Skip to main content
Erschienen in: AIDS and Behavior 6/2011

Open Access 01.08.2011 | Original Paper

Neurocognitive Consequences of HIV Infection in Older Adults: An Evaluation of the “Cortical” Hypothesis

verfasst von: J. Cobb Scott, Steven Paul Woods, Catherine L. Carey, Erica Weber, Mark W. Bondi, Igor Grant, The HIV Neurobehavioral Research Center (HNRC) Group

Erschienen in: AIDS and Behavior | Ausgabe 6/2011

Abstract

The incidence and prevalence of older adults living with HIV infection is increasing. Recent reports of increased neuropathologic and metabolic alterations in older HIV+ samples, including increased cortical beta-amyloid, have led some researchers to suggest that aging with HIV may produce a neuropsychological profile akin to that which is observed in “cortical” dementias (e.g., impairment in memory consolidation). To evaluate this possibility, we examined four groups classified by HIV serostatus and age (i.e., younger ≤40 years and older ≥50 years): (1) Younger HIV− (n = 24); (2) Younger HIV+ (n = 24); (3) Older HIV− (n = 20); and (4) Older HIV+ (n = 48). Main effects of aging were observed on episodic learning and memory, executive functions, and visuoconstruction, and main effects of HIV were observed on measures of verbal learning and memory. The interaction of age and HIV was observed on a measure of verbal recognition memory, which post hoc analyses showed to be exclusively attributed to the superior performance of the younger HIV seronegative group. Thus, in this sample of older HIV-infected individuals, the combined effects of HIV and aging do not appear to result in a “cortical” pattern of cognitive deficits.

Introduction

Since the introduction of combined antiretroviral therapies (cART) in 1996, the mortality rates associated with HIV infection and AIDS have decreased dramatically [1]. As a consequence, a larger proportion of individuals infected with HIV are living into increasingly later years of adulthood. At the same time, the number of incident HIV infection cases among older adults continues to grow, accounting for approximately 15% of new HIV diagnoses in 2005 [1]. These convergent factors have significantly increased the prevalence of older adults living with HIV infection over the past decade; at present, approximately 27% of HIV-infected persons are aged 50 years and older [2]. In the United States, older adults currently represent 29% of AIDS cases [1], with prevalence rates estimated to rise above 50% by 2015 [3]. Although controversy remains regarding the impact of aging on HIV disease outcomes [4], older HIV-infected adults may experience greater premorbid immune down-regulation [5], more rapid disease progression [6], and higher morbidity rates [7] than younger individuals.
Independent of one another, HIV and aging are each associated with significant neuropathological alterations (e.g., reduced synaptodentritic complexity). Increasing evidence also suggests that the combination of these two risk factors may have additive or synergistic effects on the central nervous system (CNS). Moreover, the neuropathological alterations associated with aging and HIV both preferentially affect the structure and function of prefonto-striato-thalamo-cortical circuits and temporolimbic networks [8, 9]. For example, several studies have found increased amyloid deposition in the hippocampus and frontal lobes in older HIV-infected individuals [10, 11], and older age has been associated with increased prevalence of ubiquitinated proteins in the temporal lobe white matter at autopsy, perhaps indicating synaptodendritic injury, inflammatory mechanisms, and gliosis [12]. Recent neuroimaging studies in HIV also indicate that older age is associated with disproportionate frontal systems alterations, including smaller frontal grey matter volumes [13], elevated markers of glial activation in frontal white matter (e.g., myo-inositol) [14], and reduced neuronal integrity (as measured by levels of N-acetylaspartate) in the basal ganglia [14], areas long recognized as being highly vulnerable to HIV-associated neuropathologies [15]. Together, the presence of these neuropathologic and metabolic alterations suggests that increased cerebral and subcortical damage may be evident in older HIV-infected individuals, thereby amplifying the risk of neurocognitive impairment [16].
Although the conceptual commonalities between the neuropsychological profiles of aging and HIV infection were first described over 20 years ago [17], only recently have investigators begun to elucidate the combined effects of these risk factors on cognition. Older adults infected with HIV are more likely than their younger counterparts to develop HIV-associated neurocognitive disorders (HAND), including HIV-associated dementia (HAD) and HIV-associated minor neurocognitive disorder (MND) [1821]. Valcour et al. [19] reported that HIV-infected individuals aged 50 years and older had a three-fold increased risk of HAD as compared to younger adults with HIV, even after controlling for other demographic characteristics, HIV disease severity, psychiatric comorbidities, and cART use. Nondemented older adults with HIV also evidence milder forms of neuropsychological impairment at disproportionately higher levels [cf. 22], particularly in the domains of episodic memory, executive functions, and psychomotor speed [23, 24].
With the advancing age of the HIV population, greater attention has also been paid to additional conditions that have the potential to affect cognition and whose risk increases with age. For example, the aging process itself is oftentimes accompanied by a host of medical and neurological conditions including hypertension, elevated cholesterol and triglyceride levels, cerebrovascular comorbidity (e.g., stroke), and metabolic dysregulation (e.g., insulin resistance), all of which may be accelerated in the setting of HIV infection [2527]. Some researchers have also postulated that, in the context of aging, HIV infection may lead to increased rates of neurodegenerative disease by lowering the threshold for clinical manifestations of such disorders, via axonal injury or other mechanisms, in accordance with the cerebral reserve hypothesis [28, 29]. One frequently offered hypothesis is that older HIV-infected individuals may have an amplified risk of developing Alzheimer’s disease [3032] or evidence neurocognitive results similar to an Alzheimer’s presentation (e.g., a “cortical” pattern of cognitive deficits), with findings of increased levels of brain beta amyloid and plaque-like lesions in older HIV+ cohorts supporting this hypothesis [10, 11, 33, 34, cf. 12].
With this complex combination of risk factors, some researchers have suggested that the development and expression of HAND may be evolving in older adults with HIV [28, 29, 31]. In other words, older HIV+ adults may display a qualitatively different expression of HAND from that which is observed in younger and middle-aged adults, who typically display a “subcortical” cognitive profile of bradykinesia, bradyphrenia, executive dysfunction, and deficient memory encoding and retrieval [35, 36]. More specifically, older adults with HAND may be more vulnerable to cortical dysfunction and more likely to show deficits typically associated with temporal (e.g., rapid forgetting, dysnomia) and parietal lobe functioning [31], especially given the aforementioned neuropathological alterations in mediotemporal structures that are associated with aging [10, 12]. This possibility may be even more likely considering the shifting pattern of neurocognitive disorders that may already be occurring in the cART era [37, 38]. To this end, recent studies have shown dysfunction in mediotemporal regions on positron emission tomography scans [31] and functional magnetic resonance imaging (fMRI) during memory encoding and retrieval [39], as well as cortical thinning in primary sensorimotor, premotor, and visual areas [40].
Despite the growing popularity of this hypothesis, no study to our knowledge has specifically tested the possibility of HAND evolving to reflect increased “cortical” neuropsychological deficits by examining potential interactions between aging and HIV with appropriate age and HIV comparison participants, as has been recommended [41, 42]. We therefore attempted to investigate the pattern of deficits in an older HIV seropositive sample in comparison to three other groups: (1) an older HIV seronegative sample; (2) a younger HIV seropositive sample; and (3) a younger HIV seronegative sample. We hypothesized that there would be significant main effects of HIV serostatus and age on measures of episodic learning and memory and executive functions, as well as main effects of age on semantic knowledge. In addition, we hypothesized that a significant interaction would be observed between HIV and age on neuropsychological measures of episodic learning and memory, semantic knowledge, and visuospatial functioning.

Methods

Participants

The study sample included 116 participants who were classified with respect to HIV serostatus (HIV− and HIV+) and age (i.e., younger ≤40 years and older ≥50 years). This yielded four groups: (1) Younger HIV− (n = 24); (2) Younger HIV+ (n = 24); (3) Older HIV− (n = 20); and (4) Older HIV+ (n = 48). Although the “older” adults represent a relatively youthful cohort (range = 50–79 years) in comparison to the cognitive aging literature, the use of 50 years as the cutoff age was informed both by the nature of current HIV epidemic [1] and NIMH neuroAIDS research recommendations [42]. Participants were recruited from local HIV clinics and community organizations from 2005 to 2008. HIV serostatus was determined by enzyme linked immunosorbent assays and confirmed by a Western Blot test. Study exclusion criteria included: (1) severe psychiatric illness (e.g., schizophrenia); (2) neurological disease (e.g., seizure disorders, closed head injuries with loss of consciousness greater than 15 min, and CNS neoplasms or opportunistic infections); (3) an estimated verbal IQ score less than 70 on the Wechsler Test of Adult Reading (WTAR) [43]; (4) substance dependence within 6 months of evaluation; and (5) a urine toxicology screen positive for illicit drugs (other than marijuana) on the day of evaluation. Note that, a positive marijuana toxicology screen was not grounds for exclusion because drug metabolites are detectable up to 30 days after last use and several medications commonly used in HIV (e.g., efavirenz, marinol) can produce positive toxicology screens.
Regarding the demographic characteristics of the study samples, Table 1 shows that the two Younger groups were comparable in age, as were the two Older groups. However, both HIV+ groups achieved fewer years of education and contained a smaller proportion of women compared to the Younger HIV seronegative group (Ps < 0.05). In addition, the Older HIV+ group obtained lower scores on the HIV Dementia Scale (HDS) [44] compared to the other three samples, which is consistent with prior research [19]. Finally, the HIV+ groups had significantly higher proportions of individuals with lifetime diagnoses of major depression and displayed a trend-level (P < 0.10) difference in current affective distress as compared to the HIV− groups. As concerns the general medical characteristics of the samples, Table 2 shows that the Older HIV+ group had the highest prevalence of hepatitis C infection, a trend-level (P < 0.10) difference in hypercholesterolemia, and, along with the Older HIV− group, also had higher rates of hypertension. Table 2 also shows that the Younger and Older HIV+ groups did not differ in terms of cART status, HIV CSF RNA viral loads, or current CD4 lymphocyte counts (all P values > 0.10), although the Older HIV+ group had a lower nadir CD4 count (P < 0.05) and the Younger HIV+ group evidenced trends for having both a higher median viral load and a higher proportion of detectable plasma viral load (Ps < 0.10). As might be expected, the Older HIV+ group had a longer duration of infection and a higher prevalence of AIDS diagnoses than the Younger HIV+ group.
Table 1
Demographic and psychiatric characteristics of the study sample
 
Younger HIV− (n = 24)
Older HIV− (n = 20)
Younger HIV+ (n = 24)
Older HIV+ (n = 48)
F2
P
 
Demographic characteristics
 Age (years)
30.1 (7.1)
56.6 (6.3)
34.3 (5.0)
55.5 (6.4)
135.21
<0.001
Y = Y+ < O = O+
 Education (years)
15.4 (2.4)
14.5 (2.3)
13.4 (2.8)
13.6 (2.8)
3.15
0.028
Y > Y+, O+
 WTAR verbal IQ
108.3 (9.8)
105.1 (10.2)
100.8 (13.4)
102.6 (14.1)
1.69
0.174
 HIV Dementia Scale
15.8 (0.5)
15.5 (0.9)
15.6 (0.9)
14.3 (3.0)
3.77
0.01
O+ < O, Y, Y+,
 Sex (% female)
66.7%
40.0%
16.7%
20.8%
18.92
<0.001
Y > Y+, O+
 Ethnicity (% Caucasian)
50.0%
50.0%
33.3%
62.5%
18.00
0.115
Psychiatric characteristics
 Major depressiona
25.0%
25.0%
50.0%
56.3%
9.73
0.021
Y+ = O+  > O = Y
 Generalized anxietya
0.0%
5.0%
8.3%
8.5%
2.29
0.514
 Substance dependencea
25.0%
40.0%
54.2%
50.0%
5.36
0.148
 POMS total
41.6 (24.2)
44.9 (28.6)
62.8 (33.0)
48.3 (26.6)
2.53
0.064
POMS Profile of Mood States, WTAR Wechsler Test of Adult Reading, Y Younger HIV seronegative, O Older HIV seronegative, Y + Younger HIV seropositive, O + Older HIV seropositive
aDenotes a lifetime diagnosis
Table 2
HIV disease and medical characteristics of the study samples
 
Younger HIV− (n = 24)
Older HIV− (n = 20)
Younger HIV+ (n = 24)
Older HIV+ (n = 48)
χ2
P
 
Medical characteristics
 Hepatitis C
4.2%
0.0%
4.2%
25.5%
13.50
0.004
O+ > Y, Y+, O
 Hypertension
8.3%
60.0%
25.0%
41.7%
15.08
0.002
O = O+ > Y, Y+
 Hypercholesterolemia
0.0%
0.0%
0.0%
10.4%
7.40
0.060
 Diabetes mellitus
0.0%
10.0%
4.2%
10.4%
3.28
0.350
HIV disease characteristics
 AIDS (%)
37.5%
68.8%
6.43
0.011
O+ > Y+
 HAART (%)
66.7%
75.0%
1.75
0.416
 ARV regimen duration (months)
29.4 (51.8)
12.9 (20.3)
1.54
0.216
 HIV duration (years)
9.6 (6.7)
15.6 (7.0)
9.63
0.002
O+ > Y+
 Nadir CD4a (cells/μl)
253 [185, 344]
102 [29, 282]
4.88
0.027
Y+ > O+
 Current CD4a (cells/μl)
640 [299, 916]
512 [253, 693]
0.85
0.358
 Plasma HIV RNAa (log10)
1.9 [1.7, 4.1]
1.7 [1.7, 1.8]
2.73
0.098
 CSF HIV RNAa,b (log10)
1.7 [1.7, 1.8]
1.7 [1.7, 1.7]
0.18
0.670
 % Detectable plasma HIV RNA
45.8%
23.9%
3.52
0.061
 % Detectable CSF HIV RNA
20.0%
16.1%
0.11
0.745
Y Younger HIV seronegative, O Older HIV seronegative, Y + Younger HIV seropositive, O + Older HIV seropositive
aData represent medians with interquartile ranges in brackets
bO+ n = 31 and Y+ n = 15

Materials and Procedure

All participants provided written, informed consent prior to completing a comprehensive medical, psychiatric, and neuropsychological research evaluation for which they received financial compensation. The study was approved by the university’s human subjects committee.

Neuropsychological Assessment

All participants were administered a comprehensive neuropsychological assessment battery by trained research assistants. The WTAR was administered as an estimate of premorbid verbal intellectual functioning. Measures for analysis were selected from the larger battery that were known to be the most sensitive tests to cortical dysfunction, in the domains of episodic memory, semantic knowledge, executive functions, and visuospatial functioning. These measures included (1) the Boston Naming Test (BNT) [45] a standardized category (animals) verbal fluency test (Category Fluency) [46]; (2) the Famous Faces subtest of the Kaufman Adolescent and Adult Intelligence Test (KAIT) [47]; (3) the Logical Memory subtest from the Wechsler Memory Scale, 3rd edition (WMS-3) [48]; (4) the California Verbal Learning Test, 2nd edition (CVLT-II) [49]; (5) the Boston Qualitative Scoring System (BQSS) for the Rey–Osterrieth Complex Figure [50]; (6) Trailmaking Test, Part B [51]; and (7) Tower of London Test (Drexel Version) [52]. Table 3 indicates the specific variables that were analyzed from each measure. We also classified each individual into one of three memory profiles using a discriminant function algorithm based on three CVLT-II variables. This algorithm was originally derived for the first version of the CVLT [53] and classifies individuals as having a CVLT-II profile indicating an encoding/storage deficit, a retrieval deficit, or a normal profile. The utility of this algorithm has been shown in a number of studies examining the pattern of memory performance in various neurological groups, including HIV [5456].
Table 3
Neuropsychological test performance in the four study samples
 
HIV−
HIV+
Effects (F values)
Young (n = 24)
Old (n = 20)
Young (n = 24)
Old (n = 48)
HIV
Aging
HIV × Aging
Boston Naming Test (total)
55.25 (4.63)
55.55 (4.57)
53.77 (5.66)
55.00 (5.05)
0.91
0.39
0.27
KAIT Famous Faces
12.71 (3.99)
14.60 (3.33)
9.80 (3.99)
13.75 (4.63)
4.91*
11.31**
1.44
Animal fluency
22.75 (5.34)
20.47 (4.33)
21.50 (5.35)
20.77 (5.26)
0.62
1.56
0.81
WMS-3 Logical Memory
 I Total
45.29 (14.61)
48.65 (7.76)
41.50 (10.12)
38.62 (11.70)
9.38**
0.19
1.89
 II Total
33.12 (7.33)
29.95 (7.16)
24.29 (8.32)
22.21 (8.92)
24.79**
1.72
0.17
 Recognition
26.92 (2.06)
27.25 (1.68)
24.96 (2.69)
25.04 (2.86)
16.15**
0.10
0.10
 % Retained
90.71 (9.09)
85.70 (13.23)
80.88 (16.96)
78.29 (19.25)
6.91**
1.20
0.33
CVLT-II
 Total 1–5
58.83 (9.19)
51.05 (9.75)
50.63 (12.77)
46.51 (11.99)
9.29**
4.96*
1.54
 Total Cued Recall Intrusions
0.29 (0.69)
1.35 (1.57)
2.58 (2.62)
2.91 (4.09)
10.18**
1.23
0.65
 Long Delay Free Recall
13.13 (2.13)
10.45 (2.46)
10.17 (4.35)
9.30 (4.13)
9.79**
3.95*
2.31
 Recognition Discriminability
3.69 (0.36)
2.77 (0.71)
2.86 (0.94)
2.78 (0.95)
7.15**
7.37**
9.95**
Rey-O BQSS
 Copy Presence and Accuracy
18.08 (1.42)
17.05 (2.16)
17.92 (2.12)
17.35 (1.77)
0.01
3.15
1.65
 Immediate Recall Presence and Accuracy
14.20 (3.16)
11.40 (2.39)
13.08 (3.02)
10.98 (3.45)
2.49
9.40**
1.20
 Delayed Recall Presence and Accuracy
13.83 (2.85)
10.70 (3.79)
12.96 (3.68)
10.98 (3.41)
1.36
11.28**
1.71
 Delayed Retention
−1.50 (10.36)
−1.40 (16.23)
−2.96 (17.57)
2.44 (27.61)
1.36
0.51
0.38
Trailmaking Test, Part B (time)
46.75 (16.10)
69.45 (17.94)
57.25 (22.57)
80.58 (52.94)
2.44
7.28**
0.03
Tower of London (Drexel) Total Moves
24.83 (13.16)
37.95 (18.56)
27.63 (17.44)
26.83 (19.65)
1.25
2.34
3.21
Note: All data are raw scores
BQSS Boston Qualitative Scoring System, CVLT-II California Verbal Learning Test, Second Edition, KAIT Kaufman Adolescent and Adult Intelligence Test, Rey-O Rey–Osterrieth Complex Figure, WMS-3 Wechsler Memory Scale, 3rd Edition
P < 0.05; ** P < 0.01

Psychiatric Assessment

All participants underwent a structured psychiatric assessment using the Composite International Diagnostic Interview (CIDI version 2.1) [57] to generate lifetime diagnoses of Major Depressive Disorder (MDD), Generalized Anxiety Disorder (GAD), and Substance-Related Disorders per Diagnostic and Statistical Manual of Mental Disorders (4th ed.) [58] criteria. The Profile of Mood States (POMS) [59] was administered to assess overall acute (i.e., the week prior to evaluation) affective distress. The POMS is a 65-item, self-report measure of current mood states on which participants rate various adjectives (e.g., “unhappy”) on a five-point Likert-type scale ranging from 0 (“not at all”) to 4 (“extremely”). The POMS Total Mood score was used for all analyses.

Medical Assessment

Finally, all participants received a neuromedical evaluation, which included a comprehensive review of medications, medical history and current symptoms, a complete physical and neurological evaluation, CDC staging, and the HDS, which is a general cognitive screening tool that evaluates memory, attention, psychomotor speed, and construction (range = 0–16). Participants also underwent a blood draw and lumbar puncture (HIV+ n = 46). Standard flow cytometry methods were used to count CD4+ lymphocytes in blood samples. Plasma and CSF HIV RNA levels were quantified using RT-PCR (Amplicor, Roche Diagnostics, Indianapolis, IN).

Data Analyses

Power analyses showed that the study was adequately powered (>0.80) to detect interactions of medium-to-large effect sizes, using a critical alpha at 0.05, based on our sample size (N = 116). The data were screened for significant outliers (i.e., data points >3.5 SD from the overall group mean) and evaluated for normality (i.e., Shapiro–Wilk W test, P < 0.05). A series of 2 × 2 ANOVAs were run to test for main effects of age (dichotomous, ≤40 vs. ≥50 years) and HIV status (dichotomous) and the potential interaction between the two variables on the neuropsychological scores of interest. Raw neuropsychological test scores were used for all analyses. To evaluate the possible effects of confounding variables on the hypothesized effects, a series of follow-up linear regressions were performed in which the four-level aging and HIV grouping variable was used as a predictor of the neuropsychological test scores, alongside the demographic, psychiatric, and medical variables upon which omnibus group differences were observed (see Tables 1, 2). The critical alpha level was set at 0.05 for all analyses.

Results

As shown in Table 3, main effects of age were observed on select measures of learning (CVLT-II Total 1–5), memory (KAIT Famous Faces; CVLT-II Long Delay Free Recall and Recognition Discriminability; and Rey-O BQSS Immediate Recall and Delayed Recall), executive functions (Trailmaking Test, Part B), and visuoconstruction (Rey-O BQSS Copy Presence and Accuracy Score).
Main effects of HIV were observed on measures of learning (CVLT-II Total 1–5; WMS-3 Logical Memory I Total) and memory (KAIT Famous Faces; WMS-3 Logical Memory II Total, Recognition, and Percent Retained; CVLT-II Long Delay Free Recall and Recognition Discriminability). There was also a main effect of HIV on the CVLT-II classifications (χ2 (2) = 9.56, P = 0.008). Full results of the classification using the CVLT-II discriminant function algorithm are presented in Table 4.
Table 4
California Verbal Learning Test-II discriminant function classification results in the four study samples
 
Encoding/storage profile (%)
Retrieval profile (%)
Normal profile (%)
HIV− Young
0.0
16.7
83.3
HIV− Old
15.0
10.0
75.0
HIV+ Young
25.0
20.8
54.2
HIV+ Old
17.0
23.4
59.6
As shown in Table 3, an interaction effect was observed on a measure of verbal recognition memory (CVLT-II Recognition Discriminability). However, post hoc analyses revealed that this interactive effect was exclusively attributable to the superior performance of the younger HIV− group, such that the younger HIV− comparison group was significantly different from the other three groups, who were all roughly equivalent.
Post hoc analyses revealed no interpretive changes in the significance level of any HIV/aging interaction term when medical (e.g., hypertension, hepatitis C infection), psychiatric (e.g., depression), or demographic (i.e., sex and education) factors were included as covariates in the statistical models.

Discussion

With the advancing age of the HIV+ population and improved outlook for longevity in individuals with HIV infection, a number of clinical researchers have recently considered whether aging in the setting of HIV might confer additional neurocognitive risks than either condition in isolation. While it is clear that HIV and aging each contribute independent deleterious effects on the CNS, resulting in decreased cognitive functioning in a subset of individuals, indirect lines of evidence have also suggested that HIV might lower the threshold for additional neurodegenerative processes that are usually seen at a later age outside of the context of HIV. While some studies have focused on enhanced cerebrovascular [26] and metabolic [27] risk in older HIV populations, the theories that have seemingly garnered the most attention recently have focused on the potential altered expression of HAND in older adults, which may reflect increased neocortical neuropathology or dysfunction, or even an early Alzheimer’s process [28]. Given the promulgation of these theories, we sought to examine whether there might be detectable changes in the neuropsychological profile of older individuals with HIV infection that might provide behavioral evidence to support (or challenge) these theories.
In the present study, our findings converge with prior data showing that older age and HIV infection independently increase the risk for neurocognitive impairment, particularly in the areas of episodic learning and memory and executive functions. However, the combined effects of HIV and aging did not reflect what would classically be considered a “cortical” pattern of cognitive deficits. To this end, interaction effects were only found on a measure of verbal recognition memory, and these effects were exclusively due to the superior performance of the younger HIV− participants. Our findings, while preliminary, suggest that, even with potential changes in underlying neuropathology (e.g., brain beta amyloid deposition), older HIV-infected persons may not display neuropsychological deficits that appear “cortical” in nature compared to what might otherwise be expected in HIV disease and aging alone.
Given the often subtle effects of such neuropathological changes on cognition and the relative youth of our sample in comparison to the aging literature, it could be argued that the effects expected on neuropsychological test results in this study would be more likely to fall within the small-to-medium range. By this line of reasoning, having adequate statistical power for medium-to-large effect sizes may not be sufficient. However, we set a somewhat liberal alpha level in order to diminish this possibility, thereby decreasing our risk for Type II error (and inflating our Type I error risk). An even more liberal alpha value of 0.10 would result in one additional significant interactive effect on Tower of London Total Moves. Yet this result, surprisingly, was due to the poor performance of the older HIV seronegative sample, while the other three groups were equivalent.
Although we did not find interactive effects of HIV and aging in this sample, it is still unclear whether the combination of these factors confers an additive risk of neurocognitive impairment. While some prior studies have found additive effects in older HIV+ samples [19, 24], the profile of neuropsychological tests in our sample did not necessarily reflect an additive effect, although we did not explicitly test for additivity given the specific hypotheses being evaluated in this study. However, individuals in the older HIV+ group scored significantly lower on the HDS, a screening instrument designed specifically for the detection of HIV-associated dementia, suggesting potential additive effects. In addition, it should be noted that we did not examine a number of other cognitive domains that might be expected to show additive effects of HIV and aging, such as psychomotor/processing speed and working memory. Of note, these domains may be particularly important to examine in investigating the effect of other comorbidities on the profile of cognitive deficits in older HIV+ samples. For example, emerging evidence suggests that cerebrovascular risk factors associated with aging and HIV contribute significantly to performance on neurocognitive tests of psychomotor and processing speed [60], especially among individuals pharmacologically untreated for these risks [61]. Thus, it will be important to address issues of additivity and comorbidity in future studies of aging in the context of HIV, as added risk factor burden may increase the risk of functional impairment, including medication nonadherence [62].
The combined effects of HIV and advancing age raise clinically relevant questions regarding the effective diagnosis and treatment of neurocognitive symptoms in HIV infection, especially with the likelihood of additive deleterious effects [23]. The possibility that concomitant neurodegenerative processes are superimposed upon this clinical syndrome has the potential to challenge current models of diagnosis and treatment of HAND in older individuals. However, in this sample of older HIV+ individuals, we found minimal evidence for qualitatively distinct cognitive impairment due to alternative etiologies. Thus, clinicians and researchers may need to have sufficient cause to broaden their differential diagnosis to include other neurodegenerative causes in most individuals under 65 who are on cART treatment and have well controlled disease (i.e., adequate plasma and CSF viral control). This does not deny that some HIV+ individuals may experience comorbid processes associated with normal aging (e.g., cerebrovascular risk) that may be detrimental to their neurological health, although the extent to which HIV impacts this presentation remains uncertain. It is also still unclear what additional risk older HIV+ individuals with poorer disease management or additional risk factors (e.g., substance abuse) face.
A few limitations to this study suggest important avenues for future study. First, although the age of our older HIV+ group (mean = 55.4 years) is commensurate with other studies of aging in HIV [24], our sample was still relatively young in comparison to most studies of aging outside of the context of HIV. It is possible that as HIV-infected individuals advance in age beyond the range represented by our sample, their risk for cerebrovascular co-pathology and immune dysregulation may increase, especially with extended use of cART medications, which may result in a different pattern of cognitive impairment. The absence of interactive effects in this sample also raises the possibility that a lag occurs between when neuropathological markers appear in the cortex and when changes can be observed on neuropsychological tests. For example, it is clear that the underlying neuropathology of Alzheimer’s disease is laid down many years prior to the emergence of clinical symptoms [63], and future studies using an epidemiological approach may show that age and HIV interact in older HIV+ individuals to lower the age of onset for Alzheimer’s disease. However, in the case of amyloid-beta, there may be important differences in its neuropathology in HIV versus Alzheimer’s disease [64] that result in unpredictable effects on the neuropsychological profile of HAND in older adults. Future studies should examine a sample of HIV+ individuals at later ages in order to further expand on these possibilities, perhaps using neuroimaging techniques that are sensitive to subtle changes in microstructure [65, 66].
Second, our study may have suffered from survival bias, in that our older HIV+ cohort may have contained a larger proportion of long-term non-progressors or individuals with slower HIV progression, whereas post-mortem studies that have reported increased neuropathology in older HIV+ individuals may have included a larger proportion of participants susceptible to dementia (and with worse immunological health). To this end, despite having a lower nadir CD4 count, a longer duration of infection, and an increased likelihood of having AIDS, our older HIV+ cohort had similar current disease markers (e.g., current CD4) compared with the younger HIV+ group, suggesting adequate current viral suppression. Thus, it would be ideal to examine a subgroup of older HIV+ individuals with a shorter duration of infection to examine the interaction of aging and HIV in those with different patterns of progression. However, our older HIV+ sample only contained 6 participants with duration of infection less than 5 years. Therefore, in an attempt to partially address this issue, we performed post hoc correlational analyses in the older HIV+ group between duration of infection and our neuropsychological outcomes. These analyses revealed that the Boston Naming Test (r = 0.58; P < 0.0001), KAIT Famous Faces Test (r = 0.38; P = 0.01), and BQSS Delayed Retention Summary Score (r = 0.31; P = 0.04) were significantly and positively associated with duration of infection in this group. Thus, a shorter duration of infection is correlated with lower neuropsychological performances on some measures traditionally associated with a “cortical” profile of deficits in older HIV+ individuals, which is suggestive of a differential pattern of deficits in older HIV+ individuals with shorter durations of infection. Nonetheless, this finding is preliminary and awaits more detailed future analyses.
Finally, both of our HIV+ groups were relatively healthy in terms of HIV disease characteristics in comparison to many community-based samples [7]. It is possible that cognitive (and pathological) changes may be more likely to occur in individuals in later disease stages or with worse current HIV immunological health. Similarly, fewer participants evidenced HIV-associated neurocognitive disorders than might be expected—19 participants evidenced asymptomatic neuropsychological impairment, 6 participants met criteria for MND, and 1 participant met criteria for HAD. It may be that a different expression of HAND only occurs in individuals with more severe levels of cognitive impairment, such as dementia [30], which our sample failed to provide in large numbers. However, to explore this possibility, we also conducted post hoc analyses that only included individuals from the older HIV+ group with HAND diagnoses (n = 15). These analyses resulted in an interaction on the Trailmaking Test, Part B (P = 0.004), such that the Older HIV+ group with HAND had significantly greater times than the three other groups, and the Younger HIV− group had faster times than the other three groups. This finding gives some indication that older HIV+ individuals with HAND diagnoses may have increased deficits in executive functions, although these preliminary results will also need to be confirmed. Therefore, future studies should examine the pattern of neuropsychological performance in older HIV+ participants who are in later disease stages or evidence greater levels of HIV-associated neurocognitive disorders to examine for the possibility of additional neurocognitive deficits in this population.

Acknowledgments

The San Diego HIV Neurobehavioral Research Center (HNRC) group is affiliated with the University of California, San Diego, the Naval Hospital, San Diego, and the Veterans Affairs San Diego Healthcare System, and includes: Director: Igor Grant, M.D.; Co-Directors: J. Hampton Atkinson, M.D., Ronald J. Ellis, M.D., Ph.D., and J. Allen McCutchan, M.D.; Center Manager: Thomas D. Marcotte, Ph.D.; Jennifer Marquie-Beck, M.P.H.; Melanie Sherman; Naval Hospital San Diego: Braden R. Hale, M.D., M.P.H. (P.I.); Neuromedical Component: Ronald J. Ellis, M.D., Ph.D. (P.I.), J. Allen McCutchan, M.D., Scott Letendre, M.D., Edmund Capparelli, Pharm.D., Rachel Schrier, Ph.D., Terry Alexander, R.N.; Debra Rosario, M.P.H., Shannon LeBlanc; Neurobehavioral Component: Robert K. Heaton, Ph.D. (P.I.), Steven Paul Woods, Psy.D., Mariana Cherner, Ph.D., David J. Moore, Ph.D., Matthew Dawson; Neuroimaging Component: Terry Jernigan, Ph.D. (P.I.), Christine Fennema-Notestine, Ph.D., Sarah L., Archibald, M.A., John Hesselink, M.D., Jacopo Annese, Ph.D., Michael J. Taylor, Ph.D., Brian C. Schweinsburg, Ph.D.; Neurobiology Component: Eliezer Masliah, M.D. (P.I.), Ian Everall, FRCPsych., FRCPath., Ph.D., Cristian Achim, M.D., Ph.D.; Neurovirology Component: Douglas Richman, M.D., (P.I.), David M. Smith, M.D.; International Component: J. Allen McCutchan, M.D., (P.I.); Developmental Component: Ian Everall, FRCPsych., FRCPath., Ph.D. (P.I.), Stuart Lipton, M.D., Ph.D.; Participant Accrual and Retention Unit: J. Hampton Atkinson, M.D. (P.I.), Rodney von Jaeger, M.P.H.; Data Management Unit: Anthony C. Gamst, Ph.D. (P.I.), Clint Cushman (Data Systems Manager), Daniel R. Masys, M.D. (Senior Consultant); Statistics Unit: Ian Abramson, Ph.D. (P.I.), Florin Vaida Ph.D., Reena Deutsch, Ph.D., Tanya Wolfson, M.A. This research was supported by National Institute of Mental Health grants R01 MH073419 to Dr. Woods, P30 MH62512 to Dr. Grant, and National Institute on Aging grants R01 AG012674 and K24 AG026431 to Dr. Bondi. The views expressed in this article are those of the authors and do not reflect the official policy or position of the Department of the Navy, Department of Defense, or the United States Government. Authors report no conflicts of interest affecting this manuscript.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.
Open AccessThis is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License (https://​creativecommons.​org/​licenses/​by-nc/​2.​0), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Centers for Disease Control. HIV/AIDS surveillance report, 2005. Atlanta: U.S. Department of Health and Human Services, CDC; 2007. p. 1–63. Centers for Disease Control. HIV/AIDS surveillance report, 2005. Atlanta: U.S. Department of Health and Human Services, CDC; 2007. p. 1–63.
2.
Zurück zum Zitat Centers for Disease Control. HIV/AIDS surveillance report, 2007. Atlanta: U.S. Department of Health and Human Services, CDC; 2009. p. 1–54. Centers for Disease Control. HIV/AIDS surveillance report, 2007. Atlanta: U.S. Department of Health and Human Services, CDC; 2009. p. 1–54.
4.
Zurück zum Zitat Casau NC. Perspective on HIV infection and aging: emerging research on the horizon. Clin Infect Dis. 2005;41(6):855–63.PubMedCrossRef Casau NC. Perspective on HIV infection and aging: emerging research on the horizon. Clin Infect Dis. 2005;41(6):855–63.PubMedCrossRef
5.
Zurück zum Zitat Fagnoni FF, Vescovini R, Passeri G, et al. Shortage of circulating naïve cd8(+) T cells provides new insights on immunodeficiency in aging. Blood. 2000;95(9):2860–8.PubMed Fagnoni FF, Vescovini R, Passeri G, et al. Shortage of circulating naïve cd8(+) T cells provides new insights on immunodeficiency in aging. Blood. 2000;95(9):2860–8.PubMed
6.
Zurück zum Zitat Goetz MB, Boscardin WJ, Wiley D, Alkasspooles S. Decreased recovery of CD4 lymphocytes in older HIV-infected patients beginning highly active antiretroviral therapy. AIDS. 2001;15(12):1576–9.PubMedCrossRef Goetz MB, Boscardin WJ, Wiley D, Alkasspooles S. Decreased recovery of CD4 lymphocytes in older HIV-infected patients beginning highly active antiretroviral therapy. AIDS. 2001;15(12):1576–9.PubMedCrossRef
7.
Zurück zum Zitat Perez JL, Moore RD. Greater effect of highly active antiretroviral therapy on survival in people aged > or =50 years compared with younger people in an urban observational cohort. Clin Infect Dis. 2003;36(2):212–8.PubMedCrossRef Perez JL, Moore RD. Greater effect of highly active antiretroviral therapy on survival in people aged > or =50 years compared with younger people in an urban observational cohort. Clin Infect Dis. 2003;36(2):212–8.PubMedCrossRef
8.
Zurück zum Zitat Burke SN, Barnes CA. Neural plasticity in the ageing brain. Nat Rev Neurosci. 2006;7(1):30–40.PubMedCrossRef Burke SN, Barnes CA. Neural plasticity in the ageing brain. Nat Rev Neurosci. 2006;7(1):30–40.PubMedCrossRef
9.
Zurück zum Zitat Langford TD, Everall IP, Masliah E. Current concepts in HIV neuropathogenesis: neuronal injury, white matter disease, and neurotrophic factors. In: Gendelman HE, Grant I, Everall IP, Lipton S, Swindells S, editors. The neurology of AIDS. 2nd ed. London: Oxford University Press; 2005. p. 405–14. Langford TD, Everall IP, Masliah E. Current concepts in HIV neuropathogenesis: neuronal injury, white matter disease, and neurotrophic factors. In: Gendelman HE, Grant I, Everall IP, Lipton S, Swindells S, editors. The neurology of AIDS. 2nd ed. London: Oxford University Press; 2005. p. 405–14.
10.
Zurück zum Zitat Esiri MM, Biddolph SC, Morris CS. Prevalence of Alzheimer plaques in AIDS. J Neurol Neurosurg Psychiatry. 1998;65(1):29–33.PubMedCrossRef Esiri MM, Biddolph SC, Morris CS. Prevalence of Alzheimer plaques in AIDS. J Neurol Neurosurg Psychiatry. 1998;65(1):29–33.PubMedCrossRef
11.
Zurück zum Zitat Green DA, Masliah E, Vinters HV, Beizai P, Moore DJ, Achim CL. Brain deposition of beta-amyloid is a common pathologic feature in HIV positive patients. AIDS. 2005;19(4):127–35.CrossRef Green DA, Masliah E, Vinters HV, Beizai P, Moore DJ, Achim CL. Brain deposition of beta-amyloid is a common pathologic feature in HIV positive patients. AIDS. 2005;19(4):127–35.CrossRef
12.
Zurück zum Zitat Gelman BB, Schuenke K. Brain aging in acquired immunodeficiency syndrome: increased ubiquitin-protein conjugate is correlated with decreased synaptic protein but not amyloid plaque accumulation. J Neurovirol. 2004;10(2):98–108.PubMedCrossRef Gelman BB, Schuenke K. Brain aging in acquired immunodeficiency syndrome: increased ubiquitin-protein conjugate is correlated with decreased synaptic protein but not amyloid plaque accumulation. J Neurovirol. 2004;10(2):98–108.PubMedCrossRef
13.
Zurück zum Zitat Jernigan TL, Gamst AC, Archibald SL, et al. Effects of methamphetamine dependence and HIV infection on cerebral morphology. Am J Psychiatry. 2005;162(8):1461–72.PubMedCrossRef Jernigan TL, Gamst AC, Archibald SL, et al. Effects of methamphetamine dependence and HIV infection on cerebral morphology. Am J Psychiatry. 2005;162(8):1461–72.PubMedCrossRef
14.
Zurück zum Zitat Ernst T, Chang L. Effect of aging on brain metabolism in antiretroviral-naive HIV patients. AIDS. 2004;18(Suppl 1):S61–7.PubMed Ernst T, Chang L. Effect of aging on brain metabolism in antiretroviral-naive HIV patients. AIDS. 2004;18(Suppl 1):S61–7.PubMed
15.
Zurück zum Zitat Gonzalez-Scarano F, Martín-García J. The neuropathogenesis of AIDS. Nat Rev Immunol. 2005;5(1):69–81.PubMedCrossRef Gonzalez-Scarano F, Martín-García J. The neuropathogenesis of AIDS. Nat Rev Immunol. 2005;5(1):69–81.PubMedCrossRef
16.
Zurück zum Zitat Chang L, Lee PL, Yiannoutsos CT, et al. A multicenter in vivo proton-MRS study of HIV-associated dementia and its relationship to age. Neuroimage. 2004;23(4):1336–47.PubMedCrossRef Chang L, Lee PL, Yiannoutsos CT, et al. A multicenter in vivo proton-MRS study of HIV-associated dementia and its relationship to age. Neuroimage. 2004;23(4):1336–47.PubMedCrossRef
17.
Zurück zum Zitat van Gorp WG, Satz P, Hinkin C, Evans G, Miller EN. The neuropsychological aspects of HIV-1 spectrum disease. Psychiatr Med. 1989;7(2):59–78.PubMed van Gorp WG, Satz P, Hinkin C, Evans G, Miller EN. The neuropsychological aspects of HIV-1 spectrum disease. Psychiatr Med. 1989;7(2):59–78.PubMed
18.
Zurück zum Zitat Larussa D, Lorenzini P, Cingolani A, et al. Highly active antiretroviral therapy reduces the age-associated risk of dementia in a cohort of older HIV-1-infected patients. AIDS Res Hum Retroviruses. 2006;22(5):386–92.PubMedCrossRef Larussa D, Lorenzini P, Cingolani A, et al. Highly active antiretroviral therapy reduces the age-associated risk of dementia in a cohort of older HIV-1-infected patients. AIDS Res Hum Retroviruses. 2006;22(5):386–92.PubMedCrossRef
19.
Zurück zum Zitat Valcour V, Shikuma C, Shiramizu B, et al. Higher frequency of dementia in older HIV-1 individuals: the Hawaii Aging with HIV-1 Cohort. Neurology. 2004;63(5):822–7.PubMed Valcour V, Shikuma C, Shiramizu B, et al. Higher frequency of dementia in older HIV-1 individuals: the Hawaii Aging with HIV-1 Cohort. Neurology. 2004;63(5):822–7.PubMed
20.
Zurück zum Zitat Valcour V, Shikuma C, Shiramizu B, et al. Age, apolipoprotein E4, and the risk of HIV dementia: the Hawaii Aging with HIV Cohort. J Neuroimmunol. 2004;157(1–2):197–202.PubMedCrossRef Valcour V, Shikuma C, Shiramizu B, et al. Age, apolipoprotein E4, and the risk of HIV dementia: the Hawaii Aging with HIV Cohort. J Neuroimmunol. 2004;157(1–2):197–202.PubMedCrossRef
21.
Zurück zum Zitat Becker JT, Lopez OL, Dew MA, Aizenstein HJ. Prevalence of cognitive disorders differs as a function of age in HIV virus infection. AIDS. 2004;18(Suppl 1):S11–8.PubMed Becker JT, Lopez OL, Dew MA, Aizenstein HJ. Prevalence of cognitive disorders differs as a function of age in HIV virus infection. AIDS. 2004;18(Suppl 1):S11–8.PubMed
22.
Zurück zum Zitat Kissel EC, Pukay-Martin ND, Bornstein RA. The relationship between age and cognitive function in HIV-infected men. J Neuropsychiatry Clin Neurosci. 2005;17(2):180–4.PubMedCrossRef Kissel EC, Pukay-Martin ND, Bornstein RA. The relationship between age and cognitive function in HIV-infected men. J Neuropsychiatry Clin Neurosci. 2005;17(2):180–4.PubMedCrossRef
23.
Zurück zum Zitat Cherner M, Ellis RJ, Lazzaretto D, et al. Effects of HIV-1 infection and aging on neurobehavioral functioning: preliminary findings. AIDS. 2004;18(Suppl 1):S27–34.PubMed Cherner M, Ellis RJ, Lazzaretto D, et al. Effects of HIV-1 infection and aging on neurobehavioral functioning: preliminary findings. AIDS. 2004;18(Suppl 1):S27–34.PubMed
24.
Zurück zum Zitat Sacktor N, Skolasky R, Selnes OA, et al. Neuropsychological test profile differences between young and old human immunodeficiency virus-positive individuals. J Neurovirol. 2007;13(3):203–9.PubMedCrossRef Sacktor N, Skolasky R, Selnes OA, et al. Neuropsychological test profile differences between young and old human immunodeficiency virus-positive individuals. J Neurovirol. 2007;13(3):203–9.PubMedCrossRef
25.
Zurück zum Zitat Magalhães MG, Greenberg B, Hansen H, Glick M. Comorbidities in older patients with HIV: a retrospective study. J Am Dent Assoc. 2007;138(11):1468–75.PubMed Magalhães MG, Greenberg B, Hansen H, Glick M. Comorbidities in older patients with HIV: a retrospective study. J Am Dent Assoc. 2007;138(11):1468–75.PubMed
26.
Zurück zum Zitat McMurtray A, Nakamoto B, Shikuma C, Valcour V. Small-vessel vascular disease in human immunodeficiency virus infection: the Hawaii Aging with HIV Cohort Study. Cerebrovasc Dis. 2007;24(2–3):236–41.PubMedCrossRef McMurtray A, Nakamoto B, Shikuma C, Valcour V. Small-vessel vascular disease in human immunodeficiency virus infection: the Hawaii Aging with HIV Cohort Study. Cerebrovasc Dis. 2007;24(2–3):236–41.PubMedCrossRef
27.
Zurück zum Zitat Valcour V, Sacktor NC, Paul RH, et al. Insulin resistance is associated with cognition among HIV-1-infected patients: the Hawaii Aging With HIV cohort. J Acquir Immune Defic Syndr. 2006;43(4):405–10.PubMedCrossRef Valcour V, Sacktor NC, Paul RH, et al. Insulin resistance is associated with cognition among HIV-1-infected patients: the Hawaii Aging With HIV cohort. J Acquir Immune Defic Syndr. 2006;43(4):405–10.PubMedCrossRef
28.
Zurück zum Zitat Brew BJ, Crowe SM, Landay A, Cysique LA, Guillemin G. Neurodegeneration and ageing in the HAART era. J Neuroimmune Pharmacol. 2009;4(2):163–74.PubMedCrossRef Brew BJ, Crowe SM, Landay A, Cysique LA, Guillemin G. Neurodegeneration and ageing in the HAART era. J Neuroimmune Pharmacol. 2009;4(2):163–74.PubMedCrossRef
29.
Zurück zum Zitat Valcour V, Paul R. HIV infection and dementia in older adults. Clin Infect Dis. 2006;42(10):1449–54.PubMedCrossRef Valcour V, Paul R. HIV infection and dementia in older adults. Clin Infect Dis. 2006;42(10):1449–54.PubMedCrossRef
30.
Zurück zum Zitat Alisky JM. The coming problem of HIV-associated Alzheimer’s disease. Med Hypotheses. 2007;69(5):1140–3.PubMedCrossRef Alisky JM. The coming problem of HIV-associated Alzheimer’s disease. Med Hypotheses. 2007;69(5):1140–3.PubMedCrossRef
31.
Zurück zum Zitat Brew BJ. Evidence for a change in AIDS dementia complex in the era of highly active antiretroviral therapy and the possibility of new forms of AIDS dementia complex. AIDS. 2004;18(Suppl 1):S75–8.PubMed Brew BJ. Evidence for a change in AIDS dementia complex in the era of highly active antiretroviral therapy and the possibility of new forms of AIDS dementia complex. AIDS. 2004;18(Suppl 1):S75–8.PubMed
32.
Zurück zum Zitat Xu J, Ikezu T. The comorbidity of HIV-associated neurocognitive disorders and Alzheimer’s disease: a foreseeable medical challenge in post-HAART era. J Neuroimmune Pharmacol. 2009;4(2):200–12.PubMedCrossRef Xu J, Ikezu T. The comorbidity of HIV-associated neurocognitive disorders and Alzheimer’s disease: a foreseeable medical challenge in post-HAART era. J Neuroimmune Pharmacol. 2009;4(2):200–12.PubMedCrossRef
33.
Zurück zum Zitat Brew BJ, Pemberton L, Blennow K, Wallin A, Hagberg L. CSF amyloid β42 and tau levels correlate with AIDS dementia complex. Neurology. 2005;65(9):1490–2.PubMedCrossRef Brew BJ, Pemberton L, Blennow K, Wallin A, Hagberg L. CSF amyloid β42 and tau levels correlate with AIDS dementia complex. Neurology. 2005;65(9):1490–2.PubMedCrossRef
34.
Zurück zum Zitat Rempel HC, Pulliam L. HIV-1 Tat inhibits neprilysin and elevates amyloid beta. AIDS. 2005;19(2):127–35.PubMedCrossRef Rempel HC, Pulliam L. HIV-1 Tat inhibits neprilysin and elevates amyloid beta. AIDS. 2005;19(2):127–35.PubMedCrossRef
35.
Zurück zum Zitat Ances BM, Ellis RJ. Dementia and neurocognitive disorders due to HIV-1 infection. Semin Neurol. 2007;27(1):86–92.PubMedCrossRef Ances BM, Ellis RJ. Dementia and neurocognitive disorders due to HIV-1 infection. Semin Neurol. 2007;27(1):86–92.PubMedCrossRef
36.
Zurück zum Zitat Heaton RK, Grant I, Butters N, et al. The HNRC 500—neuropsychology of HIV infection at different disease stages. J Int Neuropsychol Soc. 1995;1(3):231–51.PubMedCrossRef Heaton RK, Grant I, Butters N, et al. The HNRC 500—neuropsychology of HIV infection at different disease stages. J Int Neuropsychol Soc. 1995;1(3):231–51.PubMedCrossRef
37.
Zurück zum Zitat Cysique LA, Maruff P, Brew BJ. Prevalence and pattern of neuropsychological impairment in human immunodeficiency virus-infected/acquired immunodeficiency syndrome (HIV/AIDS) patients across pre- and post-highly active antiretroviral therapy eras: a combined study of two cohorts. J Neurovirol. 2004;10(6):350–7.PubMedCrossRef Cysique LA, Maruff P, Brew BJ. Prevalence and pattern of neuropsychological impairment in human immunodeficiency virus-infected/acquired immunodeficiency syndrome (HIV/AIDS) patients across pre- and post-highly active antiretroviral therapy eras: a combined study of two cohorts. J Neurovirol. 2004;10(6):350–7.PubMedCrossRef
38.
Zurück zum Zitat Scott JC, Woods SP, Patterson KA, et al. Recency effects in HIV-associated dementia are characterized by deficient encoding. Neuropsychologia. 2006;44(8):1336–43.PubMedCrossRef Scott JC, Woods SP, Patterson KA, et al. Recency effects in HIV-associated dementia are characterized by deficient encoding. Neuropsychologia. 2006;44(8):1336–43.PubMedCrossRef
39.
Zurück zum Zitat Maki PM, Cohen MH, Weber K, et al. Impairments in memory and hippocampal function in HIV-positive vs HIV-negative women: a preliminary study. Neurology. 2009;72(19):1661–8.PubMedCrossRef Maki PM, Cohen MH, Weber K, et al. Impairments in memory and hippocampal function in HIV-positive vs HIV-negative women: a preliminary study. Neurology. 2009;72(19):1661–8.PubMedCrossRef
40.
Zurück zum Zitat Thompson PM, Dutton RA, Hayashi KM, Toga AW, Lopez OL, Aizenstein HJ, et al. Thinning of the cerebral cortex visualized in HIV/AIDS reflects CD4+ T lymphocyte decline. Proc Natl Acad Sci USA. 2005;102(43):15647–52.PubMedCrossRef Thompson PM, Dutton RA, Hayashi KM, Toga AW, Lopez OL, Aizenstein HJ, et al. Thinning of the cerebral cortex visualized in HIV/AIDS reflects CD4+ T lymphocyte decline. Proc Natl Acad Sci USA. 2005;102(43):15647–52.PubMedCrossRef
41.
Zurück zum Zitat Hardy DJ, Vance DE. The neuropsychology of HIV/AIDS in older adults. Neuropsychol Rev. 2009;19(2):263–72.PubMedCrossRef Hardy DJ, Vance DE. The neuropsychology of HIV/AIDS in older adults. Neuropsychol Rev. 2009;19(2):263–72.PubMedCrossRef
42.
Zurück zum Zitat Stoff DM. Mental health research in HIV/AIDS and aging: problems and prospects. AIDS. 2004;18(Suppl 1):S3–10.PubMed Stoff DM. Mental health research in HIV/AIDS and aging: problems and prospects. AIDS. 2004;18(Suppl 1):S3–10.PubMed
43.
Zurück zum Zitat The Psychological Corporation. Manual for the Wechsler Test of Adult Reading (WTAR). San Antonio: Author; 2001. The Psychological Corporation. Manual for the Wechsler Test of Adult Reading (WTAR). San Antonio: Author; 2001.
44.
Zurück zum Zitat Power C, Selnes OA, Grim JA, McArthur JC. HIV Dementia Scale: a rapid screening test. J Acquir Immune Defic Syndr Hum Retrovirol. 1995;8(3):273–8.PubMedCrossRef Power C, Selnes OA, Grim JA, McArthur JC. HIV Dementia Scale: a rapid screening test. J Acquir Immune Defic Syndr Hum Retrovirol. 1995;8(3):273–8.PubMedCrossRef
45.
Zurück zum Zitat Goodglass H, Kaplan E, Barresi B. Boston diagnostic aphasia examination. 3rd ed. Philadelphia: Lippincott Williams & Wilkins; 2001. Goodglass H, Kaplan E, Barresi B. Boston diagnostic aphasia examination. 3rd ed. Philadelphia: Lippincott Williams & Wilkins; 2001.
46.
Zurück zum Zitat Benton AL, Hamsher K, Sivan AB. Multilingual aphasia examination. Iowa City: AJA Associates; 1983. Benton AL, Hamsher K, Sivan AB. Multilingual aphasia examination. Iowa City: AJA Associates; 1983.
47.
Zurück zum Zitat Kaufman AS, Kaufman NL. Kaufman adolescent and adult intelligence test. Manual. Circle Pines: American Guidance Service; 1993. Kaufman AS, Kaufman NL. Kaufman adolescent and adult intelligence test. Manual. Circle Pines: American Guidance Service; 1993.
48.
Zurück zum Zitat Wechsler D. Wechsler memory scale—third edition manual. San Antonio: Psychological Corporation; 1997. Wechsler D. Wechsler memory scale—third edition manual. San Antonio: Psychological Corporation; 1997.
49.
Zurück zum Zitat Delis DC, Kramer JH, Kaplan E, Ober BA. The California verbal learning test. 2nd ed. San Antonio: The Psychological Corporation; 2000. Delis DC, Kramer JH, Kaplan E, Ober BA. The California verbal learning test. 2nd ed. San Antonio: The Psychological Corporation; 2000.
50.
Zurück zum Zitat Stern RA, Javorsky DJ, Singer EA, et al. Boston qualitative scoring system for the Rey-Osterrieth complex figure (BQSS). Lutz: Psychological Assessment Resources; 1999. Stern RA, Javorsky DJ, Singer EA, et al. Boston qualitative scoring system for the Rey-Osterrieth complex figure (BQSS). Lutz: Psychological Assessment Resources; 1999.
51.
Zurück zum Zitat Army Individual Test Battery. Manual of directions and scoring. Washington, DC: War Department, Adjutant General’s Office; 1944. Army Individual Test Battery. Manual of directions and scoring. Washington, DC: War Department, Adjutant General’s Office; 1944.
52.
Zurück zum Zitat Culbertson WC, Zillmer EA. The Tower of London, Drexel University, research version: examiner’s manual. North Tonawanda: Multi-Health Systems; 1999. Culbertson WC, Zillmer EA. The Tower of London, Drexel University, research version: examiner’s manual. North Tonawanda: Multi-Health Systems; 1999.
53.
Zurück zum Zitat Massman PJ, Delis DC, Butters N, Dupont RM, Gillin JC. The subcortical dysfunction hypothesis of memory deficits in depression: neuropsychological validation in a subgroup of patients. J Clin Exp Neuropsychol. 1992;14(5):687–706.PubMedCrossRef Massman PJ, Delis DC, Butters N, Dupont RM, Gillin JC. The subcortical dysfunction hypothesis of memory deficits in depression: neuropsychological validation in a subgroup of patients. J Clin Exp Neuropsychol. 1992;14(5):687–706.PubMedCrossRef
54.
Zurück zum Zitat Becker JT, Caldararo R, Lopez OL, Dew MA, Dorst SK, Banks G. Qualitative features of the memory deficit associated with HIV infection and AIDS: cross-validation of a discriminant function classification scheme. J Clin Exp Neuropsychol. 1995;17(1):134–42.PubMedCrossRef Becker JT, Caldararo R, Lopez OL, Dew MA, Dorst SK, Banks G. Qualitative features of the memory deficit associated with HIV infection and AIDS: cross-validation of a discriminant function classification scheme. J Clin Exp Neuropsychol. 1995;17(1):134–42.PubMedCrossRef
55.
Zurück zum Zitat Filoteo JV, Rilling LM, Cole B, Williams BJ, Davis JD, Roberts JW. Variable memory profiles in Parkinson’s disease. J Clin Exp Neuropsychol. 1997;19(6):878–88.PubMedCrossRef Filoteo JV, Rilling LM, Cole B, Williams BJ, Davis JD, Roberts JW. Variable memory profiles in Parkinson’s disease. J Clin Exp Neuropsychol. 1997;19(6):878–88.PubMedCrossRef
56.
Zurück zum Zitat Peavy G, Jacobs D, Salmon DP, et al. Verbal memory performance of patients with human immunodeficiency virus infection: evidence of subcortical dysfunction. J Clin Exp Neuropsychol. 1994;16(4):508–23.PubMedCrossRef Peavy G, Jacobs D, Salmon DP, et al. Verbal memory performance of patients with human immunodeficiency virus infection: evidence of subcortical dysfunction. J Clin Exp Neuropsychol. 1994;16(4):508–23.PubMedCrossRef
57.
Zurück zum Zitat World Health Organization. Composite international diagnostic interview (CIDI, version 2.1). Geneva: World Health Organization; 1998. World Health Organization. Composite international diagnostic interview (CIDI, version 2.1). Geneva: World Health Organization; 1998.
58.
Zurück zum Zitat American Psychiatric Association. Diagnostic and statistical manual of mental disorders—fourth edition: DSM-IV. Washington, DC: American Psychiatric Association; 1994. American Psychiatric Association. Diagnostic and statistical manual of mental disorders—fourth edition: DSM-IV. Washington, DC: American Psychiatric Association; 1994.
59.
Zurück zum Zitat McNair DM, Loor M, Droppleman LF. Profile of Mood States. San Diego: Educational and industrial testing service; 1981. McNair DM, Loor M, Droppleman LF. Profile of Mood States. San Diego: Educational and industrial testing service; 1981.
60.
Zurück zum Zitat Becker JT, Kingsley L, Mullen J, Cohen B, Martin E, Miller EN, et al. Vascular risk factors, HIV serostatus, and cognitive dysfunction in gay and bisexual men. Neurology. 2009;73(16):1292–9.PubMedCrossRef Becker JT, Kingsley L, Mullen J, Cohen B, Martin E, Miller EN, et al. Vascular risk factors, HIV serostatus, and cognitive dysfunction in gay and bisexual men. Neurology. 2009;73(16):1292–9.PubMedCrossRef
61.
Zurück zum Zitat Foley J, Ettenhofer M, Wright MJ, Siddiqi I, Choi M, Thames AD, et al. Neurocognitive functioning in HIV-1 infection: effects of cerebrovascular risk factors and age. Clin Neuropsychol. 2010;24(2):265–85.PubMedCrossRef Foley J, Ettenhofer M, Wright MJ, Siddiqi I, Choi M, Thames AD, et al. Neurocognitive functioning in HIV-1 infection: effects of cerebrovascular risk factors and age. Clin Neuropsychol. 2010;24(2):265–85.PubMedCrossRef
62.
Zurück zum Zitat Barclay TR, Hinkin CH, Castellon SA, et al. Age-associated predictors of medication adherence in HIV-positive adults: health beliefs, self-efficacy, and neurocognitive status. Health Psychol. 2007;26(1):40–9.PubMedCrossRef Barclay TR, Hinkin CH, Castellon SA, et al. Age-associated predictors of medication adherence in HIV-positive adults: health beliefs, self-efficacy, and neurocognitive status. Health Psychol. 2007;26(1):40–9.PubMedCrossRef
63.
Zurück zum Zitat Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991;82(4):239–59.PubMedCrossRef Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991;82(4):239–59.PubMedCrossRef
64.
Zurück zum Zitat Achim CL, Adame A, Dumaop W, Everall IP, Masliah E. Increased accumulation of intraneuronal amyloid beta in HIV-infected patients. J Neuroimmune Pharmacol. 2009;4(2):190–9.PubMedCrossRef Achim CL, Adame A, Dumaop W, Everall IP, Masliah E. Increased accumulation of intraneuronal amyloid beta in HIV-infected patients. J Neuroimmune Pharmacol. 2009;4(2):190–9.PubMedCrossRef
65.
Zurück zum Zitat Sperling R. Functional MRI studies of associative encoding in normal aging, mild cognitive impairment, and Alzheimer’s disease. Ann N Y Acad Sci. 2007;1097:146–55.PubMedCrossRef Sperling R. Functional MRI studies of associative encoding in normal aging, mild cognitive impairment, and Alzheimer’s disease. Ann N Y Acad Sci. 2007;1097:146–55.PubMedCrossRef
66.
Zurück zum Zitat Whitwell JL, Przybelski SA, Weigand SD, et al. 3D maps from multiple MRI illustrate changing atrophy patterns as subjects progress from mild cognitive impairment to Alzheimer’s disease. Brain. 2007;130(Pt 7):1777–86.PubMedCrossRef Whitwell JL, Przybelski SA, Weigand SD, et al. 3D maps from multiple MRI illustrate changing atrophy patterns as subjects progress from mild cognitive impairment to Alzheimer’s disease. Brain. 2007;130(Pt 7):1777–86.PubMedCrossRef
Metadaten
Titel
Neurocognitive Consequences of HIV Infection in Older Adults: An Evaluation of the “Cortical” Hypothesis
verfasst von
J. Cobb Scott
Steven Paul Woods
Catherine L. Carey
Erica Weber
Mark W. Bondi
Igor Grant
The HIV Neurobehavioral Research Center (HNRC) Group
Publikationsdatum
01.08.2011
Verlag
Springer US
Erschienen in
AIDS and Behavior / Ausgabe 6/2011
Print ISSN: 1090-7165
Elektronische ISSN: 1573-3254
DOI
https://doi.org/10.1007/s10461-010-9815-8

Weitere Artikel der Ausgabe 6/2011

AIDS and Behavior 6/2011 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.