ArticlePDF Available

Differential impact of age on verbal memory and executive functioning in chronic kidney disease

Authors:

Abstract and Figures

We compared aspects of verbal memory and executive functioning in 51 community-dwelling persons with chronic kidney disease (CKD) and 55 healthy controls matched on age and education. Depressive symptoms were assessed with the Centre for Epidemiological Studies-Depression Scale (CES-D), and illness variables included glomerular filtration rate (GFR) and hemoglobin. Findings indicate that persons with CKD exhibited poorer performance on measures of memory (CVLT-II) and executive functioning (DKEFS Trailmaking Test B and Color-Word Interference Tests) in comparison with healthy controls. Furthermore, performance decrements were magnified in older CKD participants on measures of verbal memory and inhibition. Nearly half of CKD participants aged 61 and older exhibited significant impairments in verbal memory and inhibition in comparison to matched controls. Cognitive performance in CKD was not associated with measures of illness severity. The differences observed were not accounted for by depressive symptoms, which were only weakly associated with cognitive performance, and negatively associated with age. Findings highlight the need for further exploration of the etiologies and functional consequences of the neuropsychological presentation of CKD.
Content may be subject to copyright.
Differential impact of age on verbal memory and
executive functioning in chronic kidney disease
WENDY LOKEN THORNTON,
1
R. JEAN SHAPIRO,
2
SIRAD DERIA,
1
SHANNON GELB,
1
and AMANDA HILL
2
1Department of Psychology, Simon Fraser University, Burnaby, British Columbia, Canada
2Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
(Received June 21, 2006; Final Revision October 16, 2006; Accepted October 20, 2006)
Abstract
We compared aspects of verbal memory and executive functioning in 51 community-dwelling persons with chronic
kidney disease (CKD) and 55 healthy controls matched on age and education. Depressive symptoms were assessed
with the Centre for Epidemiological Studies-Depression Scale (CES-D), and illness variables included glomerular
filtration rate (GFR) and hemoglobin. Findings indicate that persons with CKD exhibited poorer performance on
measures of memory (CVLT-II) and executive functioning (DKEFS Trailmaking Test B and Color-Word
Interference Tests) in comparison with healthy controls. Furthermore, per formance decrements were magnified in
older CKD participants on measures of verbal memory and inhibition. Nearly half of CKD participants aged 61 and
older exhibited significant impairments in verbal memory and inhibition in comparison to matched controls.
Cognitive per formance in CKD was not associated with measures of illness severity. The differences observed were
not accounted for by depressive symptoms, which were only weakly associated with cognitive per formance, and
negatively associated with age. Findings highlight the need for further exploration of the etiologies and functional
consequences of the neuropsychological presentation of CKD. ( JINS, 2007, 13, 344 –353.)
Keywords: Cerebrovascular disorders, Memory, Cognition, Kidney diseases, Aging, Neuropsychology, Dementia
INTRODUCTION
Chronic Kidney Disease (CKD) is an increasingly common
illness of mid to late adulthood, with an estimated 11% of
adults in the United States affected (Coresh et al., 2003).
The term CKD is used to describe a decrease in renal func-
tion caused by kidney damage, and this illness is typically
managed on an outpatient basis with strict dietary and med-
ication regimens (Levey et al., 2003). Left untreated, CKD
results in the gradual development of renal failure and ure-
mia secondary to the accumulation of neurotoxins and reduc-
tions in metabolic rates (Burn & Bates, 1998). Estimated
glomerular filtration rate (GFR) is considered the best over-
all indicator of level of kidney function, with lower GFR
levels indicating a decrease in the overall number and0or
filtration rate of the kidney nephrons (Levey et al., 2003).
Renal failure eventually results when GFR drops consis-
tently below 15 mL0min per 1.73 m
2
and requires replace-
ment therapy such as dialysis. It is well-documented that
persons with end stage renal disease (ESRD) undergoing
hemodialysis are at considerably higher risk for cognitive
impairment than their age peers, and this has been linked to
metabolic derangements and cerebrovascular diseases asso-
ciated with renal failure (Burn & Bates, 1998; Fazekas et al.,
1996; Fazekas et al., 1995; Kurella et al., 2004; Lass et al.,
1999; Pereira et al., 2005). Those studies that have not
reported cognitive declines in ESRD have likely been lim-
ited by reliance on cognitive screening instruments (Maugeri
et al., 1999), and small samples ( Pliskin et al., 1996; Umans
& Pliskin, 1998).
Despite the fact that the prevalence of the earlier stage of
CKD is more than 100 times greater then that of ESRD
(Collins et al., 2003; Levey et al., 2003), neuropsycholog-
ical functioning in CKD has been virtually ignored to date.
In fact, several factors lead to the prediction that CKD may
be associated with exceptionally high risk for cognitive
impairment even before the development of renal failure.
Diabetes mellitus (Type II adult onset) and hypertension
Correspondence and reprint requests to: Wendy Loken Thornton, Ph.D.,
Department of Psychology, Simon Fraser University, 8888 University Drive,
Burnaby, B.C. V5A 1S6, Canada. E-mail: wthornto@sfu.ca
Journal of the International Neuropsychological Society (2007), 13, 344–353.
Copyright © 2007 INS. Published by Cambridge University Press. Printed in the USA.
DOI: 10.10170S1355617707070361
344
are the leading causes of CKD (Coresh et al., 2003; Levey
et al., 2003). Both of these illnesses have been indepen-
dently associated with development of cerebrovascular dis-
ease (Carmelli et al., 1999; Novak et al., 2006; Raz et al.,
2003), reductions in memory and executive functioning
(Knopman et al., 2001; MacKnight et al., 2002; Perlmuter
et al., 1984; Raz et al., 2003; Saxby et al., 2001), and accel-
erated cognitive decline in late middle-age and older adult-
hood (Hassing et al., 2004; MacKnight et al., 2002; Ott
et al., 1999; Posner et al., 2002). Thus, whereas underlying
cerebrovascular disease is recognized as contributing to the
cognitive presentation of ESRD (Pereira et al., 2005), it is
likely that these effects may emerge earlier in the disease
course as well. Other potential risks for cognitive dysfunc-
tion associated with CKD include elevated levels of homo-
cysteine ( Reutens & Sachdev, 2002), and anemia caused by
decreased production of erythropoietin (Pliskin et al., 2001).
To date, only a few studies have attempted to character-
ize cognitive functioning in persons with CKD. In a cross-
sectional comparison of persons with CKD and persons with
ESRD on hemodialysis, findings suggested a graded rela-
tion between cognitive function and severity of renal dis-
ease, with ESRD participants performing worse than those
with CKD, and persons with CKD performing worse than
published neuropsychological norms on measures of verbal
memory and executive functioning (Trails B; Kurella et al.,
2004). Nonetheless, the inclusion of participants with overt
CNS pathologies confounds the etiology and significance
of the findings. For example, 28% of the CKD sample had
a history of stroke, which may be independently associated
with significant and lasting neuropsychological impair-
ments (Desmond et al., 1999). Similarly, a recent study of
cognition in menopausal women with coronary artery dis-
ease and co-morbid CKD repor ted reductions in neuropsy-
chological performance associated with worsening renal
functioning (Kurella et al., 2005). Whereas these findings
may suggest that specific subgroups of persons with CKD
may be at risk for cognitive impairments, the significance
for the larger CKD population without overt neurological
co-morbidities remains unknown. Furthermore, the restricted
age ranges employed has limited the strength of the conclu-
sions that can be drawn to date. If cerebrovascular disease
contributes toward the cognitive presentation of this ill-
ness, one may expect that the reported reductions in mem-
ory and executive function in CKD may be differentially
exacerbated with increasing age (Hassing et al., 2004; Mac-
Knight et al., 2002).
To address these issues, we assessed cognitive function-
ing in a consecutive outpatient sample of persons with CKD,
and compared their performance with that of healthy con-
trols drawn from the same community. Given previous asso-
ciations between memory0executive functioning and renal
disease, we examined these functions in a CKD sample that
did not have overt CNS pathologies, such as stroke, that
may confound the cognitive presentation. Secondly, we
examined the roles of three potential variables predicted to
have a negative effect on cognitive performance in CKD,
increasing age, a higher number of depressive symptoms,
and increased metabolic derangement (i.e., estimated GFR
and hemoglobin). Toward these ends, we examined (a) the
bivariate associations between age, metabolic derange-
ment, and depressive symptoms in CKD participants, and
(b), the utility of age, health status, and their interaction in
predicting individual differences in verbal memory and exec-
utive functioning before and after controlling for depres-
sive symptoms.
METHODS
Subjects
We tested 51 consecutive outpatients (age 38–89), with a
current diagnosis of CKD who were referred to the Renal
Clinic at Vancouver General Hospital for treatment. Poten-
tial participants were considered to meet criteria for CKD if
they had an average estimated GFR ,60 mL0min 01.73 m
2
and were not receiving renal replacement therapy (i.e., hemo-
dialysis, peritoneal dialysis). Testing of CKD participants
was completed at the Renal Clinic between August 2003
and December 2005. Participants were considered eligible
for inclusion if they met the following criteria: (a) they
were fluent in the English language, (b) had completed a
minimum grade 6 education; and (c) had been followed by
the renal clinic for at least six months to ensure medical
stabilization of acute illness parameters. Participants were
ineligible if they had a history of other major illnesses with
known direct CNS effects (e.g., stroke, head injury, CNS
malignancies, and Parkinson’s disease), or had previously
identified cognitive impairments (e.g., diagnosis of demen-
tia). Additional exclusion criteria included diagnosi s of con-
current terminal illness, major psychiatric illness, or other
major organ failure (e.g., end stage liver disease). Because
of the visual nature of some tasks, we screened partici-
pants’ visual acuity, with the lower limit of corrected vision
set at 20050. Consecutive patients who met these criteria
were invited to participate during their regularly scheduled
clinic visit by a research coordinator. Recruitment success
was approximately 70%. Control participants were recruited
through local community centers and through university
staff union email lists, and were subject to the same exclu-
sion criteria. The study protocol was approved by both the
University of British Columbia and Simon Fraser Univer-
sity research ethics boards, and all participants signed
informed consents.
Measures
Measures were individually administered and scored by
trained research assistants according to standardized proce-
dures. Healthy controls were tested at community testing
sites (i.e., local libraries or community centers) or at the
Simon Fraser University Human Neuropsychology Labora-
tory. Participants received monetary compensation for their
time and travel expenses. Measures included:
Cognition in CKD 345
1. Vocabulary: An untimed multiple choice vocabulary test
was created by combining Vocabulary (V2) and Extended
Vocabulary (V3) from the ETS kit (Ekstrom et al., 1976).
2. Verbal Learning and memory functioning were mea-
sured with the California Verbal Learning Test-2 (CVLT-
II). The CVLT-II assesses learning over repeated trials,
susceptibility to memory interference, and delayed ver-
bal memory. The sum of performance across Trials 1–5
represents the total number of items learned across five
repetitions. Delayed recall on the CVLT-II provides an
estimate of information retained after a 20-minute delay
(Delis et al., 2000).
3. Executive functions were assessed with the Trailmaking
and Color-Word Interference subtests of the Delis-
Kaplan Executive Functioning System ( DKEFS; Delis
et al., 2001). For the Trailmaking test, Trails Test B (letter-
number sequencing) was used as an index of mental set
shifting. In addition to the commonly used versions of
this task, we also utilized the Motor Speed Trails task
(Delis et al., 2001), which requires participants to trace
a dotted line over the Trails path without the numbers or
letters present. We computed a “mental set shifting”
variable by subtracting an individual’s difference in psy-
chomotor speed from the Trails B total score. For the
Color-Word Interference test, we used the difference
between the Inhibition trial and the Color Naming trial
(aka “Stroop effect”) to assess cognitive inhibition abil-
ities while controlling for differences in reading speed.
4. Depressive symptoms were assessed with the Centre for
Epidemiological Studies-Depression Scale (CES-D; Rad-
loff, 1977).
5. A measure of Instrumental Activities of Daily Living
(IADL’s) was included (Lawton & Brody, 1969) to assess
to what extent participants could complete various tasks
(i.e. financial, housekeeping) autonomously, or with assis-
tance. A maximum score of eight reflects autonomy on
all items.
6. All CKD and healthy control par ticipants completed a
health questionnaire to evaluate general medical his-
tory. In persons with CKD, information confirming spe-
cific medical diagnoses, medication regimens, and
duration of illness was obtained from their medical
records. Medical diagnoses (e.g., hypertension, diabetes)
were made by the treating nephrologists. For controls
we followed published recommendations for obtaining
valid and reliable prevalence estimates of target ill-
nesses (Campbell et al., 2005) (i.e., to be considered
hypertensive, participants must report a diagnosis of
“high blood pressure” and currently be prescribed anti-
hypertensive medication).
The state of metabolic compensation was determined by
examining blood test results obtained during the most recent
clinic visit, and testing was scheduled to ensure that blood
work was obtained within two weeks of neuropsychologi-
cal testing.The following parameters were considered: hemo-
globin concentration to assess the degree of anemia and
estimated glomerular filtration rate (GFR) to assess clear-
ance capacity of nitrogenous wastes. Estimated GFR was
calculated with the widely used 4-variable equation (Levey
et al., 1999).
Statistical Analysis
Sample differences across the demographic, affective, and
cognitive variables were examined with independent sam-
ple t-tests or nonparametric tests ( Pearson x
2
) where appro-
priate. For purposes of sample description and presentation,
age 60 was determined as a cut-off for middle-aged (30
60), and older (61–89) age groups. To assess learning and
memory abilities, two measures were selected from the
CVLT-II, the total number of words remembered across the
5 trials, and the number of words correctly recalled after a
20-minute delay. The mean performances on these mea-
sures are presented in Table 2. Because these two measures
were highly correlated within the full sample (r5.78), a
composite “verbal memory” T-score combining these mea-
sures was generated based on the control sample means,
and used in all subsequent analyses to reduce the number of
dependent variables. The two measures of executive func-
tioning (inhibition and mental set shifting) were treated inde-
pendently for the following reasons. First, because executive
functioning has received little previous attention in CKD,
we wished to expand our assessment to include two aspects
of executive functions, mental set shifting and inhibition
(Miyake et al., 2000). Secondly, these measures exhibited
only a modest relationship ( r5.55), and were thus not
considered redundant.
For the regression analyses, results of evaluation of
assumptions led to logarithmic transformation of the CES-D
variable to reduce skewness and improve the normality of
the distribution. For all participants, distributions were exam-
ined for extreme values and departures from normality. Data
from 2 participants (1 CKD and 1 control) were identified
as extreme values for the Mental Set Shifting variable, and
two (both CKD) were identified as extreme values for the
Inhibition variable. Consequently, the data was evaluated
after adjusting these extreme values to make them contig-
uous with the next closest value while maintaining the rank
ordering of the distribution (see Tabachnick & Fidell, 2007).
As this adjustment did not significantly impact the regres-
sion findings, the results are presented on the non-adjusted
values. The continuous independent variable (Age) was cen-
tered to reduce multicollinearity in examination of the inter-
action term. To facilitate presentation of the relevant findings
in the Figures, T-scores (mean of 50, SD of 10) were cal-
culated for the dependent variables to place these scores on
a consistent metric. Analyses were conducted using SPSS
14 software (SPSS Inc. Chicago, Il).
346 W.L. Thornton et al.
RESULTS
Subject Characteristics
Information regarding demographic and illness characteris-
tics is presented in Table 1. The 51 CKD and 55 control
participants were well matched on age and education, and
they did not significantly differ in their gender composi-
tion, or on a measure of verbal knowledge (Vocabulary). As
expected, persons with CKD had significantly higher rates
of diabetes, hypertension, and hypercholesterolemia than
controls (see Table 1). All CKD participants scored 7 or 8
on a measure of IADL’s (Lawton & Brody, 1969; mean
7.89, SD 5.37), and all controls obtained a maximal score
of 8 on this measure. Thus, all participants can be consid-
ered functionally independent for daily living skills.
Severity and Moderators of Cognitive
Impairment in CKD
The percentage of CKD participants considered to meet
criteria for cognitive impairment was calculated using the
recommended cut-off score of performance at least 1.5 SD’s
below matched controls ( Tuokko et al., 2001). As can be
seen in Table 2, the estimates of cognitive impairment in
CKD varied with age. In younger CKD participants (ages
38– 60), estimates of cognitive impairments ranged from
0% to 16%, with the largest performance differences noted
on the delayed recall measure. In older CKD par ticipants
(ages 61–89), estimates of cognitive impairment ranged from
27% to 46%, with largest performance differences again
noted on the delayed recall measure. Overall, CKD partici-
pants reported a higher number of depressive symptoms
than healthy controls, although when stratified by age group,
these differences only reached significance in younger par-
ticipants (see Table 2). As can be seen in Table 3, increasing
age was associated with worse performance on all of the
cognitive tasks in CKD participants. Importantly, age was
not significantly associated with increased severity of ill-
ness as indexed by either estimated GFR (Pearson r5 2.04)
or hemoglobin (Pearson r5.05). Furthermore, neither hemo-
globin nor estimated GFR were significantly correlated with
cognitive performance.
To examine the predictive utility of age, health status,
and their interaction in accounting for individual differ-
ences in verbal memory and executive functioning, we con-
ducted a series of two-step hierarchical regression analyses,
the results of which are presented in Table 4. The first step
of the model included participant’s age (centered), group
(CKD vs. controls) and the interaction of these variables. In
addition, because persons with CKD reported a higher level
of depressive symptoms than controls, we wanted to deter-
mine whether this variable improved prediction of perfor-
mance on the cognitive measures. Therefore, scores on the
CES-D (log) were entered into the second step of the model.
As can be seen in Table 4, increasing age, CKD, and their
interaction were each significant predictors of poorer per-
formance on measures of verbal memory and inhibition.
Together, these variables explained 38% of the variance in
verbal memory performance with 95% confidence limits
from .21 to .51 (Steiger & Fouladi, 1992), and 37% of the
variance in Stroop (inhibition) performance (95% confi-
Table 1. Demographic and clinical variables
Participant characteristics
CKD
(n551)
Controls
(n555) p-value*
Age (mean 6SD) 63.24 613.57 60.53 615.15 ns
Female % 27 (53%) 34 (62%) ns
Education (mean years 6SD) 13.41 63.16 14.13 62.29 ns
Vocabulary 21.69 69.98 24.92 67.26 ns
Hypertension % 46 (90%) 16 (29%) ,.001
Diabetes mellitus % 16 (31%) 0 (0%) ,.001
Benzodiazepines % 5 (10%) 1 (2%) ns
Opiates % 3 (6%) 0 (0%) ns
CKD participants
Duration of renal disease (yrs.) 5.56 66.68
Stage of renal disease† (%)
Stage 3 (GFR 30–59) 12 (24%) Age range 540–83
Stage 4 (GFR 15–29) 28 (55%) Age range 540–84
Stage 5 (GFR ,14) 11 (22%) Age range 538–89
GFR 24.11 611.05
Hemoglobin (g0L) 124.06 612.61
*: p-value obtained from independent sample t-tests of group means; Vocabulary 5ETS V2 1
V3; †—5 stage model of kidney disease recommended by the Kidney Disease Outcome Quality
Initiative Practice Guidelines; GFR 5glomerular filtration rate (mL 0min 01.73m2).
Cognition in CKD 347
dence limits from .20 to .50; Steiger & Fouladi, 1992).
Both older age and CKD were associated with worse per-
formance on these measures. Furthermore, examination of
the regression slopes revealed that age-related performance
decrements were differentially exacerbated in CKD partici-
pants. These relationships can be observed in Figures 1
and 2. Whereas older age and diagnosis of CKD were pre-
dictive of worse performance on the mental set shifting
task (R
2
5.35; confidence limits from .18 to .48; Steiger &
Fouladi, 1992), the interaction of these variables was non-
contributory (see Fig. 3).
Depressive symptoms were not a significant predictor of
either verbal memory or mental set shifting performance
(R
2
change 5.000, and .001, respectively). However, de-
pressive symptoms were a significant predictor of Stroop
performance, resulting in an additional 3% of variance
explained. Interestingly, the direction of this relationship
was negative, as a lesser number of depressive symptoms
predicted greater inter ference on the Stroop task. To eluci-
date this relationship, the age by depressive symptoms”
and “group by depressive symptoms” interaction terms were
entered into two hierarchical regression models along with
Table 2. Verbal memory, executive functioning, and depressive symptoms by age and group
Measures (mean 6SD) CKD Healthy controls Effect size* p-value** CKD impaired
CVLT-II Trial 1–5 Total
All 40.69 69.63 48.38 69.90 .79 ,.001
Younger 46.20 66.67 49.53 610.05 .38 ns n50
Older 35.39 69.12 47.00 69.73 1.23 ,.001 n511 (42%)
CVLT-II delayed recall
All 8.26 63.52 11.33 63.20 .91 ,.001
Younger 9.96 62.35 11.50 63.18 .54 ,.05 n54 (16%)
Older 6.62 63.71 11.12 63.28 1.28 ,.001 n512 (46%)
Mental set shifting
All 80.39 645.43 61.55 633.98 .47 ,.05
Younger 55.04 626.66 49.37 623.04 .23 ns n53 (12%)
Older 104.77 646.75 76.16 639.34 .66 ,.05 n57 (27%)
Inhibition
All 34.53 615.38 27.05 610.35 .57 ,.01
Younger 25.68 67.86 24.17 68.57 .18 ns n53 (12%)
Older 43.04 616.14 30.52 611.41 .89 ,.005 n511 (42%)
CES-D
All 10.59 66.71 6.06 66.06 .70 ,.001
Younger 12.56 67.50 6.13 65.63 .97 ,.005
Older 8.69 65.33 5.96 66.66 .45 ns
Note. For comparisons of all participants, n551 for CKD and 55 for Controls groups, for Younger group comparisons,
n525 for CKD and 30 for Controls, for older group comparisons, n526 for CKD and 25 for Controls; CVLT-II items: more
words r etained 5better performance; mental set shifting (Trails B-Motor Speed ) 5time to completion (lowe r scores 5better
performance); inhibition (aka “Stroop effect”) 5difference between inhibition trial and color naming trial ( lower scores 5
better per formance); *: Effect size 5Cohen’s d(mean 1 2mean 2 0pooled SD), and all effects favor controls; **: p-value
obtained from independent sample t-tests o f group mea ns, †: % I mpaired 5percentage falling 1.5 or more SD below the mean
of the controls, CES-D 5Centre for Epidemiological Studies-Depression Scale.
Table 3. Correlations between illness variables, and cognitive variables in CKD par ticipants
Variable Age 2 3 4 5 6 7 8
1Age
2Education 2.22
3CVLT 1–5 2.63** .33*
4CVLT LD 2.55** .54** .77**
5Inhibition .59** 2.19 2.46** 2.44**
6Mental set shifting .54** 2.42** 2.56** 2.62** .54**
7CES-D 2.26 .23 .11 .13 2.32* 2.06
8GFR 2.04 .04 .21 .21 2.14 .01 2.17
9Hemoglobin .05 .03 2.17 .03 2.02 2.01 2.08 .30*
*p52-tailed correlation ,.05, **p,.01; GFR 5glomerular filtration rate (mL 0min01.73m2; higher 5better renal func-
tioning).
348 W.L. Thornton et al.
Table 4. Hierarchical regression results predicting verbal
memory, mental set shifting, and inhibition in persons
with CKD and healthy controls
Predictors B SE bp R
2
Verbal memory
Age 2.19 .073 2.27 ,.05
Group (CKD0Controls) 27.65 1.60 2.38 ,.001
Age by group 2.25 .11 2.23 ,.05
R
2
* .38
Mental set shifting
Age .32 .07 .46 ,.001
Group (CKD0Controls) 3.58 1.60 .18 ,.05
Age by group .12 .11 .11 ns
R
2
* .35
Inhibition: Step 1
Age .25 .07 .36 ,.005
Group (CKD0Controls) 4.52 1.58 .23 ,.01
Age by group .24 .11 .23 ,.05
R
2
.37
Inhibition: Step 2
Age .25 .07 .35 ,.005
Group (CKD0Controls) 5.85 1.68 .29 ,.005
Age by group .22 .11 .21 ,.05
Depressive symptoms 24.42 2.12 2.18 ,.05
R
2
.39
DR
2
.02
*Step 2 did not result in a significant increase in R2
Fig. 1. Regression slopes and scatter plot showing the effects of
Age, Group (CKD and Controls), and their interaction on the ver-
bal memory measure.
Fig. 2. Regression slopes and scatter plot showing the effects of
Age, Group (CKD and Controls), and their interaction on the Inhi-
bition measure.
Fig. 3. Regression slopes and scatter plot showing the effects of
Age and Group (CKD and Controls) on the Mental Set Shifting
task.
Cognition in CKD 349
the appropriate main effects, and were non-contributory.
Examination of the intercorrelations revealed that in CKD
participants, a higher number of depressive symptoms were
significantly associated with better performance on the Inhi-
bition task ( Pearson r5 2.32; see Table 3). Furthermore, it
was noted that this might reflect the fact that increasing age
was marginally associated with a reduction in depression
symptoms ( Pearson r5 2.26, p5.07). In fact, after con-
trolling for the effects of age through partial correlation, the
relationship between depressive symptoms and inhibition
was reduced to non-significance ( pr 5 2.22).
Finally, in CKD participants only, we conducted a series
of hierarchical regression analyses to determine the predic-
tive utility of metabolic factors (estimated GFR and hemo-
globin) after adjusting for the effects of age, which is
embedded in the MDRD GFR estimate. As expected, increas-
ing age emerged as a significant predictor of worse perfor-
mance on each of the cognitive dependent measures, whereas
the inclusion of the metabolic factors added no further pre-
diction over and above the effects of age.
DISCUSSION
Little attention to date has been directed toward cognitive
functioning in persons with CKD in the years prior to the
initiation of dialysis. We examined the pattern of cognitive
performance in a Canadian CKD sample with a broad range
of age and illness severity. In addition, we examined the
roles of three potential variables predicted to have a nega-
tive effect on cognitive performance in CKD, increasing
age, a higher number of depressive symptoms, and increased
illness severity (as indexed by estimated GFR and hemo-
globin). The current evidence extends previous findings
(Kurella et al., 2004) by demonstrating that even persons
with CKD who do not present with overt evidence of cere-
brovascular pathology (e.g., stroke) are at high risk for cog-
nitive impairments in comparison with age peers. In addition,
our findings suggest that age is an important moderator of
the cognitive presentation in CKD. Whereas cer tain aspects
of executive functioning and learning may be relatively pre-
served in younger CKD participants, older CKD partici-
pants exhibited worse per formance than matched controls
on all aspects of memory and executive functioning assessed
in the current study. Furthermore, examination of the effect
size estimates indicates that the magnitude of the cognitive
differences observed is moderate to large (see Table 2) with
very large differences noted in verbal memory in older CKD
participants (Cohen, 1992).
An important question remains, what may be underlying
these significant cognitive impairments in persons with
CKD? The current findings allow us to first approach this
question by ruling out some potentially important vari-
ables. Whereas CKD participants exhibited more self-
reported depressive symptoms than controls, these were only
weakly associated with cognitive performance after con-
trolling for the effects of age. Furthermore, we could find
no evidence for an association between cognitive perfor-
mance and illness progression in persons with CKD. Regard-
ing the effects of age, it is important to note that there was
no indication in the current sample of a direct association
between increasing age and our indices of severity of renal
disease. Specifically, whereas increasing age was associ-
ated with greater extent and severity of impairments in ver-
bal memory and inhibition, age was not significantly
associated with either estimated GFR (Pearson r5 2.04)
or hemoglobin (Pearson r5.05). Fur thermore, age was
evenly distributed across the stages of renal disease repre-
sented in the current sample (see Table 1).
To understand the age effects in this study, it is important
to note that we excluded participants with a previous diag-
nosis of dementia, and all participants were considered inde-
pendent for activities of daily living based on their IADL
performance. Nonetheless, the fact that older CKD partici-
pants exhibited such high rates of cognitive impairments
suggests that these results may reflect emergent cerebrovas-
cular disease that may exert a negative effect on cognition
even in the absence of overt symptoms such as stroke. As
reported previously and observed in the current sample,
hypertension is nearly ubiquitous in CKD (Levey et al.,
2003). Furthermore, hypertension and diabetes together form
the leading causes of renal disease (Collins et al., 2003;
Coresh et al., 2003), and both have been associated with
accelerated cognitive decline in late mid-age and late-life
(Abate et al., 2001; Gunning-Dixon & Raz, 2000; Hassing
et al., 2004; Knopman et al., 2001; MacKnight et al., 2002).
Whereas there has been little direct neuroradiological exam-
ination to date in persons with CKD, it has recently been
argued that cerebrovascular disease may be an important
cause of cognitive impairment in persons with the more
severe form of renal failure known as ESRD (Pereira et al.,
2005). Recent brain imaging studies have reported high
rates of cerebrovascular disease in persons with ESRD, with
50% to 80% exhibiting white matter hyperintensities (WMH)
and other evidence of ischemic brain lesions (Fazekas et al.,
1996; Fazekas et al., 1995; Lass et al., 1999). The presence
of WMH’s has been associated with cerebrovascular dis-
ease (Stewar t, 1999) and with impairments in executive
functioning and memory in elderly populations (Gunning-
Dixon & Raz, 2000). In addition, CKD is associated with
several other cerebrovascular risk factors, such as athero-
sclerosis, hyperhomocysteinemia, and oxidative stress (Leon-
cini et al., 2003; Pereira et al., 2005). Thus, the pattern of
memory and executive functioning impairments we observe
in older CKD par ticipants may in part reflect what has pre-
viously been deemed “vascular cognitive impairment
(Hachinski, 1994), in patient populations with known
cerebrovascular risks. Such an explanation may underlie
previously reported associations between moderate renal
impairment and vascular dementia in older samples (Seliger
et al., 2004). An important direction for future research will
be to further elucidate the relative contributions of these
and other potential cognitive risk factors in CKD. Further-
more, it will be important to track cognitive performance
longitudinally in persons with CKD to determine how declin-
350 W.L. Thornton et al.
ing cognitive status may influence treatment decisions and
outcomes.
Limitations
These findings should be considered in light of certain lim-
itations. It is possible that our decision to exclude individ-
uals with other illnesses with CNS effects (i.e., stroke and
dementia) may reduce the generalizability of our findings
to the broader CKD population. In fact, it is likely that more
liberal inclusion would have led to even higher rates of
significant impairment observed. Nonetheless, we felt our
criteria allowed more precise elucidation of the cognitive
presentation of CKD. In addition, because we did not have
access to laboratory blood work on our control participants,
it is it possible that some of our “healthy controls” may
have had undetected renal disease (i.e., reduced GFR). In
any case, the inclusion of such individuals in the control
group would be expected to reduce the magnitude of the
differences observed and would not serve to confound inter-
pretation of the current results.
Regarding the dependent measures employed, we elected
to specifically examine verbal memory and aspects of exec-
utive functioning. The reasons for this were threefold. First,
a decline in these functions has previously been reported in
CKD samples with a large percentage of participants with a
history of stroke. We wished to extend these findings to
CKD participants without a history of stroke to better char-
acterize the pervasiveness and nature of the impairments.
Secondly, it is important to note that our measures of exec-
utive functioning were derived after controlling for the effects
of both psychomotor speed and reading speed. Thus, we
believe we have reported a more accurate and nuanced pic-
ture of the true nature of the deficits associated with CKD.
Thirdly, these functions have been shown to be particularly
sensitive to cerebrovascular risks that are highly prevalent
in this population. We acknowledge that postulating that
cerebrovascular disease may underlie at least some of the
deficits observed is speculative, but consistent with previ-
ous findings indicating high rates of cerebrovascular pathol-
ogy in renal disease (for a review, see Pereira et al., 2005).
Developing a direct link between cerebral integrity and func-
tion in CKD remains an important question for future
research.
Summary and Conclusions
In summary, our findings suggest that community-dwelling
adults with CKD display high rates of cognitive impair-
ments relative to matched controls. Furthermore, these
impairments are most pronounced in persons aged 61 and
older and are not associated with metabolic illness param-
eters such as estimated GFR or hemoglobin. The differ-
ences observed were not accounted for by depressive
symptoms, which were only weakly associated with cogni-
tive performance, and negatively associated with age. With
more than half of persons with renal disease over age 60
(Coresh et al., 2003), the current findings have important
implications for medical management of CKD, and high-
light the need for further neuropsychological investigation
of this highly prevalent and costly illness. Developing a
better understanding of the factors underlying neuropsycho-
logical per formance in CKD is crucial to detect persons
that may be at risk for cognitive compromise years before
the initiation of dialysis, to assist in treatment planning, to
identify and treat potentially reversible causes of cognitive
impairment, and to provide essential support to patients
required to manage increasingly complex medication and
dietary regimens.
ACKNOWLEDGMENTS
The authors thank Heike Dumke, Stacey Kesten, and Sarah
Meachen for their help in data collection, Teri Pentland for her
efforts in subject recruitment and study coordination, Dr. Ray Koop-
man for statistical consultation, and Dr. Allen E. Thornton for his
helpful comments on an earlier version of this manuscript. The
authors also address special thanks to those who agreed to partici-
pate in this study. This work was supported in part by a Simon
Fraser University0Social Sciences and Humanities Research Coun-
cil (SFU0SSHRC) Institutional Grant and an SFU0Institute of
Health Research and Education (SFU0IHRE) Seed Grant awarded
to Dr. Wendy Loken Thornton, and by a University of British
Columbia ( UBC) Division of Geriatric Medicine Grant awarded
to Dr.’s Thornton and Hill. Sirad Deria was supported by the For-
mation de Chercheurs et l’Aide a la Recherche - Fonds de la
recherche en santé du Québec Master’s Scholarship ( FCAR-
FRSQ), and by a Canadian Institute of Health Research (CIHR)
Professional Student Research Award. Shannon Gelb was sup-
ported by a SSHRC Canada Graduate Master’s Scholarship and
by a CIHR 0Michael Smith Foundation for Health Research
(MSFRH) Transplant Research Training Award. Portions of this
work were presented in abstract form at the 2nd Annual Congress
of the International Society for Vascular, Cognitive, and Behav-
ioural Disorders, and at the Gerontological Society of America’s
56th Annual Scientific Meeting.
REFERENCES
Abate, G., Zito, M., Ferrari-Ramondo, V., & Di Iorio, A. (2001).
Blood pressure and dementia: A review. Archives of Gerontol-
ogy and Geriatrics,33(Suppl. 7), 7–18.
Burn, D.J. & Bates, D. (1998). Neurology and the kidney. Journal
of Neurology, Neurosurgery, and Psychiatry,65, 810 –821.
Campbell, N.R.C., Joffres, M.R., & McKay, D.W. (2005). Hyper-
tension surveillance in Canada: Minimum standards for assess-
ing blood pressure in surveys. Canadian Journal of Public
Health,96, 217–220.
Carmelli, D., Swan, G.E., Reed, T., Wolf, P.A., Miller, B.L., &
DeCarli, C. (1999). Midlife cardiovascular risk factors and brain
morphology in identical older male twins. Neurology,52,
1119–1124.
Cohen, J. (1992). A power primer. Psychological Bulletin,112,
155–159.
Collins, A.J., Kasiske, B., Herzog, C., Chen, S.C., Everson, S.,
Constantini, E., Grimm, R., McBean, M., Xue, J., Chavers, B.,
Matas, A., Manning, W., Louis, T., Pan, W., Liu, J., Suying, L.,
Cognition in CKD 351
Roberts, T., Dalleska, F., Snyder, J., Ebben, J., Frazier, E., Sheets,
D., Johnson, R., Li, S., Dunning, S., Berrini, D., Guo, H., Solid,
C., Arko, C., Daniels, F., Wang, X., Forrest, B., Gilbertson, D.,
St. Peter, W., Frederick, P., Eggers, P., & Agodoa, L. (2003).
U.S. Renal Data System. Excerpts from the USRDS 2002
Annual Data Report: Atlas of end-stage renal disease in the
United States. American Journal of Kidney Disease,42(Suppl.
6), 1–260.
Coresh, J., Astor, B.C., Greene, T., Eknoyan, G., & Levey, A.S.
(2003). Prevalence of chronic kidney disease and decreased
kidney function in the adult US population: Third National
Health and Nutrition Examination Survey. American Journal
of Kidney Disease,41, 1–12.
Delis, D., Kramer, J.A., Kaplan, E., & Ober, B. (2000). The Cali-
fornia Verbal Learning Test (2nd ed.) adult version. San Antonio,
TX: The Psychological Corporation.
Delis, D.C., Kaplan, E., & Kramer, J.H. (2001). Delis-Kaplan Exec-
utive Function System. San Antonio, TX: The Psychological
Corporation.
Desmond, D.W., Erkinjuntti, T., Sano, M., Cummings, J.L., Bowler,
J.V., Pasquier, F., Moroney, J.T., Ferris, S.H., Stern, Y., Sach-
dev, P.S., & Hachinski, V.C. (1999). The cognitive syndrome
of vascular dementia: Implications for clinical trials. Alzhei-
mer’s Disease and Associated Disorders,13 (Suppl. 3), 21–29.
Ekstrom, R.B., Harmon, D., & Dermen, D. (1976). Manual for kit
of factor-referenced cognitive tests. Princeton, NJ: Educational
Testing Services.
Fazekas, G., Fazekas, F., Schmidt, R., Flooh, E., Valetitsch, H.,
Kapeller, P., & Krejs, G.J. (1996). Pattern of cerebral blood
flow and cognition in patients undergoing chronic haemodial-
ysis treatment. Nuclear Medicine Communications,17, 85–88.
Fazekas, G., Fazekas, F., Schmidt, R., Kapeller, P., Offenbacher,
H., & Krejs, G.J. (1995). Brain MRI findings and cognitive
impairment in patients undergoing chronic hemodialysis treat-
ment. Journal of the Neurological Sciences,134, 85–88.
Gunning-Dixon, F. & Raz, N. (2000). The cognitive correlates of
white matter abnormalities in normal aging: A quantitative
review. Neuropsychology,14, 224 –232.
Hachinski, V. (1994). Vascular dementia: A radical redefinition.
Dementia,5, 130 –132.
Hassing, L.B., Hofer, S.M., Nilsson, S.E., Berg, S., Pedersen, N.L.,
McClearn, G., & Johansson, B. (2004). Comorbid type 2 dia-
betes mellitus and hypertension exacerbates cognitive decline:
Evidence from a longitudinal study. Age and Ageing,33,
355–361.
Knopman, D., Boland, L.L., Moseley, T., Howard, G., Liao, D.,
Szklo, M., McGovern, P., & Folsom, A.R. (2001). Cardiovas-
cular risk factors and cognitive decline in middle-aged adults.
Neurology,56, 42– 48.
Kurella, M., Chertow, G.M., Luan, J., & Yaffe, K. (2004). Cogni-
tive impairment in chronic kidney disease. Journal of the Amer-
ican Geriatric Society,52, 1863–1869.
Kurella, M., Yaffe, K., Shlipak, M.G., Wenger, N.K., & Chertow,
G.M. (2005). Chronic kidney disease and cognitive impair-
ment in menopausal women. American Journal of Kidney Dis-
ease,1, 66–76.
Lass, P., Buscombe, J.R., Harber, M., Davenport, A., & Hilson,
J.W. (1999). Cognitive impairment in patients with renal fail-
ure is associated with multiple-infarct dementia. Clinical
Nuclear Medicine,24, 561–565.
Lawton, M.O. & Brody, E.M. (1969). Assessment of older people:
Self-maintaining and instrumental activities of daily living.
Gerontologist,9, 179–186.
Leoncini, G., Viazzi, F., Parodi, D., Vettoretti, S., Ratto, E., Ra-
vera, M., Tomolillo, C., Del Sette, M., Bezante, G.P., Defer-
rari, G., & Pontremoli, R. (2003). Mild renal dysfunction and
subclinical cardiovascular damage in primary hypertension.
Hypertension,42, 14–18.
Levey, A.S., Bosch, J.P., Lewis, J.B., Greene, T., Rogers, N., &
Roth, D. (1999). A more accurate method to estimate glomer-
ular filtration rate from serum creatinine: A new prediction
equation. Modification of Diet in Renal Disease Study Group.
Annals of Internal Medicine,130, 41– 49.
Levey, A.S., Coresh, J., Balk, E., Kausz, A.T., Levin, A., Steffes,
M.W., Hogg, R.J., Perrone, R.D., Lau, J., & Eknoyan, G. (2003).
National Kidney Foundation practice guidelines for chronic
kidney disease: Evaluation, classification, and stratification.
Annals of Internal Medicine,139, 137–147.
MacKnight, C., Rockwood, K., Awalt, E., & McDowell, I. (2002).
Diabetes mellitus and the risk of dementia, Alzheimer’s dis-
ease, and vascular cognitive impairment in the Canadian Study
of Health and Aging. Dementia and Geriatric Cognitive Dis-
orders,14, 77–82.
Maugeri, D., Malaguarnera, M., Panebianco, P., Barbagallo, P.,
Curasi, M.P., Bonanno, M.R., Speciale, S., Santangelo, A., &
Russo, M.S. (1999). Assessment of cognitive and affective dis-
orders in an elderly population undergoing hemodialysis.
Archives of Gerontology and Geriatrics,29, 239–248.
Miyake, A., Friedman, N.P., Emerson, M.J., Witzki, A.H., & Ho-
werter, A. (2000). The unity and diversity of executive func-
tions and their contributions to complex “frontal lobe” tasks: A
latent variable analysis. Cognitive Psychology,41, 49–100.
Novak, V., Last, D., Alsop, D.C., Abduljalil, A.M., Hu, K.,
Lepicovsky, L., Ca vallerano, J. & Lipsitz, L.A. (2006).
Cerebral blood flow velocity and periventricular white matter
hyperintensities in type 2 diabetes. Diabe tes Care,29,
1529–1534.
Ott, A., Stolk, R.P., van Harskamp, F., Pols, H.A.P., Hofman, A.,
& Breteler, M.M.B. (1999). Diabetes mellitus and the risk of
dementia. Neurology,53, 1937–1942.
Pereira, A.A., Weiner, D.E., Scott, T., & Sarnak, M.J. (2005). Cog-
nitive function in dialysis patients. American Journal of Kid-
ney Disease,45, 448– 462.
Perlmuter, L.C., Hakima, M.K., Hodgson-Harrington, C., Gins-
berg, J., Katz, J., Singer, D.E., & Nathan, D.M. (1984). Decreased
cognitive functioning in aging non-insulin dependent diabetes
patients. American Journal of Medicine,77, 1043–1048.
Pliskin, N.H., Kiolbasa, T.A., Hart, R.P., & Umans, J.G. (2001).
Neuropsychological function in renal disease and its treat-
ment. In S.R. Beers (Ed.), Medical Neuropsychology, 2nd edi-
tion ( pp. 107–126). New York: Kluwer Academic0Plenum
Publishers.
Pliskin, N.H., Yurk, H.M., Ho, L.T., & Umans, J.G. (1996). Neuro-
cognitive function in chronic hemodialysis patients. Kidney
International,49, 1435–1440.
Posner, H.B., Tang, M.X., Luchsinger, J., Lantigua, R., Stern, Y.,
& Mayeux, R. (2002). The relationship of hypertension in the
elderly to AD, vascular dementia, and cognitive function. Neu-
rology,58, 1175–1181.
Radloff, L.S. (1977). A self-report depression scale for research in
general populations. Applied Psychological Measurement,1,
385– 401.
352 W.L. Thornton et al.
Raz, N., Rodrigues, K.M., & Acker, J.D. (2003). Hypertension
and the brain: Vulnerability of prefrontal regions and executive
functions. Behavioral Neuroscience,117, 1169–1180.
Reutens, S. & Sachdev, P. (2002). Homocysteine in neuropsychi-
atric disorders of the elderly. International Journal of Geriat-
ric Psychiatry,17, 859–864.
Saxby, B.K., Harrington, F., McKeith, I.G., Wesnes, K., & Ford,
G.A. (2001). Effects of hypertension on attention, memory,
and executive function in older adults. Health Psychology,22,
587–591.
Seliger, S.L., Siscovick, D.S., Stehman-Breen, C.O., Gillen, D.L.,
Fitzpatrick, A., Bleyer, A., & Kuller, L.H. (2004). Moderate
renal impairment and risk of dementia among older adults: The
Cardio-vascular Health Cognition Study. Journal of the Amer-
ican Society of Nephrology,15, 1904 –1911.
Steiger, J.H. & Fouladi, R.T. (1992). R2: A computer program for
interval estimation, power calculation, and hypothesis testing
for the squared multiple correlation. Behaviour Research Meth-
ods, Instruments, and Computers,4, 581–582.
Stewart, R. (1999). Hypertension and cognitive decline. British
Journal of Psychiatry,174, 286 –287.
Tabachnick, B.G., & Fidell, L.S. (2007). Using Multivariate Sta-
tistics (5th ed.). Boston: Allyn and Bacon.
Tuokko, H.A., Frerichs, J., & Kristjansoon, B. (2001). Cognitive
impairment, no dementia: Concepts and issues. International
Psychogeriatrics,13, 183–202.
Umans, J.G. & Pliskin, N.H. (1998). Attention and mental process-
ing speed in hemodialysis patients. American Journal of Kid-
ney Disease,32, 749–751.
Cognition in CKD 353
... In terms of comorbid conditions, seven studies (Dixit et al., 2013;Egbi, Ogunrin, & Oviasu, 2015;Nasser, Shawki, El Shahawy, & Sany, 2012;Owolabi et al., 2016;Pi et al., 2016) excluded individuals with diabetes mellitus and/or cardiac risk or history. Five studies did not exclude these and reported rates of presentation (Hailpern, Melamed, Cohen, & Hostetter, 2007;Owolabi et al., 2016;Sanchez-Roman et al., 2011;Thornton, Shapiro, Deria, Gelb, & Hill, 2007;Tsai, Wang, & Fuh, 2010), with three conducting analyses comparing them between the groups and all finding no significant differences. One other study reporting diabetes controlled for its presence in their analyses (Silverwood et al., 2014). ...
... One other study reporting diabetes controlled for its presence in their analyses (Silverwood et al., 2014). Several other studies (Bae & Park, 2008;Dixit et al., 2013;Egbi et al., 2015;Nasser et al., 2012;Owolabi et al., 2016;Pi et al., 2016;Sanchez-Roman et al., 2011;Thornton et al., 2007) also excluded participants with other comorbidities such as history of Parkinson's disease and/or cerebrovascular diseases, hypertension, and head injuries. Two studies (Chen et al., 2015;Gad, Ramzy, Abdelhamid, ElMassry, & Masoud, 2012) did not report any related conditions to CKD. ...
... Two of these studies have been discussed above Cognitive impairment (CI %) was significantly poorer for ESKD compared to undialyzed stage 3-5 group in executive function, delayed memory, visuospatial ability, and overall CI Nasser et al. ( Hailpern et al., 2007;Tsai et al., 2010). Of the remaining seven studies, four compared the differences between two groups; a healthy cohort and a clinical group of patients with both stage 3 and 4 CKD inclusive (Gad et al., 2012;Nasser et al., 2012;Sanchez-Roman et al., 2011), and a healthy cohort and a clinical group of patients with stage 3 to 5 CKD (Thornton et al., 2007). Gad et al (2012) found that a moderate CKD group performed significantly worse than the (2012), reported moderate CKD participants to have significantly poorer scores (d = 3.34 to 6.33) than healthy controls in general cognition, processing speed, switching, and working memory (MMSE, TMT-B, D-SYM, and digit span). ...
Article
Full-text available
Objectives: Recognition of cognitive impairment in chronic kidney disease (CKD) and its impact on functioning in adults is growing. The vast majority of studies to date have been conducted in older populations where CKD is more pronounced; however, the degree to which age-related cognitive changes could be influencing these findings remains unaddressed. This current study thus aimed to review cognitive impairment findings by stage in non-elderly CKD samples. Methods: PubMed and Medline via Scopus were searched for cross-sectional or cohort studies and randomized controlled trials that assessed cognitive function in individuals with CKD in any research setting. CKD studies including patients at any illness stage were included providing participants were below 65 years old, were not on peritoneal dialysis and had not undergone a kidney transplant. Results: Fifteen studies, with a total of 9304 participants, were included. Cognitive function broadly deteriorated from stage 1 to stage 5. Early stage CKD was associated with a drop in speed of processing, attention, response speed, and short-term memory abilities. Moderate stage CKD was associated with deficits in executive functioning, verbal fluency, logical memory, orientation and concentration. People with end stage kidney disease manifested significant deficits in all previous cognitive domains, along with cognitive control, delayed and immediate memory, visuospatial impairment, and overall cognitive impairment. Conclusions: Cognitive impairment is evident across the stages of CKD, independent of age-related changes, for both lower-order and higher-order cognitive abilities. These impairments also increase between the stages, suggesting a cumulative effect. Future directions for research are discussed. (JINS, 2018, 22, 1-14).
... However, another risk factor that remains relatively understudied to date involves the role of cognition. Indeed, relative to their healthy peers, adults with chronic kidney disease (CKD) demonstrate weaknesses in memory and executive functioning across the course of their illness [7,8] and following successful transplant [9,10]. These cognitive difficulties in CKD and RTR have been theorized to stem from cerebrovascular insufficiencies commonly reported in CKD and in dialysis patients that may not be reversed with transplantation [11]. ...
... Historically, cognitive functioning in chronic illness populations has focused on traditional neurocognitive abilities (i.e., learning, memory, and executive functions [7,8,10]). Notably, there is emerging interest in the role of "everyday" cognition, specifically, everyday problem solving (EPS) in health behaviors. While performance on EPS tasks may in part rely on traditional abilities such as executive function and episodic memory [17,18], EPS appears to represent a distinct aspect of cognition with additive utility in predicting real world outcomes, including self-rated functioning, life-skills functioning, and mortality [19][20][21]. ...
... Neurocognitive measures. Well-established tasks assessing multiple cognitive domains were utilized to provide measurement of aspects of cognition commonly examined in RTR [7,8,10]. The Kaufman Brief Intelligence Test (KBIT-2 [53]) assesses verbal and non-verbal intellectual functioning. ...
Article
Full-text available
Objective Estimates indicate that 20–70% of renal transplant recipients are medication non-adherent, significantly increasing the risk of organ rejection. Medication adherence is negatively impacted by lower everyday problem solving ability, and associations between depressive symptoms, self-efficacy, and adherence are reported in renal transplant recipients. Nonetheless, to date, these associations have not been examined concurrently. Given the relationship between non-adherence and organ rejection, it is critical to gain a better understanding of the predictors of adherence in renal transplant recipients. To this end, we modeled relationships among cognitive abilities, depressive symptoms, self-efficacy, and adherence in this group. Methods Participants (N = 211) underwent renal transplant at least one year prior to participation. Adherence was measured via self-report, medication possession ratio, and immunosuppressant blood-level. Traditionally-measured neurocognitive and everyday problem-solving abilities were assessed. Depressive symptoms were measured via self-report, as were general and medication adherence related self-efficacy. Structural equation modeling was used to assess the fit of the model to available data. Results Everyday problem solving and self-efficacy had direct positive associations with adherence. Depressive symptoms were negatively associated with self-efficacy, but not adherence. Traditionally-measured neurocognitive abilities were positively associated with self-efficacy, and negatively associated with depressive symptoms. Conclusions We present a comprehensive investigation of relationships between cognitive and psychosocial factors and adherence in medically stable renal transplant recipients. Findings confirm the importance of everyday problem solving and self-efficacy in predicting adherence and suggest that influences of depressive symptoms and neurocognitive abilities are indirect. Findings have important implications for future development of interventions to improve medication adherence in renal transplant recipients.
... With the aging of society, the morbidity and mortality of CKD and the risk of cognitive impairment are increasing in countries with long life expectancy [33]. This study found that CKD increased the risk of impairment of memory, executive function, information processing speed, and sustained attention, in line with the previous literature [34][35][36]. A prospective cohort study demonstrated that kidney dysfunction was associated with higher cognitive impairment, even in subjects with early-stage CKD [37]. ...
Article
Full-text available
Background Cognitive dysfunction is prevalent among the elderly diagnosed with chronic kidney disease (CKD). Low protein diets are used for retarding the progression of CKD in clinical practice. Nonetheless, the impact of dietary protein consumption on cognitive function in this population remains uncertain. Methods We recruited 2306 participants (≥60 years) from 2011 to 2014 National Health and Nutrition Examination Survey (NHANES). 24-h dietary recall questionnaire was utilized to evaluate protein intake. Cognitive function was measured using the Consortium to Establish a Registry for Alzheimer’s Disease (CERAD), Animal Fluency Test (AFT), and Digit Symbol Substitution Test (DSST). Participants’ characteristics were analyzed, and the interaction between protein consumption and CKD on cognitive impairment were analyzed using a logistic regression model. Results We divided participants into three groups based on CKD stages: no CKD, CKD stage G1 to G2 (19%), and CKD stage G3 to G5 (18%). The average protein intake was 0.97 g/(kg·d). In the higher protein intake group, CKD stages G1 to G2 elevated the risk of immediate memory impairment (OR: 2.441, 95% Cl: 1.161–5.132 for protein consumption in 1.0-1.2g/(kg·d); OR: 2.225, 95% Cl: 1.015–4.876 for protein consumption in >1.2 g/(kg·d)). However, no similar resuts were observed in the lower protein intake group. In addition, the interaction between CKD status and protein intake on immediate memory was statistically significant (p = .041). Conclusion A higher probability of cognitive impairment in the elderly with early-stage CKD may be linked to higher protein intake. Low protein diets may be a potential strategy to release cognitive impairment in the elderly with early-stage CKD.
... In laboratory studies of cognition, ESRD has been associated with multiple impairments (Hart & Kreutzer, 1988), for example, of visual attention (Etgen et al., 2009), executive functioning (Jassal, Devins, Chan, Bozanovic, & Rourke, 2006) and psychomotor speed (Griva et al., 2003). In addition, various aspects of memory have been shown to be impaired: verbal and visual memory (Elias et al., 2009;Kurella, Chertow, Luan, & Yaffe, 2004), working memory (Buchman et al., 2009), episodic memory (Thornton, Shapiro, Deria, Gelb, & Hill, 2007) and conceptual memory (Jones et al., 2015b). However, it is uncertain whether patients are aware of these impairments, and to what extent they impact on everyday life. ...
Article
Introduction: End-stage renal disease (ESRD) has been associated with a range of cognitive deficits, including impaired retrospective memory and attention. Prospective memory (PM) is memory for future intentions, such as remembering to take medication on time. Prospective memory has not been examined in any ESRD patients; yet, the implications upon diet and medication management could potentially have detrimental effects on patient welfare. This is the first study to examine PM in ESRD patients being treated with hemodialysis (HD). Methods: Hemodialysis patients (n = 18) were compared with age-matched and education-matched controls (n = 18) on a boardgame task that emulates a typical week of activities (i.e., grocery shopping, meetings with friends), requiring the participant to remember a series of upcoming tasks. Other measures were also examined, including general cognitive decline, measures of independent living, IQ, and mood. Findings: Patients recalled significantly fewer upcoming events than the control group, suggesting an impairment of PM. No significant relationship was found between PM performance and any other measures, suggesting the difference between groups is likely due to the effects of ESRD, HD treatment, or some associated comorbidity. Discussion: This is the first study to demonstrate a PM deficit in patients undergoing HD treatment. This finding contributes to the current knowledge of the cognitive profile of patients undergoing HD while also highlighting the implications that a PM deficit may have on patient quality of life. The finding may go some way to explaining variances in patients' ability to monitor and adhere to medication and dietary regimes and, ultimately, to live independently. The study also highlights the necessity of viewing treatment for ESRD as a holistic process to maximize patient well-being.
... visuospatial and constructional abilities. Attentional control and executive functioning are domains often impaired in most chronic, systemic diseases [38][39][40]. ...
Article
Full-text available
Background: The cornerstone of effective management in heart failure (HF) is the ability to self-care. Aims include i) To determine factors influencing self-care in HF patients with cognitive impairment (CI) and ii) to determine the influence of cognitive domains on self-care in patients with HF and CI. Methods: MEDLINE, CINAHL, EMBASE, EBSCOHost, PsychINFO, ProQuest Research Library, Health Technology Assessment Database, The Cochrane Library, Web of Science and Scopus databases were systematically searched. Original research describing the relationship between cognition and HF self-care in community-dwelling older persons with dementia/CI in English, published in a peer-reviewed journal from 1stJanuary(2000)-22ndMarch(2016) was identified. Study and population characteristics, data sources, self-care processes, methods of cognitive assessment, cognitive domains affected, study outcomes, impact of impairment, and other risk factors of self-care impairment were abstracted by two reviewers. Results: Of 10,688 studies identified, 14 met the inclusion criteria. Patients with HF and CI ranged from 14 to 73%. Where reported, self-care maintenance adequacy ranged from 50 to 61%; self-care management adequacy ranged from 14 to 36% and self-care confidence adequacy ranged from 0 to 44% on the Self-care of Heart Failure Index (SCHFI). All but one study predicted poor self-care ability according to poor outcome on cognitive testing. Additionally, specific cognitive domain deficits impaired self-care. Subjects with lower cognitive scores were less likely to seek assistance while subjects with depression had poor self-care abilities. Conclusions: Clinicians must consider the type and severity of impairments in cognitive domains to tailor management. Awareness of depression, self-confidence and support access may modulate self-care ability.
... CKD patients present several comorbidities, such as immune disorders and neurological complications, which contribute to the morbidity and mortality of these patients [4][5][6]. CKD is frequently associated with cognitive impairments, and it has been estimated that more than 85% of hemodialyzed (HD) patients with terminal CKD have cognitive deficits [7][8][9][10]. The neurological disease in ...
Article
Full-text available
Chronic kidney disease (CKD) involves multiple organ dysfunction, and the neurological complications that are often present in CKD patients support the idea of a crosstalk between the kidneys and the brain. Evidence suggests a possible role for products accumulating in these patients as uremic toxins in various CKD complications, including neurodegeneration. Indoxyl sulfate (IS), derived from tryptophan metabolism, is well-known as a uremic nephron-vascular toxin, and recent evidence suggests it also has a role in the immune response and in neurodegeneration. Inflammation has been associated with neurodegenerative diseases, as well as with CKD. In this study, we demonstrated that sera of CKD patients induced a significant inflammation in astrocyte cells which was proportional to IS sera concentrations, and that the IS adsorbent, AST-120, reduced this inflammatory response. These results indicated that, among the uremic toxins accumulating in serum of CKD patients, IS significantly contributed to astrocyte inflammation. Moreover, being also chronic inflammation associated with CKD, here we reported that IS further increased inflammation and oxidative stress in primary central nervous system (CNS) cells, via Nuclear Factor-κB (NF-κB) and Aryl hydrocarbon Receptor (AhR) activation, and induced neuron death. This study is a step towards elucidating IS as a potential pharmacological target in CKD patients.
... In an investigation of the effects of HD frequency on cognitive performance, Kurella Tamura et al. [39] found that higher HD frequency did not improve EFs, suggesting that residual uremia is not the main factor responsible for neuropsychological impairment in these patients. However, the negative effect of uremic status on cognitive performance is well documented [1,40,41]. In conclusion, the contribution of urea to executive functioning in ESRD-HD patients remains unclear, and is likely mediated by other biochemical-clinical variables and nutritional status. ...
Article
Full-text available
Objective There is evidence of cognitive impairment in patients with end-stage renal disease in hemodialysis (ESRD-HD). However, few studies have exhaustively analyzed executive functions (EFs) in this population, especially considering the influence of a wide range of clinical variables. This study analyzes performance in different EF components in ESRD-HD patients compared to a group of healthy controls (HCs), in addition to the acute effects of HD and the associations of cognitive performance with clinical variables. Method EFs were evaluated pre- and post-HD in 43 ESRD-HD patients and 42 HCs, using a battery of tests designed to assess EF domains. Age, schooling, mood and blood pressure were statistically controlled. Associations between performance and clinical factors were computed by correlations and hierarchical multiple regression analyses. Results The performance of the ESRD-HD patients was significantly lower than that of HCs in all the EF domains except for planning. Group differences were marginally significant for reasoning. HD produced no acute changes in global performance, with improvements see only in inhibition and working memory. EF scores were positively associated with total number of months previously transplanted, body mass index (BMI), dry weight, and levels of hemoglobin, albumin, ferritin, calcium, phosphorus, sodium, urea, and creatinine. Conclusions Global EF functioning was lower in ESRD-HD patients than in HCs. No major acute HD-related EF changes were detected. These findings underline the importance of an adequate nutritional status for maintaining executive functioning in ESRD-HD patients.
Article
Background: Patients with chronic kidney disease on hemodialysis (HD) often have cognitive deficits. However, there are few studies that have examined the neuropsychological impairments of patients receiving peritoneal dialysis (PD). Methods: Executive functions, processing speed and verbal memory were assessed in 27 PD patients, 42 HD patients, and 42 healthy participants (HP). Systolic blood pressure and total time on renal replacement therapy (RRT) were controlled statistically. Associations between performance and clinical factors were analyzed using correlations and multiple regression. Results: The DP group showed better performance compared to the HD group in verbal fluency, working memory, cognitive flexibility, planning and decision making. The DP group showed worse execution than the HP group in verbal inhibition and memory. Executive function scores were positively associated with total months on PD, total months on RRT, total months on HD, albumin, total cholesterol, and phosphorus, and negatively with ferritin. Conclusion: Global executive functioning was more optimal in PD patients than in HD patients. The results show the positive effect of PD on executive functions, which must be taken into account when choosing the TRS. The associations observed between biochemical factors and performance show the importance of maintaining an adequate nutritional status in these patients.
Article
Objective The Delis-Kaplan Executive Function System (D-KEFS) is a battery of tests designed to measure executive functions (EF). Additionally, the D-KEFS contains lower-order tasks, designed to control for speed of visual scanning, sequencing, and verbal and graphomotor output. The construct and criterion validities of D-KEFS scores that are time-based are well established. However, the constructs measured by the D-KEFS error scores are poorly understood, making clinical interpretations of such scores difficult. This study examined the construct validity of D-KEFS errors committed on EF tasks and tasks designed to measure lower-order processes (i.e., non-EF tasks), across the adult lifespan. Method Participants were 427 adults (18–93 years) who completed the timed subtests of the D-KEFS. Four hundred two participants also completed the Push-Turn-Taptap (PTT; a separate measure of EF) to allow cross-validation. Results General linear regressions showed that D-KEFS errors committed on the EF tests were associated with EF timed performance (assessed using the D-KEFS time-based scores and the PTT), but only among older adults. Importantly, errors committed on the D-KEFS tasks of lower-order processes were also associated with D-KEFS time-based EF performance, and this relationship held across the adult lifespan. Conclusions These findings suggest that among older adults EF errors on the D-KEFS can be interpreted as indices of EF, but such interpretations are not automatically warranted for younger adults. Additionally, errors committed on non-EF tasks contained within the D-KEFS battery can be interpreted as reflecting EF weaknesses across the adult lifespan.
Chapter
Our understanding of neuropsychological function in end-stage renal disease (ESRD) and chronic hemodialysis has evolved a great deal in the past 30 years. Early studies in the 1960s and 1970s documented the neurocognitive effects of uremia and chronic renal failure, while investigations in the 1970s and 1980s focused on neurocognitive outcome following initiation of hemodialysis, neuropsychological status in chronic hemodialysis and peritoneal dialysis, and correction for anemia and aluminum toxicity. In the 12 years since this chapter was first published, medical standards of dialysis adequacy have changed dramatically. Thus, many patients studied in the 1960s to 1980s may have been relatively underdialyzed at the time of assessment compared to current standards for dialysis prescription (Lowrie, Laird, Parker, & Sargent, 1981; Gotch & Sargent, 1985; National Kidney Foundation, 1997a, 1997b). Recent investigations quantifying dialysis delivery have provided new insights into the neuropsychological effects of dialysis. Several investigations into the effects of renal transplantation on cognition also have appeared within the past decade.
Article
Background: Serum creatinine concentration is widely used as an index of renal function, but this concentration is affected by factors other than glomerular filtration rate (GFR). Objective: To develop an equation to predict GFR from serum creatinine concentration and other factors. Design: Cross-sectional study of GFR, creatinine clearance, serum creatinine concentration, and demographic and clinical characteristics in patients with chronic renal disease. Patients: 1628 patients enrolled in the baseline period of the Modification of Diet in Renal Disease (MDRD) Study, of whom 1070 were randomly selected as the training sample ; the remaining 558 patients constituted the validation sample. Methods: The prediction equation was developed by stepwise regression applied to the training sample. The equation was then tested and compared with other prediction equations in the validation sample. Results: To simplify prediction of GFR, the equation included only demographic and serum variables. Independent factors associated with a lower GFR included a higher serum creatinine concentration, older age, female sex, nonblack ethnicity, higher serum urea nitrogen levels, and lower serum albumin levels (P < 0.001 for all factors). The multiple regression model explained 90.3% of the variance in the logarithm of GFR in the validation sample. Measured creatinine clearance overestimated GFR by 19%, and creatinine clearance predicted by the Cockcroft-Gault formula overestimated GFR by 16%. After adjustment for this overestimation, the percentage of variance of the logarithm of GFR predicted by measured creatinine clearance or the Cockcroft-Gault formula was 86.6% and 84.2%, respectively. Conclusion: The equation developed from the MDRD Study provided a more accurate estimate of GFR in our study group than measured creatinine clearance or other commonly used equations.
Article
The MDRD study equation has many advantages. It is more accurate and precise than the Cockcroft–Gault equation for persons with a GFR less than approximately 90 mL/min per 1.73 m²(34–35). This equation predicts GFR as measured by using an accepted method (urinary clearance of ¹²⁵I-iothalamate). It was developed on a large (n > 1000) database containing persons with various kidney diseases and was tested on a validation database containing more than 500 additional patients. It does not require height or weight and has been validated in kidney transplant recipients and African-Americans with nephrosclerosis (36). Nonetheless, questions remain about the equation's generalizability because it has not been validated in diabetic kidney disease, in patients with serious comorbid conditions, in normal persons, or in persons older than 70 years of age. Clinical conditions in which it may be necessary to measure GFR by using clearance methods include extremes of age and body size, severe malnutrition or obesity, diseases of skeletal muscle, paraplegia or quadriplegia, vegetarian diet, rapidly changing kidney function, and calculation of the dose of potentially toxic drugs that are excreted by the kidneys. Chronic kidney disease affects approximately 11% of the U.S. adult population (20 million people from 1988 to 1994). The prevalence of earlier stages of disease (10.8%) is more than 100 times greater than the prevalence of kidney failure (0.1%). Adverse outcomes of chronic kidney disease, including loss of kidney function and development of kidney failure and CVD, can often be prevented or delayed through early detection and treatment. In particular, physicians should consider using interventions to slow the progression of kidney disease in all patients with chronic kidney disease and should place patients with chronic kidney disease in the highest-risk group for CVD risk factor reduction and other treatments for CVD. Each patient with chronic kidney disease should have a clinical action plan, based on the stage of disease, as defined by the NKF K/DOQI guidelines. All patients with chronic kidney disease and persons at increased risk for chronic kidney disease should undergo measurement of proteinuria (as a marker of kidney damage) and GFR. Quantitative assessment of proteinuria is useful for detection, differential diagnosis, prognosis, and treatment of chronic kidney disease. The ratio of concentration of albumin to creatinine in untimed urine samples should be used to detect and monitor proteinuria. Glomerular filtration rate, as estimated by prediction equations based on serum creatinine concentration, age, race, sex, and body size, is useful for detecting chronic kidney disease, classifying its severity, estimating progression, managing complications, and deciding on referral to a nephrologist.