbe, A.E., Thomas, D.L., Diehl, A.M., 2003. Hyperlactatae- peripheral lipoatrophy. Aids 15 (14), 1801–1809.
mia syndromes associated with HIV therapy. Lancet Infect. Dis. Shor-Posner, G., Lecusay, R., Morales, G., Campa, A., Miguez-
3 (6), 329–337. Burbano, M.J., 2002. Neuroprotection in HIV-positive drug
Olinski, R., Gackowski, D., Foksinski, M., Rozalski, R., users: implications for antioxidant therapy. J. Acquir. Immune
Roszkowski, K., Jaruga, P., 2002. Oxidative DNA damage: Def. Syndr. 31 (Suppl. 2), S84–S88.
V. Valcour, B. Shiramizu / Mitochondrion 4 (2004) 119–129 129

Sims, N.R., Finegan, J.M., Blass, J.P., Bowen, D.M., Neary, D., Turchan, J., Pocernich, C.B., Gairola, C., Chauhan, A., Schifitto, G.,
1987. Mitochondrial function in brain tissue in primary Butterfield, D.A., et al., 2003. Oxidative stress in HIV demented
degenerative dementia. Brain Res. 436 (1), 30–38. patients and protection ex vivo with novel antioxidants.
Smith, M.A., Harris, P.L., Sayre, L.M., Perry, G., 1997. Iron Neurology 60 (2), 307–314.
accumulation in Alzheimer disease is a source of redox- Valcour, V., HIV-associated, N.S., 2002. dementia and aging.
generated free radicals. Proc. Natl Acad. Sci. USA 94 (18), J. Ment. Health Aging 8 (4), 295–306.
9866–9868. Valcour, V.S., Shikuma, B., Poff, C., Watters, P., Grove, J.,
Smola, S., Justice, A.C., Wagner, J., Rabeneck, L., Weissman, S., Selnes, O., Sacktor, N., 2002. Neurocognitive function among
Rodriguez-Barradas, M., 2001. Veterans aging cohort three-site older compared to younger HIV-1 seropositive individuals In:
study (VACS 3): overview and description. J. Clin. Epidemiol. fourth International Symposium on Neurovirology, 2002,
54 (Suppl. 1), S61–S76. Dusseldorf, Germany 2002.
Soong, N.W., Hinton, D.R., Cortopassi, G., Arnheim, N., 1992. Valcour, V.S.C., Watters, M., Sacktor, N., 2004. Cognitive
Mosaicism for a specific somatic mitochondrial DNA mutation impairment in older HIV-1 seropositive individuals: prevalence,
in adult human brain. Nat. Genet. 2 (4), 318–323. characteristics, and potential mechanisms. AIDS 18 (Suppl. 1),
Staal, F.J., Roederer, M., Israelski, D.M., Bubp, J., Mole, L.A., S79–S86.
McShane, D., et al., 1992. Intracellular glutathione levels in T
Walker, U.A., Venhoff, N., 2001. Multiple mitochondrial DNA
cell subsets decrease in HIV-infected individuals. AIDS Res.
deletions and lactic acidosis in an HIV-infected patient under
Hum. Retroviruses 8 (2), 305–311.
antiretroviral therapy. Aids 15 (11), 1449–1450.
Staal, F., 2000. Antioxidant therapy for AIDS. Eur. J. Clin. Invest.
Wallace, D.C., 1999. Mitochondrial diseases in man and mouse.
30 (10), 841–842.
Science 283 (5407), 1482–1488.
Sulkowski, M.S., Thomas, D.L., 2003. Hepatitis C in the HIV-
Wei, W., Kim, Y., Boudreau, N., 2001. Association of smoking
Infected Person. Annu. Intern. Med. 138 (3), 197–207.
with serum and dietary levels of antioxidants in adults:
Swerdlow, R.H., Kish, S.J., 2002. Mitochondria in Alzheimer’s
NHANES III, 1988–1994. Am. J. Public Health 91 (2), 258–
disease. Int. Rev. Neurobiol. 53, 341–385.
Tang, A.M., Smit, E., 2000. Oxidative stress in HIV-1-infected 264.
injection drug users. J. Acquir. Immune Def. Syndr. 25 (Suppl. White, D.J., Mital, D., Taylor, S., St John, J.C., 2001. Sperm
1), S12–S18. mitochondrial DNA deletions as a consequence of long
Thyagarajan, D., Byrne, E., 2002. Mitochondrial disorders of the term highly active antiretroviral therapy. Aids 15 (8), 1061–
nervous system: clinical, biochemical, and molecular genetic 1062.
features. Int. Rev. Neurobiol. 53, 93–144. Xiong, H., Zeng, Y.C., Lewis, T., Zheng, J., Persidsky, Y.,
Toborek, M., Lee, Y.W., Pu, H., Malecki, A., Flora, G., Garrido, R., Gendelman, H.E., 2000. HIV-1 infected mononuclear phago-
et al., 2003. HIV-Tat protein induces oxidative and inflamma- cyte secretory products affect neuronal physiology leading to
tory pathways in brain endothelium. J. Neurochem. 84 (1), 169– cellular demise: relevance for HIV-1-associated dementia.
179. J. Neurovirol. 6 (Suppl. 1), S14–S23.
Tong, L., Toliver-Kinsky, T., Edwards, M., Rassin, D.K., Werr- A cluster of Kaposi’s sarcoma and Pneumocystis carinii pneumonia
bach-Perez, K., Perez-Polo, J.R., 2002. Attenuated transcrip- among homosexual male residents of Los Angeles and Orange
tional responses to oxidative stress in the aged rat brain. Counties. California. MMWR Morb Mortal Wkly Rep. 31 (23)
J. Neurosci. Res. 70 (3), 318–326. 1982 305–307.

Das könnte Ihnen auch gefallen