Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Archives of Gerontology and Geriatrics 75 (2018) 20–27

Contents lists available at ScienceDirect

Archives of Gerontology and Geriatrics


journal homepage: www.elsevier.com/locate/archger

Full Length Article

Association of nutrition and immune-endocrine dysfunction with muscle T


mass and performance in cognitively impaired older adults

L. Taya, , B.P. Leungb, S. Weec, K.S. Tayb, N. Alic,d, M. Chanc,d, W.S. Limc,d
a
Department of General Medicine, Sengkang General Hospital, Singapore
b
Department of Rheumatology, Allergy and Immunology, Tan Tock Seng Hospital, Singapore
c
Institute of Geriatrics and Active Ageing, Tan Tock Seng Hospital, Singapore
d
Department of Geriatric Medicine, Tan Tock Seng Hospital, Singapore

A R T I C L E I N F O A B S T R A C T

Keywords: Background: With lean mass declining early in Alzheimer’s disease, muscle quality beyond quantity is relevant to
Sarcopenia physical performance. We sought to identify potentially modifiable factors for the differential loss of muscle
Cognitive impairment mass (pre-sarcopenia) and its performance (sarcopenia) in older adults with mild cognitive impairment (MCI)
Inflammation and mild-to-moderate Alzheimer’s disease (AD).
Endocrine
Methods: This is a cross-sectional study of 108 community-dwelling older adults with MCI and mild-to-moderate
AD. Participants were categorized as: (i) No sarcopenia (normal muscle mass), (ii) Pre-sarcopenia (low muscle
mass without weakness or slowness), (iii) Sarcopenia (low muscle mass AND weak grip strength and/or slow gait
speed) using Asian cut-offs. Muscle quality was defined as the ratio of grip and knee extension strength to
average arm and leg lean mass respectively. We measured cognitive, functional and physical (Short Physical
Performance Battery, SPPB) performance; physical activity level; nutritional status; and blood biomarkers of
inflammation and endocrine dysfunction.
Results: SPPB (p = 0.033) and activity level (p = 0.010) were highest in the pre-sarcopenic group. Pre-sarco-
penic group had highest arm muscle quality [10.6 (7.7–12.2) vs 13.9 (12.6–15.7) vs 11.3 (9.7–12.8),
p < 0.001], despite significantly lower appendicular lean mass than non-sarcopenic group. In multi-nomial
logistic regression reference to non-sarcopenic group, malnutrition independently increased risk for both pre-
sarcopenia (Relative risk = 7.53, 95% C.I 1.20–47.51, p = 0.032) and sarcopenia (Relative risk = 11.91, 95%
C.I 2.85–49.77, p = 0.001). A combined pro-inflammatory and endocrine deficient state significantly increased
the risk of sarcopenia (Relative risk = 5.17, 95% C.I 1.31–20.37, p = 0.019).
Conclusion: Malnutrition is a precursor for progressive loss of muscle mass, but a pro-inflammatory and endo-
crine deficient state may potentially aggravate decline in muscle quality to culminate in frank sarcopenia.

1. Introduction There is substantial overlap between the physical frailty phenotype


and sarcopenia, which is widely operationalized as decreased muscle
Body composition changes with progressive weight loss have been function accompanying the decline in muscle mass with advancing age,
observed in the earliest clinical stages of Alzheimer’s disease (AD) and and spans a continuum through pre-sarcopenia (low muscle mass),
may precede onset of dementia, being driven predominantly by the loss sarcopenia (low muscle mass with low muscle strength or poor per-
of lean mass (Burns, Johnson, Watts, Swerdlow, & Brooks, 2010; formance), and severe sarcopenia (low muscle mass with low muscle
Johnson, Wilkin, & Morris, 2006). Weight loss, along with declines in strength and poor performance) (Cruz-Jentoft et al., 2010). Indeed,
muscle mass and performance, feature amongst the major components sarcopenia may be considered the biological substrate for the devel-
of the physical frailty syndrome (Fried, Ferruci, Darer, Williamson, & opment of physical frailty. There is, however, dissociation between the
Anderson, 2004) which has been recognized as a distinct entity asso- loss of muscle mass and that of muscle strength, with higher rate of
ciated with aggravated risk for adverse outcomes amongst cognitively decline in muscle strength and its greater impact on future disability
impaired older adults (Bilotta et al., 2012; Ni Mhaolain et al., 2012; relative to muscle mass (Visser et al., 2005; von Haehling, Morley, &
Oosterveld et al., 2014). Anker, 2010). In addition, muscle strength but not muscle mass


Corresponding author at: Department of General Medicine, Sengkang General Hospital, 378 Alexandra Road, 159964, Singapore.
E-mail address: laura.tay.b.g@singhealth.com.sg (L. Tay).

https://doi.org/10.1016/j.archger.2017.11.008
Received 12 July 2017; Received in revised form 8 October 2017; Accepted 14 November 2017
Available online 16 November 2017
0167-4943/ © 2017 Elsevier B.V. All rights reserved.
L. Tay et al. Archives of Gerontology and Geriatrics 75 (2018) 20–27

independently predicted mortality in older adults (Newman et al., CDR of 0.5, 1 or 2, corresponding to very mild, mild or moderate de-
2006). This observed discrepancy between muscle mass and muscle mentia respectively. We excluded subjects with a diagnosis of possible
strength has shifted attention towards muscle quality as a more clini- AD in view of the confounding co-morbid diagnoses and differing
cally relevant determinant of physical performance in older adults clinical course in these individuals.
(Barbat-Artigas, Rolland, Zamboni, & Aubertin-Leheudre, 2012).
The consequences of muscle mass and strength declines extend be- 2.1.2. Eligibility criteria
yond physical limitations, having been linked to cognitive impairment Subjects were eligible if they were aged > 55 years, with a diag-
and dementia (Nourhaehemi et al., 2002; Shin, Kim, Kim, Shin, & Yoon, nosis of MCI or mild to moderate AD at baseline, community-dwelling,
2002). Further, low muscle quality as represented by strength per unit and accompanied by a reliable informant.
muscle mass has been associated with poorer cognitive performance We excluded subjects with presence of other central nervous con-
(Canon & Crimmins, 2011). The significant association between sar- ditions (stroke disease, Parkinson’s disease, subdural hematoma,
copenia and cognitive impairment raises the possibility of shared normal pressure hydrocephalus, and brain tumor); presence of systemic
common pathways (Chang, Hsu, Wu, Huang, & Han, 2016). Endocrine conditions that can contribute to cognitive impairment (hypothyr-
systems involved in muscle anabolism such as insulin-like growth factor oidism, B12 deficiency, and hypercalcaemia); and presence of any ac-
1 (IGF-1) and dehydroepiandrosterone sulphate (DHEAS) have also tive neuropsychiatric conditions producing disability. Subjects living in
been implicated in age-related cognitive decline (Cruz-Jentoft et al., a sheltered or nursing home were also excluded.
2014; Landi et al., 2012; Maggio et al., 2012; Watanabe et al., 2005), The validity of the overall cognitive evaluation process and CDR
while a chronic inflammatory state has been linked to both muscle scoring has been previously established (Chong and Sahadevan, 2003;
catabolism and AD (Holmes et al., 2009; Schaap et al., 2009). Beyond Lim, Chin, Lam, Lim, & Sahadevan, 2005). Laboratory investigations to
their individual detrimental effects, the potential for additive effects of exclude potentially reversible causes of dementia via blood tests and
inflammation and endocrine dysregulations has also been suggested neuroimaging were done. A multidisciplinary consensus meeting was
(Cappola et al., 2008). conducted to review all relevant results for accurate clinical pheno-
The observed association of sarcopenia but not pre-sarcopenia with typing of the cognitive disorder (MCI or mild-moderate probable AD).
dual impairments in cognitive and physical performance may be espe- Patients meeting study eligibility criteria were then recruited.
cially pertinent in cognitively impaired older adults (Tolea & Galvin,
2015), and suggests sarcopenia as a risk factor for accelerating disease 2.2. Measures
progression and disability. Compared with sarcopenic individuals who
are less likely to transition out, the pre-sarcopenic state with its greater 2.2.1. Sarcopenia assessment and muscle quality
potential for reversibility offers an intervention target to avoid pro- Grip strength was measured using the hydraulic hand dynamometer
gression (Murphy et al., 2014). Little is known about risk factors that (North Coast@ Hydraulic Hand Dynamometer), with two trials of grip
differentiate pre-sarcopenia from non-sarcopenia and sarcopenia states, strength for each hand and all 4 trials averaged to yield strength. Knee
although the inconsistent translation of gains in muscle mass to im- extensor muscle strength of each leg was measured twice using an
provements in muscle strength raises the hypothesis for differential electronic dynamometer (BASELINE PUSHPULL Dynamometer), with
pathways underlying muscle mass and function (Schroddder et al., the participant seated at the edge of a chair and maintaining the trunk
2012). Thus, this study sought to identify potentially modifiable factors in the upright position. The average of the 4 readings provided a
associated with the differential loss of muscle mass (pre-sarcopenia) measure of knee extension strength. Gait speed was based on the time to
and its performance (sarcopenia) in cognitively impaired older adults. walk 3m.
Percentage body fat and lean mass measures were obtained via a
2. Methods dual-energy X-ray absorptiometry system (Discovery™ APEX 13.3;
Hologic, Bedford, MA, USA). Appendicular skeletal mass was derived
2.1. Study population from the summation of muscle mass measurements in the four limbs.
We adopted the European Working Group on Sarcopenia in Older
This is a cross-sectional analysis of community-dwelling older adults People (EWGSOP) consensus criteria (Cruz-Jentoft et al., 2010), but
with mild cognitive impairment (MCI) and mild-moderate Alzheimer’s employing Asian gender-specific cut-off values for muscle mass, grip
dementia (AD) attending a tertiary Memory Clinic, Tan Tock Seng strength and gait speed (Chen et al., 2014), to delineate 3 groups: (i) No
Hospital, Singapore. Informed written consent was obtained from the sarcopenia (normal muscle mass), (ii) Pre-sarcopenia (low muscle mass
patient or legally acceptable representative where appropriate, and the without impact on muscle strength or gait speed), (iii) Sarcopenia (low
study was approved by the Domain Specific Review Board (DSRB) of muscle mass AND weak grip strength and/or slow gait speed).
the National Healthcare Group (NHG). A measure of muscle quality was derived for each individual from
the ratio of grip and knee extension strength to average arm and leg
2.1.1. Diagnostic categories lean mass respectively.
MCI was defined as follows: (1) global Clinical Dementia Rating
(CDR) (Morris, 1993) score of 0.5; (2) presence of subjective memory 2.2.2. Cognitive assessment
complaint which was corroborated by a reliable informant; (3) delayed We used the CDR (Morris, 1993), a structured clinician rating, to
recall > 1 SD below the age and education-adjusted means of healthy determine dementia severity. The CDR is a global dementia rating scale
community-dwelling subjects derived from an earlier normative study and ratings in each of the six domains can be summed for a CDR sum-of-
(Sahadevan, Lim, Tan, & Chan, 2002); (4) relatively normal general boxes (CDR-SB) score (range 0–18). The attending geriatrician, trained
cognitive function, defined as a score ≥21 for subjects with ≤6 years in administration of the CDR, rated each patient’s CDR at baseline.
education and ≥24 for those with > 6 years of education on the locally Cognitive performance was assessed using the locally-validated
validated modified Chinese version of Mini Mental State Examination modified Chinese version of Mini-Mental State Examination (CMMSE),
(CMMSE) (Sahadevan, Lim, Tan, & Chan, 2000), (5) largely intact ac- and MCI subjects also underwent a neuropsychological assessment
tivities of daily living; and (6) no clinical dementia. (Sahadevan et al., 2000).
Mild-moderate AD subjects fulfilled National Institute of
Neurological and Communicative Disorders and Stroke and the 2.2.3. Other clinical co-variates
Alzheimer’s Disease and Related Disorders Association (NINCDS- Demographic data and co-morbid vascular risk factors – hyperten-
ADRDA) criteria for probable AD (McKhann et al., 1984), with global sion, hyperlipidemia, diabetes mellitus, atrial fibrillation, peripheral

21
L. Tay et al. Archives of Gerontology and Geriatrics 75 (2018) 20–27

vascular disease, smoking history, ischemic heart disease – were cap- subsequently categorized as (i) neither pro-inflammatory nor endocrine
tured at baseline. The presence of hypertension, hyperlipidaemia, dia- deficient, (ii) pro-inflammatory (IL-6 or TNF-R1, or both, being in
betes mellitus, atrial fibrillation, peripheral vascular disease and highest quartile) but not endocrine deficient, (iii) endocrine deficient
smoking was defined by self- or informant-report, documentation in (IGF-1 or DHEAS, or both, being in lowest quartile) but not pro-in-
clinical notes or by the use of disease-specific medications. The pre- flammatory and (iv) both pro-inflammatory and endocrine deficient.
sence of ischemic heart disease was defined as having a history of an-
gina, myocardial infarction, congestive heart failure or previous cor- 2.3. Statistical analyses
onary artery stenting or bypass surgery, verified against the patients’
medical records. Medical records were also reviewed for the presence of Descriptive data are presented as means (+SD) or median (inter-
chronic inflammatory disease and any active treatment with steroids or quartile range, IQR) for quantitative variables and as absolute and re-
immunosuppressant medication. lative frequencies for categorical variables. We performed univariate
Standing height (m), body weight (kg) and waist circumference analyses comparing no sarcopenia, pre-sarcopenia and sarcopenia
(cm) were measured, and body mass index (BMI) was calculated using groups in baseline demographics, clinical measures of cognitive, func-
the formula weight/height2 (kg/m2). With increasing recognition for tional and physical performance, vascular co-morbidities and blood
the limitation of BMI as an indicator of obesity as it includes lean mass biomarker measurements using One-way ANOVA and Kruskal-Wallis
in its calculation without discriminating between muscle and fat dis- tests for parametric and non-parametric continuous variables respec-
tribution, A Body Shape Index (ABSI) was calculated for each individual tively, and Chi-square and Fisher’s exact tests for categorical variables.
[ABSI = Waist circumference/(BMI2/3Height1/2)], which allows for Multinomial logistic regression, with the referent group being non-
better representation of central body volume (Krakauer & Krakauer, sarcopenia, was performed to identify factors independently associated
2012). with the differential loss of skeletal muscle mass and function. The
Functional ability was evaluated using Barthel’s basic activities of model included potentially modifiable factors that were observed to be
daily living (ADL) index (Mahoney & Barthel, 1965) and Lawton and significant on univariate analysis, adjusted a priori for age, gender and
Brody’s instrumental ADL (iADL) index (Barbeger-Gateau et al., 1992). severity of cognitive impairment.
Physical performance was measured using the Short Physical Perfor- Statistical analyses were performed using STATA version 14 (Stata
mance Battery (SPPB) (Guralnik et al., 1994), which assessed balance in Corp., College Station, TX). All statistical tests were two-tailed, with p
the side-by-side, semi-tandem and tandem positions; gait speed; and value < 0.05 considered statistically significant
lower limb strength and power on the single and repeated chair stands.
Physical activity level was captured using the Frenchay activity scale 3. Results
(Wade, Legh-Smith, & Langton Hewer, 1985). Nutrition was system-
atically assessed using the locally validated mini nutritional assessment 3.1. Clinical characteristics (Table 1)
(MNA) questionnaire (Chan et al., 2010).
We reviewed each subject’s neuroimaging scan – brain computed We recruited 129 subjects between December 2012 and July 2015.
tomography (CT) scan or magnetic resonance imaging (MRI). White 108 subjects had complete baseline clinical assessments, blood bio-
matter lesion (WML) severity was graded using the Age-Related White marker measurements and DXA imaging, and were included for ana-
Matter Changes (ARWMC) scale by a blinded rater (Wahlund et al., lysis, with the mean age being 76.7 + 6.5 years, female predominance
2001). Medial temporal atrophy (MTA) score reflecting neurodegen- (64.8%) and majority of Chinese ethnicity (92.6%). There was no dif-
eration was scored on T1-weighted coronal slices parallel to the ference in age, gender and severity of cognitive impairment between
brainstem axis and perpendicular to the hippocampal axis, by a con- participants with DXA imaging and those who were excluded due to
sensus method where the scores range from 0 (no atrophy) to 4 (severe unavailability of DXA results.
atrophy) (Wahlund, Julin, Johansson, & Scheltens, 2000). Forty-one (38%) subjects were non-sarcopenic, 14 (13%) were pre-
Total 25-hydroxy vitamin D level was measured at baseline, and sarcopenic and 53 (49%) were sarcopenic at baseline. Sarcopenic sub-
APOE genotyping into APOEε2,3,4 isoforms was performed via re- jects were significantly older (76.5 + 5.5 vs 72.4 + 7.6 vs 78.3 + 6.5,
striction enzyme analysis using applied biosystems platform- ABI Prism p = 0.008), while the pre-sarcopenic group had significantly more
310 Genetic Analyser. male subjects. While a significant difference in CMMSE scores was
observed across groups, being highest in pre-sarcopenia, the 3 groups
2.2.4. Blood biomarkers were similar in baseline severity of cognitive impairment as assessed
All subjects underwent baseline blood biomarker measurements. using diagnostic categories (MCI, mild AD or moderate AD) and CDR
Blood was drawn following an 8-h fast and the aliquoted serum was (global and sum-of-boxes) scores. All vascular co-morbidities examined
frozen and stored at −80 °C until the tests were performed. were similarly distributed across groups, and there was no difference in
Inflammatory status was assessed by serum levels of soluble tumour severity of white matter lesions on neuroimaging scans between groups.
necrosis factor-α receptor-1 (TNF-R1, R&D Systems, Minneapolis, MN, Only 95 subjects had the appropriate neuroimaging coronal slices for
USA), interleukin-6 (high sensitive IL-6, eBioscience, San Diego, CA, evaluation of hippocampal atrophy, with no observed difference in
USA), and the acute phase reactant C-reactive protein (CRP, MTA scores between groups. Overall prevalence of chronic in-
eBioscience) via ELISA. The endocrine markers IGF-1 and DHEAS were flammatory disease was low in our cohort (1.9%).
quantified using commercial ELISA assays (BioVendor, Brno, Czech Physical performance and activity level were significantly different
Republic and Abcam, Cambridge, UK respectively). All biomarkers were across groups, with pre-sarcopenic group having highest SPPB and FAI
measured in duplicates according to manufacturers’ recommendations, scores. No subject was overtly malnourished based on MNA total
and the average value was reported for all assays. Detection limits were score < 17, and the non-sarcopenic group was least likely to be at-risk
as follows: DHEAS, 0.2 μmol/L; IGF-1, 5 ng/ml; IL-6, 0.1 pg/ml; TNF- of malnutrition (MNA total score 17–23.5: 7.3% vs 42.9% vs 47.2%,
R1, 20 pg/ml. p < 0.001). There was no difference in serum Vitamin D levels and
To examine the impact of an elevated inflammatory state or endo- ApoE4 status between groups.
crine deficiency, we adopted quartile cut-offs from a local cohort of
cognitively well and independent older adults, with the highest quartile 3.2. Body composition and muscle quality (Table 2)
for IL-6 and TNF-R1 considered pro-inflammatory, and the lowest
quartile for IGF-1 being deficient. The lowest quartile of DHEAS in the ABSI was similar across groups, despite observed significant dif-
current cohort was used to define DHEAS deficiency. Each subject was ference in BMI, being highest in non-sarcopenic group. Not

22
L. Tay et al. Archives of Gerontology and Geriatrics 75 (2018) 20–27

Table 1 unexpectedly, appendicular lean mass was highest in the non-sarco-


Baseline Clinical Characteristics. penic group. We observed lowest average fat mass in both upper and
lower extremities in the pre-sarcopenic group, who also had sig-
Sarcopenia Status p value
nificantly lowest percentage body fat. Muscle quality as a ratio of grip
No sarcopenia Pre-sarcopenia Sarcopenia strength to arm lean mass was highest in the pre-sarcopenic group [10.6
(n = 41) (n = 14) (n = 53) (7.7–12.2) vs 13.9 (12.6–15.7) vs 11.3 (9.7–12.8), p < 0.001].
With recognized differences in body composition between genders,
Demographics
Age 76.2 (5.5) 72.4 (7.6) 78.3 (6.5)* 0.008 we performed subgroup analysis separately for men and women. ABSI
Gender (male, %) 11 (26.8%) 9 (64.3%) 18 (34%) 0.039 was similar across groups regardless of gender. While percentage body
Ethnicity 37 (90.2%) 13 (92.9%) 50 (94.3%) 0.433 fat was similarly distributed across groups in women, pre-sarcopenic
(Chinese, %) men exhibited a trend for having lowest percentage body fat. Muscle
Diagnostic Categories (%) quality in the upper limbs differed significantly and was highest in the
Mild cognitive 8 (19.5%) 4 (28.6%) 6 (11.3%) pre-sarcopenic group in both genders [men: 11.9 (10.8–13.5) vs 13.9
impairment
(12.6–14.8) vs 11.5 (9.2–12.5), p = 0.008; women: 9.2 (7.4–12.2) vs
Mild AD 28 (68.3%) 9 (64.3%) 37 (69.8%) 0.471
Moderate AD 5 (12.2%) 1 (7.1%) 10 (18.9%) 13.8 (13.0–6.2) vs 11.3 (9.8–12.8), p = 0.002].
Due to the observed significantly poorer physical performance on
Co-morbidities
Hypertension 28 (68.3%) 10 (71.4%) 34 (64.2%) 0.882 SPPB in non-sarcopenic compared with pre-sarcopenic group despite
Diabetes mellitus 10 (24.4%) 3 (21.4%) 20 (37.7%) 0.280 normal muscle mass, we performed sub-group analysis for the non-
Hyperlipidaemia 30 (73.2%) 12 (85.7%) 32 (60.4%) 0.158 sarcopenic group, comparing participants with impaired muscle per-
Ischemic heart 6 (14.6%) 1 (7.1%) 13 (24.5%) 0.269 formance (weak grip strength and/or slow gait speed) versus their
disease
counterparts with preserved muscle performance. 29 (70.1%) non-sar-
Atrial fibrillation 2 (4.9%) 1 (7.1%) 1 (1.9%) 0.462
Smoking 5 (13.9%) 3 (21.4%) 9 (17%) 0.735 copenic participants had impaired muscle performance, with sig-
Stroke/ transient 2 (4.9%) 0 1 (1.9%) 0.723 nificantly lower SPPB [10 (9.5–11) vs 7 (5–9), p = 0.002] and FAI [26
ischemic attack (21–34.5) vs 18 (14–21), p = 0.002] scores, although appendicular
Chronic 1(2.4%) 0 1 (1.9%) 1.00
lean mass was similar between groups (6.8 + 0.34 vs 6.3 + 0.16,
inflammatory
disease
p = 0.096). While there was no difference in age, non-sarcopenic par-
ticipants with impaired muscle performance were more likely to be
Major depressive 2 (4.9%) 0 1 (1.9%) 0.723
disorder
female (p = 0.007) and had more severe cognitive impairment [CRD
sum-of-boxes: 5 (2–6) vs 2.25 (1.25–5), p = 0.039] (Supplementary
Cognitive Performance
Table).
CDR global 0.213
0.5 16 (39%) 5 (35.7%) 10 (18.9%)
1 20 (48.8%) 8 (57.1%) 32 (60.4%) 3.3. Inflammatory-endocrine biomarkers (Table 3)
2 5 (12.2%) 1 (7.1%) 11 (20.8%)
CDR sum-of- 4.4 (3.0) 4.1 (2.5) 5.4 (2.9) 0.140
The pro-inflammatory cytokines IL-6 and TNF-R1 did not differ-
boxes (mean,
SD)
entiate between non-, pre- and sarcopenic groups, in both absolute
CMMSE (mean, 18.5 (5.5) 21.5 (3.4) 17.4 (5.3)* 0.036 serum concentrations and when upper quartile cut-offs were employed.
SD) Serum concentrations of the acute phase protein CRP were similar
Neuroimaging across groups.
ARWMC score 6.17 (4.58) 4.64 (3.10) 5.16 (4.52) 0.410 In the endocrine axes, serum IGF-1 concentration was significantly
MTA score 1.43 (1.01) 1.29 (0.91) 1.47 (1.05) 0.828 different across groups, being lowest in the sarcopenic group and
(n = 95)
highest in pre-sarcopenic. Adopting the lowest quartile of serum IGF-1
Neuropsychiatric Symptoms concentration to define an IGF-1 deficient state, sarcopenic subjects
NPI severity 4 (1–6) 2 (0–2) 3 (1–5) 0.069
were most likely to be IGF-1 deficient, while pre-sarcopenic subjects
(median, IQR)
were least likely to exhibit IGF-1 deficiency (29.3% vs 14.3% vs 56.6%,
Functional Performance p = 0.003). There was no significant difference in serum DHEAS con-
MBI (median, 100 (95–100) 100 (100–100) 100 (95–100) 0.269
IQR)
centration across groups, although a trend for higher prevalence of a
iADL (median, 15 (11–19) 17.5 (15–20) 14 (10–18) 0.088 DHEAS-deficient state was observed with sarcopenia.
IQR) When pro-inflammatory and endocrine-deficient states were ex-
Physical Performance amined in tandem, sarcopenic group was least likely to be in the “ideal”
SPPB (median, 8 (7–10) 10 (10–11) 9 (7–10) 0.033 state of being neither pro-inflammatory nor endocrine deficient, and
IQR) had the highest proportion of subjects being in the combined pro-in-
FAI (mean, SD) 21 (8.6) 26.9 (8.0) 19.4 (7.3)* 0.010 flammatory and endocrine-deficient state (17.1% vs 14.3% vs 41.5%,
Nutritional Status p = 0.016).
MNA-at risk of 3 (7.3%) 6 (42.9%) 25 (47.2%) < 0.001
malnutrition
3.4. Factors independently associated with pre-sarcopenia and sarcopenia
Vitamin D (mean, 20.7 (9.1) 24.8 (8.2) 20.3 (11.5) 0.341
SD) (Table 4)
ApoE status
ApoE4 positive 17 (41.5%) 8 (57.1%) 14 (27.5%) 0.091 We performed multi-nomial logistic regression with non-sarcopenia
as the reference group, and included in the model nutritional status and
AD: Alzheimer’s disease; ARWMC: age-related white matter changes (range 0–30); CDR: inflammatory-endocrine state as the independent potentially modifi-
Clinical Dementia Rating; CMMSE: Chinese Mini-Mental State Examination (range able variables, adjusted a priori for age, gender, and CDR sum-of-boxes
0–28);) FAI: Frenchay Activity Index (range: 0–45); iADL: Lawton and Brody’s instru-
score. We excluded percentage body fat owing to collinearity with
mental Activities of Daily Living (range 0–23); MBI: Modified Barthel Index (range
nutritional status. Malnutrition was associated with significantly in-
0–100); MNA: Mini Nutrition Assessment; SPPB: Short Physical Performance Battery
(range 0–12). creased risk for both pre-sarcopenia (Relative risk ratio = 7.53, 95% C.I
* p < 0.05 between sarcopenia and pre-sarcopenia. 1.20–47.51, p = 0.032) and sarcopenia (Relative risk ratio = 11.91,
95% C.I 2.85–49.77, p = 0.001). Inflammatory-endocrine state was not

23
L. Tay et al. Archives of Gerontology and Geriatrics 75 (2018) 20–27

Table 2
Body Composition and Muscle Quality.

Sarcopenia Status p value

No sarcopenia Pre-sarcopenia Sarcopenia


(n = 41) (n = 14) (n = 53)

Body Composition
BMI (mean, SD) 25.4 (3.3) 21.2 (2.3) 21.7 (3.2) < 0.001
ABSI 0.083 (0.005) 0.085 (0.006) 0.086 (0.006) 0.155

DXA Imaging
Appendicular Lean Mass (kg/m2) (mean, SD) 6.45 (0.86) 5.83 (0.85)** 5.32 (0.78)# < 0.001
Average fat mass arm (g) (mean, SD) 1240.2 (451.5) 913.0 (308.4)** 1002.1 (322.5)# 0.003
Average fat mass leg (g) (mean, SD) 3391.8 (1050.5) 2649.0 (884.4)** 2661.8 (740.1)# < 0.001
% body fat 38.5 (9.3) 30.8 (8.5)** 36.3 (6.6) 0.010

Muscle Quality
Arm muscle quality (kg/kg) (median, IQR) 10.6 (7.7–12.2) 13.9 (12.6–15.7) 11.3 (9.7–12.8) < 0.001
Leg muscle quality (kg/kg) (mean, SD) 5.8 (1.8) 5.8 (1.5) 6.0 (1.8) 0.901

Men (n = 38) Women (n = 70)

No sarcopenia Pre-sarcopenia Sarcopenia No sarcopenia Pre-sarcopenia Sarcopenia


(n = 11) (n = 9) (n = 18) (n = 30) (n = 5) (n = 35)

DXA Imaging
Appendicular Lean Mass (kg/m2) (mean, SD) 7.56 (0.71) 6.35 (0.54)** 6.16 (0.66)*,# 6.05 (0.46) 4.9 (0.27)** 4.89 (0.40)*,#
% body fat 36.3 (14.9) 26.3 (6.6) 30.9 (6.3) 39.3 (6.4) 39.0 (4.2) 39.2(4.7)

Muscle Quality
Arm muscle quality (kg/kg) (median, IQR) 11.9 (10.8–13.5) 13.9 (12.6–14.8) 11.5 (9.2–12.5)* 9.2 (7.4–12.2) 13.8 (13.0–6.2) 11.3 (9.8–12.8)*
Leg muscle quality (kg/kg) (mean, SD) 6.2 (1.1) 5.4 (1.1) 5.5 (1.7) 5.7 (2.0) 6.4 (1.9) 6.2 (1.8)

ABSI: A New Body Shape Index; BMI: Body Mass Index.


* p < 0.05 across groups.
** p < 0.005 between pre-sarcopenia and no sarcopenia.
#
p < 0.05 between sarcopenia and no sarcopenia.

associated with pre-sarcopenia, but the combined state of being pro- independent risk factor for decline in muscle mass, with incremental
inflammatory and endocrine deficient significantly increased the risk of risk for pre-sarcopenia and sarcopenia. However, in the pre-sarcopenic
sarcopenia (Relative risk ratio = 5.17, 95% C.I 1.31–20.37, group, the poorer nutritional status was not sufficient to yield a com-
p = 0.019). promise in muscle quality and, consequently, physical performance.
While women were significantly less likely to be pre-sarcopenic, While 2 out of 3 malnourished older adults were physically frail, only
sarcopenia risk was independent of gender. Severity of cognitive im- 10% of physically frail seniors were noted to be malnourished (Verlaan
pairment was not associated with pre-sarcopenia or sarcopenia. et al., 2017). Our findings thus further suggest that nutrition may be a
key substrate for muscle mass, but other mechanisms may be driving
the qualitative decline beyond quantitative muscle loss, to culminate in
4. Discussion functional deficits clinically manifest as sarcopenia and its sequelae of
physical frailty. This will be of clinical significance in light of the recent
Our study supports the clinical relevance of muscle quality beyond systematic review in which nutritional intervention in isolation failed to
muscle mass as a determinant of functional performance in cognitively yield improvement in muscle strength among older people with re-
impaired older adults, evident by the pre-sarcopenic group being the duced functional ability (Beck, Dent, & Baldwin, 2016).
most physically active and exhibiting highest physical performance As further support of our hypothesized differential mechanisms for
despite their low muscle mass. While malnutrition is a precursor for the quantitative and qualitative declines in skeletal muscle, in-
progressive loss of muscle mass, being the common risk factor for pre- flammatory-endocrine dysregulation conferred a five-fold higher risk
sarcopenia and sarcopenia, a combined pro-inflammatory and endo- for sarcopenia, despite not being associated with pre-sarcopenia. The
crine deficient state aggravates decline in muscle performance culmi- differential association suggests that against a background of declining
nating in frank sarcopenia. muscle mass, a heightened state of inflammation in tandem with en-
Previous research had shown that changes in muscle mass explained docrine deficiency impairs muscle performance. Our findings also
only 6–8% of the variability in muscle strength among older adults parallel previously reported differences in related factors for muscle
(Goodpaster et al., 2006). Indeed, the dissociation between muscle mass mass and function, as declines in growth hormone and IGF-1 have been
and physical performance in the pre-sarcopenic group, such that muscle linked to loss of muscle mass but not muscle power (Morley, 2016),
quality as reflected by grip strength per unit arm lean mass and SPPB which corroborate the observed lack of a significant impact of an iso-
scores are higher relative to the non-sarcopenic group, suggests that the lated endocrine deficient state on the risk of sarcopenia. The detri-
efficiency of muscle tissue has greater impact on functional con- mental effect of a combined pro-inflammatory and endocrine deficient
sequences compared with an isolated decline in muscle mass. This is state, over the isolated presence of either inflammation or endocrine
further reinforced by findings within the non-sarcopenic group whereby deficiency, is concordant with previously reported synergistic effects of
70% of subjects had impairments in performance on grip strength or IGF-1 and IL-6 on progressive disability (Cappola et al., 2008) as well as
gait speed despite normal muscle mass, with significantly poorer both in-vivo and in-vitro observed interactions between anabolic hor-
muscle quality compared with their counterparts with preserved muscle mones and pro-inflammatory cytokines (Andreassen et al., 2010;
performance measures. Lelbach, Scharf, & Ramadori, 2001). The current findings seemingly
Consistent with widely reported literature, malnutrition was an

24
L. Tay et al. Archives of Gerontology and Geriatrics 75 (2018) 20–27

Table 3 when occurring in isolation rather than being in tandem with an en-
Inflammatory and Endocrine Biomarkers. docrine-deficient state (Tay, Lim, Chan, Ye, & Chong, 2016). However,
the observed physical performance limitations despite normal muscle
Sarcopenia Status p value
mass in approximately one-quarter of our cohort suggest that there may
No Pre- Sarcopenia be non-muscle pathways to physical frailty. Indeed, slowness and
sarcopenia sarcopenia weakness – the core characteristics of sarcopenia and physical frailty-
(n = 41) (n = 14) (n = 53) are commonly associated with neurological conditions. This is also re-
Pro-inflammatory Cytokines inforced by the observation of Huang et al., who noted that slowness
IL-6 (pg/ml, median, 0.17 0.28 0.2 0.521 and weakness with non-muscle etiology were strongly associated with
IQR) (0.1–0.47) (0.06–0.57) (0.1–0.69) cognitive impairment (Huang et al., 2016). Dysfunctions in the per-
Elevated IL-6 (%) 2 (4.9%) 0 8 (15.1%) 0.150 ipheral and central nervous system have been implicated in muscle
TNF-R1 (pg/ml, 4883.5 4892.6 5256.5 0.504
quality (Moore et al., 2014), and may account for the observed higher
mean, SD) (1648.0) (1149.6) (1735.7)
Elevated TNF-R1 16 (39%) 7 (50%) 27 (50.9%) 0.494 cognitive performance scores in the pre-sarcopenic group compared
(%) with both non-sarcopenic and sarcopenic groups. In addition, we ob-
CRP (mg/L, median, 0.14 0.19 0.10 0.458 served worse cognitive performance within the non-sarcopenic group
IQR) (0.06–0.33) (0.11–0.45) (0.05–0.36)
with impaired muscle performance. Yet, the overall lack of an asso-
Endocrine ciation between the stage of severity of cognitive impairment and sar-
IGF-1 (ng/ml, mean, 139.2 (80.0) 155.0 (89.7) 97.3 (64.9)*,# 0.006 copenia status is similar to the findings of the EPIDOS cohort, which
SD)
IGF-1 deficient (%) 12 (29.3%) 2 (14.3%) 30 (56.6%) 0.003
found no link between different operative definitions of sarcopenia and
DHEAS (μmol/L, 1.95 1.49 1.50 0.135 cognitive impairment, despite independent and significant associations
median, IQR) (1.3–2.96) (1.04–3.30) (0.81–2.12) between grip strength and gait speed with cognition (Abellan van Khan
DHEAS deficient (%) 7 (17.1%) 2 (14.3%) 18 (34.0%) 0.119 et al., 2013). Together, these observations suggest that functional per-
Inflammatory-Endocrine (%) formance measures rather than muscle mass parameters are associated
Non-inflammatory- 16 (39%) 5 (35.7%) 8 (15.1%) with cognition. Further investigations detailing the mechanisms that
Non-endocrine
link cognitive impairment, sarcopenia and physical frailty may be an-
deficient
Pro-inflammatory 10 (24.4%) 5 (35.7%) 8 (15.1%) 0.016
ticipated to allow a more targeted interventional approach.
Endocrine deficient 8 (19.5%) 2 (14.3%) 15 (28.3%) Beyond neurological factors, changes in muscle architecture in re-
Pro-inflammatory- 7 (17.1%) 2 (14.3%) 22 (41.5%) lation to anthropometrics have been implicated in functional impair-
Endocrine ment of muscle (Goodpaster et al., 2001; Vilaca et al., 2014). In parti-
deficient
cular, intermuscular adipose tissue (IMAT) is an ectopic depot of
DHEAS: dehydroepiandrosterone sulphate; IGF-1: insulin-like growth factor-1; TNF-R1: adipose tissue underneath the fascia and within muscle that has been
tumour necrosis factor-alpha receptor-1. implicated in muscle quality and function in older adults, and may
* p < 0.05 between sarcopenia and pre-sarcopenia. reflect metabolic profile (Beavers et al., 2013). Of significance within
#
p < 0.05 between sarcopenia and no sarcopenia. our study is the lower fat mass in the pre-sarcopenic group, with cor-
respondingly lowest percentage body fat, that highlights the potential
Table 4 for a detrimental effect of adiposity on muscle quality. The relationship
Multinomial Logistic Regression for Factors Associated with Sarcopenia Status. between adiposity and muscle quality is further clarified in a recent
study reporting poorer muscle quality and physical performance in
Relative Risk 95% Confidence p value
Ratio Interval obese women despite a greater quantity of fat free and lean mass
(Vilaca et al., 2014). In parallel with earlier evidence that gender dif-
Pre-sarcopenia ferences may exist in muscle quality measures (Frontera, Hughes, Lutz,
Age 0.88 0.77–1.01 0.070
& Evans, 1991), men were more likely to exhibit preserved physical
Gender (female) 0.13 0.02–0.67 0.015
CDR sum-of-boxes 1.02 0.76–1.35 0.917 performance despite loss of muscle mass (pre-sarcopenia). We have
previously reported differential pathophysiological mechanisms for
MNA-at risk of malnutrition 7.53 1.20–47.51 0.032
sarcopenia in community-dwelling cognitively intact older adults (Tay
Inflammatory-Endocrine et al., 2015), although the sample size of the current study limits further
Proinflammatory 1.89 0.27–13.12 0.520
Endocrine deficient 1.06 0.12–9.16 0.960
sub-group analysis by gender. Earlier studies demonstrating that im-
Proinflammatory-Endocrine 1.59 0.17–14.49 0.681 provement in muscle function need not be accompanied by gains in
deficient muscle mass suggest that muscle weakness culminates from impaired
Sarcopenia muscle quality rather than quantity. Such qualitative changes have also
Age 1.04 0.95–1.14 0.359 been linked to loss of motor units, muscle fibrosis, degeneration of
Gender (female) 0.83 0.26–2.64 0.751 neuromuscular junction, reduced muscle aerobic capacity and reduced
CDR sum-of-boxes 1.08 0.90–1.29 0.432
excitation-contraction coupling (McGregor et al., 2014).
MNA-at risk of malnutrition 11.91 2.85–49.77 0.001 The prevalence of sarcopenia (49%) in our current cognitively im-
Inflammatory-Endocrine paired cohort of older adults is almost twice as high as the previously
Proinflammatory 1.31 0.29–5.37 0.727 reported prevalence of sarcopenia in our local cohort of cognitively
Endocrine deficient 2.12 0.53–8.50 0.289 intact community-dwelling older adults, applying similar diagnostic
Proinflammatory-Endocrine 5.17 1.31–20.37 0.019
algorithms and cut-offs for diagnosis of sarcopenia (Tay et al., 2015).
deficient
The higher prevalence is consistent with the findings of a recent sys-
R2 = 0.234. tematic review, which found a positive association between sarcopenia
Reference state: Non-sarcopenia. and impaired cognition, suggesting that the co-occurrence of both
CDR: Clinical Dementia Rating; MNA: Mini Nutritional Assessment. conditions is common (Chang et al., 2016). Further, the prevalence of
sarcopenia has also been shown to vary across populations and settings
contradict our previous study in which the detrimental effect of a pro- (Cruz-Jentoft et al., 2014).
inflammatory state on physical frailty – typically considered a sequelae We acknowledge several limitations in our study, including the re-
of sarcopenia – in cognitively impaired older adults was significant only latively small sample size, in particular the pre-sarcopenic group, with

25
L. Tay et al. Archives of Gerontology and Geriatrics 75 (2018) 20–27

consequent possible type 2 error. The cross-sectional analysis limits (2013). Associations between body composition and gait speed decline: Results from
inference on temporal relationships between sarcopenia and its risk the Health, Aging and Body Composition study. American Journal of Clinical Nutrition,
97, 552–560.
factors, and the potential for alterations in the measured blood bio- Beck, A. M., Dent, E., & Baldwin, C. (2016). Nutritional intervention as part of functional
markers being consequent to changes in body composition cannot be rehabilitation in older people with reduced functional ability: A systematic review
confidently dismissed. Additionally, the assessment of muscle strength and meta-analysis of randomised controlled studies. Journal of Human Nutrition and
Dietetics, 29(6), 733–745. http://dx.doi.org/10.1111/jhn.12382.
could have been influenced by cognitive function, given the observed Bilotta, C., Bermagaschini, L., Nicolini, P., Case, A., Pina, G., Rossi, S. V., et al. (2012).
higher cognitive performance scores in the pre-sarcopenic group and Frailty syndrome diagnosed according to the Study of Osteoporotic Fractures criteria
more severe cognitive impairment in non-sarcopenic subjects with and mortality in older outpatients suffering from Alzheimer’s disease: A one-year
prospective cohort study. Aging & Mental Health, 16(3), 273–280.
physical performance limitations. However, the recruited subjects had a Burns, J. M., Johnson, D. K., Watts, A., Swerdlow, R. H., & Brooks, W. M. (2010). Lean
generally milder severity of cognitive impairment, having excluded mass is reduced in early Alzheimer’s Disease and associated with brain atrophy.
subjects with advanced dementia, and they were able to perform all Archives of Neurology, 67(4), 428–433.
Canon, M. E., & Crimmins, E. M. (2011). Sex differences in the association between
clinical assessments as instructed. Our careful phenotyping and exclu-
muscle quality, inflammatory markers and cognitive decline. Journal of Nutrition,
sion of participants with other central nervous conditions such as stroke Health and Aging, 15(8), 695–698.
disease reduces the likelihood for physical performance being influ- Cappola, A. R., Xue, Q. L., Ferrucci, L., Guralnik, J. M., Volpato, S., & Fried, L. P. (2008).
enced by neurological conditions other than neurodenegeration. Insulin-like growth factor 1 and interleukin-6 contribute synergistically to disability
and mortality in older women. The Journal of Clinical Endocrinology and Metabolism,
Nonetheless, the study’s inclusion/exclusion criteria limits general- 88, 2019–2025.
izability beyond the earlier stages of dementia, and its restriction to a Chan, M., Lim, Y. P., Ernest, A., & Tan, T. L. (2010). Nutritional assessment in an Asian
Memory Clinic population potentially limits interpretation in less spe- nursing home and its association with mortality. Journal of Nutrition, Health and
Aging, 14(January (1)), 23–28.
cialized settings such as primary care. Chang, K. V., Hsu, T. H., Wu, W. T., Huang, K. C., & Han, D. S. (2016). Association between
In conclusion, the present study corroborates existing literature for sarcopenia and cognitive impairment: A systematic review and meta-analysis, 17(12),
the dissociations between muscle mass, strength and physical perfor- 1164 e7–1164. e15.
Chen, L. K., Liu, L. K., Woo, J., Assantachai, P., Auyeung, T. W., Bahyah, K. S., et al.
mance, but being specific to cognitively impaired older adults. The (2014). Sarcopenia in asia: Consensus report of the Asian Working Group for
ability to maintain physical performance despite the loss of muscle Sarcopenia. Journal of the American Medical Directors Association, 15, 95–101.
mass reiterates the important concept of muscle quality relevant to Chong, M. S., & Sahadevan, S. (2003). An evidence-based clinical approach to the diag-
nosis of dementia. Annals of the Academy of Medicine, Singapore, 23, 740–749.
delaying disability in cognitively impaired older adults. While opti- Cruz-Jentoft, A. J., Maeyens, J. P., Bauer, J. M., Boirie, Y., Cederholm, T., Landi, F., et al.
mizing nutritional status will undoubtedly remain important to prevent (2010). Sarcopenia: European consensus on definition and diagnosis: Report of the
progressive loss of muscle mass, more targeted interventions that si- European Working Group on Sarcopenia in Older People. Age and Ageing, 1(12),
412–423.
multaneously address immune and endocrine dysfunctions will be ne-
Cruz-Jentoft, A. J., Landi, F., Schneider, S. M., Zunigna, C., Arai, H., Boirie, Y., et al.
cessary to ensure maintenance of functional independence through (2014). Prevalence of and interventions for sarcopenia in ageing adults: A systematic
preserving muscle quality. review. Report of the International Sarcopenia Initiative (EWGSOP and IWGS). Age
and Ageing, 43, 748–759.
Fried, L. P., Ferruci, L., Darer, J., Williamson, J. D., & Anderson, G. (2004). Untangling
Conflicts of interest the concepts of disability, frailty and co-morbidity: Implications for improved tar-
geting and care. The Journals of Gerontology Series A: Biological Sciences and Medical
All the authors have no conflict of interest or financial disclosure of Sciences, 59, 255–263.
Frontera, W. R., Hughes, V. A., Lutz, K. J., & Evans, W. J. (1991). A cross-sectional study
note. of muscle strength and mass in 45- to 78-yr-old men and women. Journal of Applied
Physiology, 71(2), 644–650.
Funding support Goodpaster, B. H., Carlson, C. L., Visser, M., Kelley, D. E., Scherzinger, A., Harris, T. B.,
et al. (2001). Attenuation of skeletal muscle and strength in the elderly: The Health
ABC study. Journal of Applied Physiology, 90(6), 2157–2165.
This study was funded by NHG Clinician Scientist Career Goodpaster, B. H., Park, S. W., Harris, T. B., Kritchevsky, S. B., Nevitt, B., Schwartz, A. V.,
SchemeCSCS/12002 and NHG CSCS/13001. et al. (2006). The loss of skeletal muscle strength, mass, and quality in older adults.
The health, aging and body composition study. The Journals of Gerontology Series A:
Biological Sciences and Medical Sciences, 61, 1059–1064.
Acknowledgements Guralnik, J. M., Simonsick, E. M., Ferruci, L., Glynn, R. J., Berkman, L. F., Blazer, D. G.,
et al. (1994). A short physical performance battery assessing lower extremity func-
tion: Association with self-reported disability and prediction of mortality and nursing
We thank Prof Tan Eng King and Ms Ebonne Ng for the ApoE gen-
home admission? Journal of Gerontology, 49(2), M85–94.
otyping. We also extend our thanks to our patients and their caregivers Holmes, C., Cunningham, C., Zotova, E., Woolford, J., Dean, C., Kerr, S., et al. (2009).
who have graciously consented to participation in the study. Systemic inflammation and disease progression in Alzheimer disease. Neurology, 73,
768–774.
Huang, C. Y., Hwang, A. C., Liu, K. C., Lee, W. J., Chen, L. Y., Peng, L. N., et al. (2016).
Appendix A. Supplementary data Association of dynapenia, sarcopenia and cognitive impairment among community-
dwelling older Taiwanese. Rejuvenation Research, 19(1), 71–78.
Supplementary data associated with this article can be found, in the Johnson, D. K., Wilkins, C. H., & Morris, J. C. (2006). Accelerated weight loss may pre-
cede diagnosis in Alzheimer Disease. Archives of Neurology, 63(9), 1312–1317.
online version, at https://doi.org/10.1016/j.archger.2017.11.008. Krakauer, N. Y., & Krakauer, J. C. (2012). A new body shape index predicts mortality
hazard independently of body mass index. PLoS One, 7(7), e39504.
References Landi, F., Liperoti, R., Rusco, D., Mastrapaolo, S., Quatttrociocchi, D., Proja, A., et al.
(2012). Prevalence and risk factors of Sarcopenia among nursing home older re-
sidents. The Journals of Gerontology Series A: Biological Sciences and Medical Sciences,
Abellan van Khan, G., Cesari, M., Gillette-Guyonnet, S., Dupuy, C., Nourhashemi, F., 67, 48–55.
Schott, A. M., et al. (2013). Sarcopenia and cogntiive impairment in elderly women: Lelbach, A., Scharf, J. G., & Ramadori, G. (2001). Regulation of insulin-like growth factor-
Results from the EPIDOS cohort. Age and Ageing, 42, 196–202. I and of insulin- like growth factor binding protein-1, -3 and -4 in cocultures of rat
Andreassen, M., Raymond, I., Hillbrandt, P., Kistorp, C., Rathcke, C., Vestergaard, H., hepatocytes and Kupffer cells by interleukin-6. Journal of Hepatology, 35(November
et al. (2010). Associations between plasma insulin-like growth factor-1 and the (5)), 558–567.
markers of inflammation interleukin-6, c-reactive protein and YKL-40 in an elderly Lim, W. S., Chin, J. J., Lam, C. K., Lim, P. P., & Sahadevan, S. (2005). Clinical dementia
background population. Inflammation Research, 59(7), 503–510. rating: Experience of a multi-racial Asian population. Alzheimer Disease and Associated
Barbat-Artigas, S., Rolland, Y., Zamboni, M., & Aubertin-Leheudre, M. (2012). How to Disorders, 19, 135–142.
assess functional status: A new muscle quality index. Journal of Nutrition, Health and Maggio, M., Dall'Aglio, E., Lauretani, F., Cattabiani, C., Ceresini, S., Caffarra, F., et al.
Aging, 16, 67–77. (2012). The hormonal pathway to cognitive impairment in older men. Journal of
Barbeger-Gateau, P., Commenges, D., Gagnon, M., Letenneur, L., Sauvel, C., & Dartiques, Nutrition, Health and Aging, 16(1), 40–54.
J. F. (1992). Instrumental activities of daily living as a screening tool for cognitive Mahoney, F. I., & Barthel, D. (1965). Functional evaluation: The Barthel Index. Maryland
impairment and dementia in elderly community dwellers. Journal of the American State Medical Journal, 14, 56–61.
Geriatrics Society, 40, 1129–1134. McGregor, R. A., Cameron-Smith, D., & Poppitt, S. D. (2014). It is not just muscle mass: A
Beavers, K. M., Beavers, D. P., Houston, D. K., Harris, T. B., Hue, T. F., Koster, A., et al. review of muscle quality, composition and metabolism during ageing as determinants

26
L. Tay et al. Archives of Gerontology and Geriatrics 75 (2018) 20–27

of muscle function and mobility in later life. Longevity & Healthspan, 3(1), 9. et al. (2012). Value of measuring muscle performance to assess changes in lean mass
McKhann, G., Drachmann, D. A., Folstein, M., Katzman, R., Price, D., Stadian, E. M., et al. with testosterone and growth hormone supplementation. European Journal of Applied
(1984). Clinical diagnosis of Alzheimer’s disease – Report to the NINCDS-ADRDA Physiology, 112(3), 1123–1131.
workgroup under the auspices of Department of Health and Human Services Task Shin, H. Y., Kim, S. W., Kim, J. M., Shin, I. S., & Yoon, J. S. (2012). Association of grip
Force on Alzheimer’s disease. Neurology, 34, 939–944. strength with dementia in a Korean older population. International Journal of Geriatric
Moore, A. Z., Caturegli, G., Metter, E. J., Makrogiannis, S., Resnick, S. M., Harris, T. B., Psychiatry, 27(5), 500–505.
et al. (2014). Difference in muscle quality over the adult life-span and biological Tay, L., Ding, Y. Y., Leung, B. P., Ismail, N. H., Yeo, A., Yew, S., et al. (2015). Sex-specific
correlates in the Baltimore Longitudinal Study of Aging. Journal of the American differences in risk factors for sarcopenia in amongst community-dwelling older
Geriatrics Society, 62(2), 230–236. adults. Age, 37, 121.
Morley, J. E. (2016). Frailty and sarcopenia: The new geriatric giants. Revista De Tay, L., Lim, W. S., Chan, M., Ye, R. J., & Chong, M. S. (2016). The independent role of
Investigacion Clinica, 68, 59–67. inflammation in baseline physical frailty among older adults with mild cognitive
Morris, J. C. (1993). The Clinical Dementia Rating (CDR): Current version and scoring impairment and mild-to-moderate Alzheimer’s disease. Journal of Nutrition, Health
rules. Neurology, 43, 2412–2414. and Aging, 20(3), 288–299.
Murphy, R. A., Edward, H. I., Zhang, Q., Boudreau, R. M., Cawthon, P. M., Newman, A. B., Tolea, M. I., & Galvin, J. E. (2015). Sarcopenia and impairment in cognitive and physical
et al. (2014). Transition to sarcopenia and determinants of transitions in older adults: performance. Clinical Interventions in Aging, 10, 663–671.
A population-based study. Journals of Gerontology Series A: Biomedical Sciences and Verlaan, S., Ligthart-Melis, G. C., Wijers, S. L. J., Cederholm, T., Maier, A. B., de van der
Medical Sciences, 69(6), 751–758. Schueren, M. A. E., et al. (2017). High prevalence of physical frailty among com-
Newman, A. B., Kupelin, V., Visser, M., Simonsick, E. M., Goodpaster, B. H., Kritchevsky, munity-dwelling malnourished older adults- A systematic review and meta-analysis.
S. B., et al. (2006). Strength, but not muscle mass, is associated with mortality in the Journal of the American Medical Directors Association, 18(5), 374–382.
health, aging and body composition study cohort. Journals of Gerontology Series A: Vilaca, K. H., Carneiro, J. A., Ferriolli, E., Lima, N. K., de Paula, F. J., Moriguti, J. C., et al.
Biomedical Sciences and Medical Sciences, 61, 72–77. (2014). Body composition, physical performance and muscle quality of active elderly
Ni Mhaolain, A. M., Gallagher, D., Crosby, L., Ryan, D., Lacey, L., Coen, R. F., et al. women. Archives of Gerontology and Geriatrics, 59(1), 44–48.
(2012). Frailty and quality of life for people with Alzheimer’s dementia and mild Visser, M., Goodpaster, B. H., Kritchevsky, S. B., Newman, A. B., Nevitt, M., Rubin, S. M.,
cognitive impairment. The American Journal of Alzheimer’s Disease & Other Dementias®, et al. (2005). Muscle mass, muscle strength, and muscle fat infiltration as predictors
27(1), 48–54. of incident mobility limitations in well-functioning older persons. The Journals of
Nourhaehemi, F., Andrieu, S., Gillette-Guyonnet, S., Reynish, E., Albarede, J. L., Gerontology Series A: Biological Sciences and Medical Sciences, 60(3), 324–333.
Grandjean, H., et al. (2002). Is there a relationship between fat-free soft tissue mass von Haehling, S., Morley, J. E., & Anker, S. D. (2010). An overview of sarcopenia: Facts
and low cognitive function? Results from a study of 7.105 women. Journal of the and numbers on prevalence and clinical impact. Journal of Cachexia, Sarcopenia and
American Geriatrics Society, 50(11), 1796–1801. Muscle, 1, 129–133.
Oosterveld, S. M., Kessels, R. P., Hamel, R., Ramakers, R. H., Aalten, P., Verhey, F. R., Wade, D. T., Legh-Smith, J., & Langton Hewer, R. (1985). Social activities after stroke:
et al. (2014). The influence of co-morbidity and frailty on clinical manifestation of Measurement and natural history using the Frenchay Activities Index. International
patients with Alzheimer’s disease. Journal of Alzheimer’s Disease, 42(2), 501–509. Rehabilitation Medicine, 7(4), 176–181.
Sahadevan, S., Lim, P. P., Tan, N. J., & Chan, S. P. (2000). Diagnostic performance of two Wahlund, L. O., Julin, P., Johansson, S. E., & Scheltens, P. (2000). Visual rating and
mental status tests in the older Chinese: Influence of education and age on cut-off volumetry of the medial temporal lobe on magnetic resonance imaging in dementia:
values. International Journal of Geriatric Psychiatry, 15, 234–241. A comparative study. The Journal of Neurology, Neurosurgery, & Psychiatry, 69,
Sahadevan, S., Lim, J. P., Tan, N. J., & Chan, S. P. (2002). Psychometric identification of 630–635.
early Alzheimer disease in an elderly Chinese population with differing educational Wahlund, L. O., Barkhof, F., Fazekas, F., Bronge, L., Augustin, M., Sjogren, M., et al.
levels. Alzheimer Disease and Associated Disorders, 16, 65–72. (2001). A new rating scale for age-related white matter changes applicable to MRI
Schaap, L. A., Plujim, S. M. F., Deeg, D. J. H., Harris, T. B., Krtichevsky, S. B., Newman, A. and CT. Stroke, 32 1318-22.23.
B., et al. (2009). Higher inflammatory marker levels in older persons: Associations Watanabe, T., Miyazaki, A., Katagiri, T., Yamamoto, H., Idei, T., Iguchi, T., et al. (2005).
with 5-year change in muscle mass and muscle strength. Journals of Gerontology Series Relationship between serum insulin-like growth factor-1 levels and Alzheimer’s dis-
A: Biomedical Sciences and Medical Sciences, 64A(11), 1183–1189. ease and vascular dementia. Journal of the American Geriatrics Society, 53, 1748–1753.
Schroddder, E. T., He, J., Yarasheski, K. E., Binder, E. F., Castaneda-Sceppa, C., Bhasin, S.,

27

You might also like