ArticlePDF AvailableLiterature Review

The Impact of Dairy Protein Intake on Muscle Mass, Muscle Strength, and Physical Performance in Middle-Aged to Older Adults with or without Existing Sarcopenia: A Systematic Review and Meta-Analysis

Authors:

Abstract and Figures

Sarcopenia is an age-related condition associated with a progressive loss of muscle mass and strength. Insufficient protein intake is a risk factor for sarcopenia. Protein supplementation is suggested to improve muscle anabolism and function in younger and older adults. Dairy products are a good source of high-quality proteins. This review evaluates the effectiveness of dairy proteins on functions associated with sarcopenia in middle-aged and older adults. Randomized controlled trials were identified using PubMed, CINAHL/EBSCO, and Web of Science databases (last search: 10 May 2017) and were quality assessed. The results of appendicular muscle mass and muscle strength of handgrip and leg press were pooled using a random-effects model. The analysis of the Short Physical Performance Battery is presented in narrative form. Adverse events and tolerability of dairy protein supplementation were considered as secondary outcomes. Fourteen studies involving 1424 participants aged between 61 and 81 y met the inclusion criteria. Dairy protein significantly increased appendicular muscle mass (0.13 kg; 95% CI: 0.01, 0.26 kg; P = 0.04); however, it had no effect on improvement in handgrip (0.84 kg; 95% CI: -0.24, 1.93 kg; P = 0.13) or leg press (0.37 kg; 95% CI: -4.79, 5.53 kg; P = 0.89). The effect of dairy protein on the Short Physical Performance Battery was inconclusive. Nine studies reported the dairy protein to be well tolerated with no serious adverse events. Although future high-quality research is required to establish the optimal type of dairy protein, the present systematic review provides evidence of the beneficial effect of dairy protein as a potential nutrition strategy to improve appendicular muscle mass in middle-aged and older adults.
Content may be subject to copyright.
REVIEW
The Impact of Dairy Protein Intake on Muscle Mass,
Muscle Strength, and Physical Performance in
Middle-Aged to Older Adults with or without
Existing Sarcopenia: A Systematic Review and
Meta-Analysis
Nivine I Hanach, Fiona McCullough, and Amanda Avery
Division of Nutritional Sciences, University of Nottingham, Leicestershire, United Kingdom
ABSTRACT
Sarcopenia is an age-related condition associated with a progressive loss of muscle mass and strength. Insufficient protein intake is a risk factor for
sarcopenia. Protein supplementation is suggested to improve muscle anabolism and function in younger and older adults. Dairy products are a
good source of high-quality proteins. This review evaluates the effectiveness of dairy proteins on functions associated with sarcopenia in middle-
aged and older adults. Randomized controlled trials were identified using PubMed, CINAHL/EBSCO, and Web of Science databases (last search:
10 May 2017) and were quality assessed. The results of appendicular muscle mass and muscle strength of handgrip and leg press were pooled
using a random-effects model. The analysis of the Short Physical Performance Battery is presented in narrative form. Adverse events and tolerability
of dairy protein supplementation were considered as secondary outcomes. Fourteen studies involving 1424 participants aged between 61 and 81
y met the inclusion criteria. Dairy protein significantly increased appendicular muscle mass (0.13 kg; 95% CI: 0.01, 0.26 kg; P=0.04); however, it had
no effect on improvement in handgrip (0.84 kg; 95% CI: 0.24, 1.93 kg; P=0.13) or leg press (0.37 kg; 95% CI: 4.79, 5.53 kg; P=0.89). The effect
of dairy protein on the Short Physical Performance Battery was inconclusive. Nine studies reported the dairy protein to be well tolerated with no
serious adverse events. Although future high-quality research is required to establish the optimal type of dairy protein, the present systematic review
provides evidence of the beneficial effect of dairy protein as a potential nutrition strategy to improve appendicular muscle mass in middle-aged
and older adults. Adv Nutr 2018;0:1–11.
Keywords: sarcopenia, muscle mass, muscle strength, physical performance, dairy protein, systematic review, meta-analysis, middle-age and older
adults
Introduction
Sarcopenia, the term used to dene age-related progressive
decline in muscle mass and muscle strength, was rst
reported by Rosenberg in 1989 (1). This condition can occur
with or without a reduction in fat mass (1). There is not a
specic age at which muscle mass and strength begin to di-
minish because of various contributing factors, including diet
and physical activity levels. However, the sarcopenic process
starts as early as the fourth or fth decade of life (2). Sarcope-
nia is recognized as an increasing public health problem (3),
The authors declared no nancial support received for this work.
Author disclosures: NIH, FM, and AA, no conicts of interest.
Address correspondence to AA (e-mail: amanda.avery@nottingham.ac.uk).
Abbreviations used: AMM, appendicular muscle mass; CG, control group; IG, intervention
group; SPPB, Short Physical Performance Battery.
with the number of people aected by sarcopenia worldwide
projected to increase from >50 million in 2010 to >200
million in 2050 (4). Sarcopenia is associated with increased
risk of adverse outcomes such as a poor quality of life, physi-
cal disability, depression, injurious falls, hospital admissions,
and death (3). As a consequence, high health care expendi-
tures of $900/person per year are attributed to sarcopenia (5).
The etiology of sarcopenia is multifactorial; it includes
increased inammatory mediators (e.g., cytokines), bed rest
or low physical activity levels, hormonal disorders (6), and
poor nutrition, particularly an inadequate energy and/or
protein intake (4,7). Nutrition is a modiable risk factor
for sarcopenia (8). For example, dietary protein enhances
the anabolic activity in skeletal muscle and provides the
© 2018 American Society for Nutrition. All rights reserved. Adv Nutr 2018;0:1–11; doi: https://doi.org/10.1093/advances/nmy065. 1
necessary amino acids to stimulate postprandial muscle pro-
tein synthesis (9). Insucient protein consumption has been
associated with muscle mass depletion and poor physical
function in older adults (8). A number of studies have been
carried out to evaluate the potential eect of protein or
amino acid supplementation on reducing the decline in func-
tions associated with sarcopenia (i.e., muscle mass, muscle
strength, and physical performance) in younger and older
adults. In a meta-analysis of 22 randomized controlled trials
(10), protein supplementation after prolonged resistance
training was shown to increase lean muscle mass by 0.69 kg
(P<0.00001) and leg press strength by 13.5 kg (P<0.005)
compared with a placebo group in both younger and older
adults. The majority of the included trials supplemented with
whey protein from dairy, alone or in combination with amino
acids or another type of dairy protein. In a recent meta-
analysis of 10 trials, Hidayat et al. (12)foundapositive
eect of milk-based protein supplementation and resistance
training on fat-free mass (0.74 kg; 95% CI: 0.30, 1.17 kg) in
older adults, suggesting a potential role of dairy protein in
promoting muscle anabolism.
Dairy products are good sources of high-quality protein,
primarily in the form of either whey or casein (11). They
are aordable and readily available throughout the world
(12). Dairy products, including milk-protein supplements,
do not require cooking or require only minimal preparation
compared with other protein-rich foods such as lean meat,
poultry, sh, and eggs (12). This makes dairy sources a
practical option for older adults to consume adequate protein
(12). Further research is required to determine if dairy
proteins could be used as a dietary strategy to reduce the
health risks associated with sarcopenia by improving muscle
mass and muscle function.
Although previous meta-analyses have evaluated the ef-
fect of protein supplementation on muscle mass and strength,
the potential dietary role of dairy protein in improving the
primary outcome parameters of sarcopenia has not been
specically investigated to our knowledge. Therefore, this
systematic review and meta-analysis aimed to evaluate the
impact of dairy protein intake on muscle mass, muscle
strength, and physical performance in middle-aged to older
adults with or without existing sarcopenia.
Methods
The current systematic review was conducted according to
the Preferred Reporting Items for Systematic Reviews and
Meta-Analyses (PRISMA) statement (13).
Eligibility criteria
The selection of the studies was restricted to full-text articles
and the English language. The relevant studies had to meet
the following criteria:
Participants: Studies conducted in humans only. Middle-
aged to older adults [middle age dened as between 45 and
65 y old (14)] with or without sarcopenia.
Types of study: Randomized controlled trials in which
the recruited subjects were randomly assigned to 1
TAB LE 1 Cutos of SPPB scores in older adults1
SPPB score Performance level
4–6 Low
7–9 Intermediate
10–12 High
1Data from referen ce 17. SPPB, Short Physical Performance Battery.
intervention group (IG) compared with a control group
(CG).
Types of intervention: The IG received dairy protein
supplementation (e.g., whey protein, milk-protein con-
centrate, casein) or a protein-based dairy product (e.g.,
ricotta cheese). The duration of the intervention was
12 wk. This period of time was decided on the basis
of the available evidence that muscle hypertrophy occurs
within 12 wk in response to dietary modications and
protein supplementations during resistance training (15,
16). Inclusion of resistance training as part of the study
intervention was optional.
Types of outcome measures: Primary outcomes were
changes in muscle strength (kilograms) of handgrip and
leg press measured by hydraulic hand dynamometer and
1-repetition-maximum strength test, respectively, and
changes in appendicular skeletal muscle mass (kilograms)
measured by DXA. Physical performance was assessed us-
ing the Short Physical Performance Battery (SPPB) score.
The SPPB is a standard measure of physical performance,
itassessestheindividualsbalance,strength,gait,anden-
durance (4). The score is calculated by summing the scores of
3 equally weighted tests: balance, gait speed, and chair stand
(4). Table 1 denes the cuto points of SPPB score in older
adults (17).
These variables have been suggested as primary outcome
domains to dene sarcopenia by the European Work Group
on Sarcopenia in Older People (4), with the proposed
measuring tools conrmed to be reliable and valid. Adverse
events and intervention adherence were reported as the
secondary outcomes. Exclusion criteria are presented in
Table 2.
Search strategy
PubMed (https://www.ncbi.nlm.nih.gov/pubmed/), CINAHL
(EBSCO) (https://www.ebsco.com/products/research-data
bases/cinahl-database), and Web of Science (https://
clarivate.com/products/web-of-science/databases/)databas-
es were used for electronic searches. References of the
retrieved studies and existing meta-analyses were also
hand-searched. The search covered the period up to 10 May
2017. There were no publication date or publication type
restrictions in this review. Key terms included in the search
engines were as follows—study design: #1 randomized
controlled trial OR controlled trial OR clinical trial;
population: #2 adult OR older adults OR middle-aged adults;
exposure: #3 milk OR milk protein OR dairy protein OR
whey OR whey protein OR whey protein supplementation
OR casein OR protein supplementation; outcome: #4 muscle
2 Hanach et al.
TAB LE 2 Exclusion criteria
Exclusion criteria
Study design Observational studies, meta-analyses, systematic reviews
Population Animals; children; subjects diagnosed with liver disease, kidney disease, or cancer; allergic/intolerant to milk protein or
lactose; taking medications that might interfere with the intervention; with low cognitive function; taking protein
supplementation other than the intervention dairy protein of study of interest
Outcome Postprandial protein synthesis, muscle protein synthesis, muscle fibers, muscle biopsy
mass OR skeletal muscle mass OR appendicular muscle mass
#5 muscle strength #6 physical performance OR physical
function OR functionality #7 (#4 OR #5 OR #6) #8 (#1 AND
#2 AND #3 AND #7).
Data extraction
The following data were extracted and tabulated: author(s),
year of publication, country of publication, study design,
duration of intervention, participants’ characteristics (e.g.,
sample size, baseline characteristics, inclusions), description
ofthestudyarms,measuredoutcomes,andvaluesofthe
outcomes of interests pre- and postintervention.
Quality assessment
The quality of the included studies was assessed according
totheCochraneCollaborationstoolforassessingrisk
of bias (18). It addresses 6 specic domains: sequence
generation, allocation concealment, blinding of participants,
personnel and outcome assessors, incomplete outcome data,
and selection reporting. In this review, as suggested by the
Cochrane Handbook for systematic reviews of interventions
(18),aqualityscaletoassesstheoverallriskofbiaswas
not used. The use of quality scales to appraise the included
randomized trials tends to combine the assessments of
aspects of the quality reporting with those of trial conduct
(19). Instead, the judgments of overall risk of bias are made
explicit by separating the assessment of internal and external
validity (19). The study selection, data extraction, and quality
assessment were primarily performed by NIH with oversight
by 2 experienced researchers (AA and FM). Dierences were
resolved by consensus.
Statistical analysis
RevMan software (Review Manager, version 5.3.5; The
Nordic Cochrane Centre, The Cochrane Collaboration, 2014;
http://community.cochrane.org/tools/review-production-
tools/revman-5/revman-5-download)wasusedtoperform
the meta-analysis. The mean change (meannal –mean
baseline)
of handgrip strength and appendicular muscle mass (AMM)
andmeanoutcomevalue(nalvalue)oflegpressstrength
were retrieved for analysis. When only the baseline and
nal SDs were reported, the change SD was computed
using the Cochrane Handbook proposed equation (18), as
follows:
SD change =(SDbaseline)2+(SDfinal )2
(2×corr ×SDbaseline ×SDfinal)(1)
where the imputed correlation coecient is 0.80. Eect sizes
are presented as mean dierences (95% CIs) for the contin-
uous outcomes. The results were pooled using the inverse
variance random-eects model (DerSimonian and Laird). I2
statistics were used to assess heterogeneity between studies.
The I2indicates the percentage of the variability in eect
estimates across studies due to heterogeneity rather than
sampling error (I2>50%: substantial heterogeneity) (19).
The xed-eects model was used when no heterogeneity was
identied. A Pvalue <0.05 was considered to be signicant.
To investigate whether the changes in primary outcomes
were aected by the subjects’ characteristics and individual
intervention, a subgroup analysis was conducted adjusting
for subjects’ mean age, subjects’ health status, and amount
of protein supplementation. In addition, a sensitivity analysis
was performed by excluding a single study, in turn, to
investigate its eect on the results of the meta-analysis.
Results
Literature search
Figure 1 shows the ow of the studies through the review
process. A total of 202 articles were identied after the
electronic search of PubMed, CINAHL/EBSCO, and Web
of Science databases. Four articles were retrieved after the
manual search of references of key articles and existing
reviews. The abstract screening resulted in the exclusion
of 191 articles not meeting inclusion criteria: 8 studies
evaluated the increase in the muscle fractional synthesis
rate, 5 studies were not randomized controlled trials, 1
study was animal-based, and the remaining studies contained
irrelevant content. After the full-text screening for eligibility,
one article was excluded for not providing the exposure of
interest. A total of 14 studies were therefore included in this
systematic review, and 11 were then included in the meta-
analysis. The publication dates ranged from 2009 to 2016.
The included studies were conducted in Mexico (2), United
States (3), Netherlands (3), Iceland (1), Finland (1), Germany
(1), Canada (1), Australia (1), and Ireland (1).
Study characteristics
Table 3 presents a summary of the characteristics of the
included studies. The eligible randomized controlled trials
included a total of 1424 participants with a mean ±SD age
range between 61 ±5yand81±1 y. Participants’ character-
istics varied between studies. Individuals with sarcopenia and
polymyalgia rheumatism were recruited in 3 trials (20–22)
and 1 trial (23), respectively, whereas the remaining studies
Dairy protein intake and sarcopenia: a review 3
Records identified through PubMed,
CINAHL/EBSCO, and Web of Science databases
searching
(n= 202)
Additional records identified through other
sources
(n=4)
Records after duplicates removed
(n=206)
Full-text articles excluded, with reasons
(n=1)
the exposure of interest is not a protein- based
supplement or dairy product
Records screened
(n=206)
Full-texts screening for eligibility
(n=15)
Records excluded
(n=191)
8 the outcome of interest was the muscle
fractional synthesis rate
2 cross-sectional studies
3 animal-based interventions
1 cohort studies
177 irrelevant studies
Studies included in qualitative synthesis
(n= 14)
Studies included in quantitative synthesis (meta-
analysis)
(n= 11)
FIGURE 1 PRISMA flow diagram of the selection of the studies. PRISMA, Preferred Reporting Items for Systematic Reviews and
Meta-Analyses.
enrolled healthy individuals. The subjects were nonfrail and
fully mobile in all studies with the exception of 3 in which
theyhadalimitedmobility(21,24,25).
Intervention
The duration of the intervention varied from 12 to
24 wk. The dairy protein supplement used in the IG varied
between studies. Two studies provided ricotta cheese with
the habitual diet (20,26). Five studies supplemented whey
protein either alone (23,25) or in combination with leucine
and vitamin D (21,22,28). Two studies supplemented
leucine- (23) and cysteine- (29) enriched whey protein. One
study supplemented skimmed milk–based high protein with
whey (28). Milk-protein concentrate was given in 2 studies
(24,31), and a milk-based matrix and casein hydrolysate were
used in 2 studies as a supplement (32,33). Only in one study
(33) was the amount of protein supplementation reported
according to body weight (0.33 g/kg), whereas the remaining
studies reported the extra protein intake according to daily
amounts ranging from 14 to 40 g/d. In all but 2 trials
the protein supplement was given daily; in the 2 studies
(27,32) it was consumed on “training” days only. The
frequency of protein supplementation varied among the
studies from 1 time/d (22,27–32), 2 times/d (21,23–25,29,
33), or 3 times/d (20,26). Seven of the included trials (22,
25,27–29,31,32) integrated resistance training as part of the
intervention, which ranged from 3 to 5 times/wk.
Comparison
In 2 studies (20,26),thesubjectsintheCGwereaskedto
follow their habitual diet. In 6 studies, an isocaloric product
wasprovidedasasupplementtotheCG(21,22,25,27,
28,33). Three studies used a placebo supplement (24,31,
32). A regular dairy product, casein, and skim milk–based
supplement were given to the CG in Björkman et al. (23),
Karelis et al. (29), and Zhu et al. (30), respectively.
Quality assessment
Figure 2 shows the risk-of-bias graph of the included
randomized controlled trials. According to the Cochrane
Collaboration tool for risk of bias (18), 64% of the tri-
als reported adequate random sequence generation and
allocation concealment, 86% had blinded participants and
personnel, 79% had blinded outcome assessors, 93% of the
4 Hanach et al.
TAB LE 3 Characteristics of the randomized controlled trials on dairy protein and the outcome variables of sarcopenia in middle-aged to older adults1
Study (ref) Region
Study
duration
Age,
y
Subjects,
nSex Health status
Type o f
protein
Total
amount
of protein2Comparator
RT
frequency Measured outcomes
Alemán-Maeto et al. (20)Mexico 3mo 60 40 F, M Sarcopenic Ricotta cheese 15.7 g/d Habitual diet NA AMM; MS of handgrip; AEs
Bauer et al. (21)Germany13wk65 380 F, M Sarcopenic,
limited
mobility
Whey protein 40 g/d Isocaloric drink NA AMM, MS of handgrip, SPPB
Rondanelli et al. (22) United States 12 wk 65 130 F, M Sarcopenic Whey protein 22 g/d Isocaloric
maltodextrin
drink
5 d/wk MS of handgrip
Björkman et al. (23) Finland 20 wk >50 46 F, M Polymyalgia
rheumatic
Whey protein 14 g/d Casein-based
dairy product
NA AMM, MS of handgrip
Tieland et al. (24) Netherlands 24 wk 65 65 F, M Frail MPC 30 g/d Placebo,
carbohydrate
drink
NA AMM; MS of handgrip; MS of leg
press; SPPB
Chalé et al. (25) United States 6 mo 70–85 67 F, M Limited
mobility
Whey protein 40 g/d Isocaloric
maltodextrin
drink
3 d/wk MS of leg press; SPPB
Alemán-Maeto et al. (26)Mexico 3mo 60 90 F, M Healthy Ricotta cheese 15.7 g/d Habitual diet NA AMM; MS of handgrip; SPPB, AEs
Arnarsonetal.(27) Iceland 12 wk 65–91 141 F, M Healthy Whey protein 20 g/d Isocaloric drink 3 d/wk AMM
Verreijen et a l. (28) Netherlands 13 wk >55 65 F, M Obese Whey protein 20–40 g/d4Isocaloric drink 3 d/wk AMM; MS of handgrip
Karelis et al. (29) Canada 135 d 65–88 80 F, M Healthy Cysteine-rich
whey
protein
20 g/d Casein 3 d/wk MS of leg press
Zhu et al. (30) Australia 2 y 70–80 181 F Healthy Skim
milk–based
high protein
30 g/d Skim milk–based
supplement
NA AMM; MS of handgrip
Leenders et al. (31) Netherlands 24 wk 70 ±1 57 F, M Healthy MPC 15 g/d Placebo, isocaloric
drink
3 d/wk MS of leg press
Verdijk et al. (32) United States 12 wk 72 ±2 28 F, M Healthy Casein
hydrolysate
20 g/d Placebo, flavored
drink
3 d/wk MS of leg press
Norton et al. (33) Ireland 24 wk 45–60 60 F, M Healthy Milk-based
protein
matrix3
0.33 g/kg Isocaloric drink NA AMM
1AE, adverse event; AMM, appendicular muscle mass; MPC, milk-protein concentrate; MS, muscle strength; NA, not available; RT, resistance training; SPPB, Short Physical Performance Battery.
2The amount of extra protein provided by the test supplement.
3The milk protein matrix is composed of a 9:2:1 ratio of milk-protein concentrate, whey-protein concentrate, and whey-protein isolate, respectively.
4The 40 g of protein was given on the training days only.
Dairy protein intake and sarcopenia: a review 5
FIGURE 2 Risk-of-bias graph. Review authors’judgments about each risk-of-bias item presented as percentages across all included
studies using the Cochrane risk-of-bias tool (18).
trials addressed adequately the incomplete outcome data,
and 100% of the trials were free of selective reporting. The
high risk of bias was mainly attributed to performance and
detection bias (20,26,28)andtoattritionbias(28). There
was insucient information to assess the degree of selection
bias in 6 trials (20,23,25,26,29,31)(Figure 3).
Primary outcomes
Muscle strength of handgrip. The meta-analysis of the
mean dierences in mean change in muscle strength of
handgrip included 7 studies with 435 participants in each
oftheIGsandCGs.Thedairyproteinhadnoeectonthe
improvement in handgrip strength (mean dierence: 0.84 kg;
95% CI: 0.24, 1.93 kg; P=0.13) (Figure 4). A signicant
substantial heterogeneity was shown between the studies
(I2=82%, P<0.00001). When a sensitivity analysis was
performed, the exclusion of Rondanelli et al. (22)ledtothe
absence of between-study heterogeneity (I2=0%), without
altering the overall results (mean dierence: 0.17 kg; 95%
CI: 0.25, 1.59 kg; P=0.43). Therefore, the results must
be interpreted with caution. In Björkman et al. (23), the
measurement of handgrip strength of both hands showed a
signicant decrease in the right handgrip strength (5.2%;
P<0.001) but did not aect the left handgrip strength (3.7%;
P=0.659) after the consumption of milk protein (IG). The
ndingsofthistrialwerenotincludedintheanalysisdueto
dierences in the way the data was reported.
Muscle strength of leg press. The meta-analysis of the
mean dierence in mean endpoint values of muscle strength
of leg press included 4 studies with 114 participants in
the IG and 109 participants in the CG. The dairy protein
supplementation had no eect on leg press strength, with
a mean dierence of 0.37 kg (95% CI: 4.79, 5.53 kg;
P=0.89) (Figure 5). Similar ndings were obtained when a
sensitivity analysis was performed. In Leenders et al. (31), the
data were reported as percentage change from the baseline,
whichwasnotconvenienttobeincludedintheanalysis.A
signicant improvement in leg press strength was found in
both women and men after the 24-wk intervention (31% and
26%, respectively; P<0.001), with no dierence between the
study arms (P=0.37)
AMM. AMM was assessed in 9 trials, of which 8 were
included in the meta-analysis. The meta-analysis of the mean
dierence in mean change of appendicular muscle mass
included a total of 444 and 457 participants in the IGs and
CGs,respectively.IncomparisontotheCG,dairyprotein
supplementation resulted in a signicant increase in AMM
(mean dierence: 0.13 kg; 95% CI: 0.01, 0.26 kg; P=0.04)
(Figure 6). No changes in the results were detected after a
sensitivity analysis was done. Björkman et al. (23)reported
no dierence in AMM improvement between the groups (IG
compared with CG: 0.9% compared with 0.2%; P=0.510).
SPPB. Four out of the 14 included studies evaluated the
SPPB (21,24–26). Tieland et al. (24)andChaléetal.(25)
reported a signicant increase in SPPB score from baseline
to postintervention compared with the CG [8.9 ±0.6 to
10 ±0.6 (P=0.02) and 8.5 ±1.1 to 10.3 ±1.5 (P<0.0001),
respectively], although Alemán-Mateo et al. (26)andBauer
et al. (21) found no eect of dairy protein on the SPPB at the
end of the study [10.7 ±1.7 to 10.8 ±1.5 (P=0.55) and 7.5
to 8.36 (P=0.51), respectively].
Subgroup analyses. A stratied analysis was conducted
accordingtosubjects’healthstatus(withorwithoutsarcope-
nia), mean age, and amount of extra protein supplemen-
tation. The subgroup analysis by sex and type of protein
supplement was dicult to be perform due to only 3 out of
the14includedtrials(20,27,31) reporting sex dierences
in the main outcomes and to the marked dissimilarity in
the given type of protein among the studies. Similarly, the
subgroup analysis stratied by the integration of resistance
training was not possible to be performed due to dierences
in the length and type of workout program among the studies.
There was no signicant change in handgrip strength and
AMM across the subgroups (Tab l e 4 ).
6 Hanach et al.
FIGURE 3 Risk-of-bias summary. Review authors’judgments
about each risk-of-bias item for each included study using the
Cochrane risk-of-bias tool (18).
Secondary outcomes
Nine of the included studies included adverse events in
their reporting. The assessment of safety and tolerability of
the study supplements varied between studies. For instance,
Alemán-Mateo et al. (20,26) measured the relative change
of lipid prole, glomerular ltration rate, kidney function
markers, and microalbuminuria. In the 7 remaining trials,
the gastrointestinal tolerance of the supplement was assessed.
None of the included trials found a signicant dierence in
the incidence of serious adverse events between study arms.
Alemàn-Mateo et al. (20) found that the consumption of
ricotta cheese was associated with early satiety among 25%
of the female participants, and Björkman et al. (23)found
that some gastrointestinal side eects (e.g., early satiety,
diarrhea, atulence, and nausea) were reported by 44.7%
in the IG compared with 32.6% participants in the CG
(P=0.180).
The intervention product was generally well tolerated, and
the compliance rate ranged from 72.1% to 100% among the
studies. In Zhu et al. (30), the adherence to the intervention
was signicantly higher in the IG compared with the CG
(87.1% compared with 80.8%, respectively; P=0.03).
Discussion
The aim of this review was to determine if dairy proteins
couldbeusedtoreducethehealthrisksassociatedwith
sarcopenia by improving muscle mass, muscle function,
and physical performance in middle-aged to older adults.
A meta-analysis was performed to assess the improvement
in muscle strength of handgrip and leg press and AMM.
The results showed a signicant favorable eect of dairy
protein, at amounts of 14–40 g/d, on AMM without having an
eect on muscle strength of handgrip and leg press. Overall,
compliance and acceptability of the protein supplementation
were reported to be good across the studies. Dierences in
the amount of protein supplementation and subjects’ mean
age and baseline health status between the studies did not
signicantly inuence the ndings.
Previous studies have reported a direct relation between
muscle mass and strength in middle-aged to older adults.
Hayashida et al. (34) conducted a cross-sectional study in 318
individuals with the aim to evaluate the correlation between
muscle mass and muscle strength on the basis of sex and
age groups. The obtained results conrmed a signicant
association between muscle strength and AMM in men aged
65 y and women aged 75 y; however, Goodpaster et al.
(35) suggested in a prospective study in 1880 older adults
with a mean age of 73.5 ±2.8 y that the loss of muscle
strength is more rapid than the loss of muscle mass and
that the decline in the age-dependent strength cannot be
explained by the loss of muscle mass alone.
It was previously argued that older adults tend to develop
a phenomenon called anabolic resistance, an impaired
response of skeletal muscle to an anabolic stimulus with
normal muscle protein synthesis (36). Nevertheless, Burd
et al. (36) observed that, although many factors contribute
to the anabolic resistance of muscle protein synthesis in
older adults, minimal dierences could be seen in muscle
protein synthesis rates between young and older adults after
protein ingestion (36). Hence, the diversity of age groups
in this review probably has little impact on the signicant
improvement of AMM reported (P=0.04).
With regard to physical performance, the analyzed data
do not allow us to make any conclusions about the potential
Dairy protein intake and sarcopenia: a review 7
FIGURE 4 Forest plot showing results for the meta-analysis of difference in mean change from baseline in muscle strength (kilograms)
for handgrip after the intervention in middle-aged to older adults. IV, inverse variance.
eect of dairy protein on SPPB score in middle-aged to
older adults. Two of the included trials reported a signicant
increase in SPPB score (24,25).InBaueretal.(21), although
the results showed no dierence in SPPB score between
the groups (P=0.51), a signicant improvement in chair-
rise time was reported in the IG compared with the CG
(P=0.018). A better chair-stand performance has been
shown to be related to a better leg extensor power in middle-
aged and older adults (37). Cesari et al. (38) claimed that the
chair-stand test has the highest prognostic value compared
with the other SPPB subtasks. Because the chair stand has
been suggested as a useful measure of physical function, this
could partially explain the potential benets of dairy protein
on physical performance.
There are limited scientic data on dairy protein and
sarcopenia available. To the best of our knowledge, this is
the rst review to determine the impact of dairy protein
intake on muscle mass, muscle strength, and physical
performance in middle-aged to older adults with or without
existing sarcopenia. However, the ndings of the review
are limited and the following factors must be considered
when interpreting the results. First, there were dierences
in health status (sarcopenic compared with healthy subjects)
and physical functionality (e.g., frailty and mobility) of the
participants in the included trials at baseline. According to
Alemán-Mateo et al. (26), the responsiveness to the anabolic
stimulus of protein supplementation is likely to be more
eective in healthy subjects than in those with sarcopenia.
Second, the amount, regimen, form of administration (food
compared with supplements), and types of dairy protein used
(e.g., whey, milk-protein concentrate, casein) varied between
the studies (Tab l e 3), which makes it challenging to interpret
the ndings. Third, the baseline dietary protein intake, an
important confounding factor, was only measured in 7 of the
included trials (21,24,25,31–33). Campbell and Leidy (39)
armed that protein supplementation induces a signicant
enhancement in muscle mass and strength in individuals
with a habitual dietary protein intake below the RDA (<0.8
gkg1d1), and this was consistent with the results of
Verdjik e t a l . ( 32)andTielandetal.(24). Only Bauer et al.
(21) adjusted for baseline dietary protein intake. This might
have inuenced the precision of the ndings and created bias.
Dairy protein was not the only supplement in all the
trials. Bauer et al. (21), Verreijen et al. (28), and Rondanelli
et al. (22) enriched the dairy protein supplement with
leucineandvitaminD.Dairyprotein,suchasnativewhey,
is naturally a good source of the essential amino acid leucine,
which has been reported to stimulate protein synthesis and
enhance muscle mass and function (40,41). In addition,
vitamin D has been shown to induce a signicant positive
impact on muscle strength in the elderly (42). Therefore,
the absence of adjustment of the postintervention serum 25-
hydroxyvitamin D in addition to the anabolic eect of leucine
could have created an overestimation of the positive eect
of dairy protein in these trials and thus have limited the
accuracy of the ndings.
A further confounding factor is the incorporation of
physical activity in some of the included studies. In Verdjik
FIGURE 5 Forest plot showing results for the meta-analysis of difference in the mean endpoint value of muscle strength for leg press
(kilograms) after the intervention in middle-aged to older adults. IV, inverse variance.
8 Hanach et al.
FIGURE 6 Forest plot showing results for the meta-analysis of difference in mean change from baseline in appendicular muscle mass
(kilograms) after the intervention in middle-aged to older adults. IV, inverse variance.
et al. (32)andChaléetal.(25), the study arms were involved
in resistance training 3 times/wk. The authors reported a
signicant improvement in muscle strength in both groups,
regardless of the consumption of protein supplementation,
which underlines the possible eect of physical activity on
the outcomes.
Overall, 6 trials were of a high quality and had all the key
domains at low risk of bias. The high risk of bias in certain
trials was attributable to the lack of blinding of participants
and/or outcome assessors (20,26,32)andtomissingdataof
the primary outcome (28). Thus, the accuracy of the ndings
of these trials might be limited and should be interpreted with
caution. Although in some studies the allocation was stated to
be randomly generated and concealed, a detailed description
of the methods used was not clearly reported. This has
raised some uncertainty about the results and rendered the
judgment of risk of bias as unclear.
This systematic review has several limitations: 1)broad
inclusion criteria with minimal restrictions on the type
of intervention; 2) the presence of a high degree of het-
erogeneity in the data reporting among the studies; 3)
lack of adjustment for baseline protein intake; 4) lack of
publication bias assessment; 5) lack of postintervention
follow-up to evaluate the long-term eect of dairy protein
supplementation; 6) the relatively small sample sizes, which
inuences the external validity of the trials and restricts
the ndings to be generalized to the entire population; 7)
lack of blinding of participants and outcome assessors and
incomplete data reporting increased the risk of bias in certain
trials (Figure 3) and thus aected the results of the meta-
analysis; 8) only 6 of the included studies were of high
quality; 9) lack of subgroup analysis stratied by gender,
type of protein, and the integration of resistance training;
10) the methodologic diversity between the studies; and 11)
the assessment of muscle mass does not predict the physical
functioning (4).
However,thesystematicreviewandmeta-analysisalsohas
the following strengths: 1) this is the rst systematic review
and meta-analysis to highlight the possible eect of dairy
protein consumption on the outcome variables of sarcopenia,
which could be clinically meaningful for the general popula-
tion; 2) the review was conducted according to the PRISMA
statement (13); 3)allthemeasuringtoolsthatwereused
to assess the outcomes of interest were proved to be valid
and reliable and thus can be applied in clinical and research
settings; 4) the inclusion of a considerable number of studies
in the meta-analysis; 5) despite the diversity in the provided
type of dairy protein, all of the included trials reported a high
adherence and tolerance to the exposure, which conrmed
the safety of protein supplementation; and 6)theassessment
TAB LE 4 Subgroup analyses of mean changes in muscle strength for handgrip and AMM according to subjects’health status, mean age,
and amount of protein consumed1
Changes in handgrip strength, kg Changes in AMM, kg
nEffect (95% CI) PI
2,% nEffect (95% CI) PI
2,%
Health status
With sarcopenia 3 1.62 (0.94, 4.18) 2 0.17 (0.00, 0.33)
0.26 21.9 0.79 0
Without sarcopenia 4 0.11 (0.40, 0.62) 6 0.13 (0.07, 0.33)
Amount of protein
<20 g/d 3 0.74 (0.95, 2.44) 3 0.30 (0.57, 0.96)
0.91 0 0.87 0
20 g/d 4 0.86 (0.44, 2.17) 5 0.14 (0.02, 0.29)
Mean age
>65 y NA 6 0.45 (0.09, 0.98) 0.20 40.4
65 y 2 0.08 (0.03, 0.20)
1AMM, appendicular muscle mass; NA, not available.
Dairy protein intake and sarcopenia: a review 9
ofhandgripstrength,whichisagoodindicatorofphysical
capability (4).
In conclusion, the ndings of this systematic review and
meta-analysis suggest that dairy proteins, at an amount of 14–
40 g/d, can signicantly increase the AMM in middle-aged
and older adults without a signicant clinical eect on muscle
strength of handgrip and leg press. The eect of dairy protein
on the SPPB was inconclusive due to insucient reported
data. This review highlights the need for larger-scale and
high-quality randomized controlled trials with longer follow-
ups using standardized primary outcomes (muscle mass,
muscle strength, and physical performance) when investigat-
ing the role of dairy protein in the prevention and treatment
ofsarcopenia;toestablishtheoptimaltype,amount,timing,
and frequency of dairy protein supplementation in middle-
aged to older adults; and subsequently, to examine its clinical
eectiveness in the improvement of the primary outcomes of
sarcopenia in middle-aged to older adults.
Acknowledgments
All authors read and approved the nal manuscript.
References
1. Rosenberg IH. Sarcopenia: origins and clinical relevance. J Nutr
1997;127(5):990S–1S.
2. Mitchell CJ, McGregor RA, D’Souza RF, Thorstensen EB, Markworth
JF, Fanning AC, Poppitt SD, Cameron-Smith D. Consumption of milk
protein or whey protein results in a similar increase in muscle protein
synthesis in middle aged men. Nutrition 2015;7(10):8685–99.
3. Ethgen O, Beaudart C, Buckinx F, Bruyère O, Reginster JY. The future
prevalence of sarcopenia in Europe: a claim for public health action.
Calcif Tissue Int 2017;100(3):229–34.
4. Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T,
Landi F, Martin FC, Michel JP, Rolland Y, Schneider SM, et al.
Sarcopenia: European consensus on denition and diagnosis report
oftheEuropeanWorkingGrouponSarcopeniainOlderPeople.Age
Ageing 2010;39(4):412–23.
5. Janssen I, Shepard DS, Katzmarzyk PT, Roubeno R. The healthcare
costs of sarcopenia in the United States. J Am Geriatr Soc
2004;52(1):80–5.
6. Malafarina V, Uriz-Otano F, Iniesta R, Gil-Guerrero L. Eectiveness of
nutritional supplementation on muscle mass in treatment of sarcopenia
in old age: a systematic review. J Am Med Dir Assoc 2013;14(1):10–7.
7. Cooper LA, Brown SL, Hocking E, Mullen AC. The role of exercise,
milk, dairy foods and constituent proteins on the prevention and
management of sarcopenia. Int J Dairy Tech 2016;69(1):13–21.
8. Baum JI, Wolfe RR. The link between dietary protein intake, skeletal
muscle function and health in older adults. Healthcare 2015;3(3):529–
43.
9. Coker RH, Miller S, Schutzler S, Deutz N, Wolfe RR. Whey protein
and essential amino acids promote the reduction of adipose tissue and
increased muscle protein synthesis during caloric restriction-induced
weight loss in elderly, obese individuals. Nutr J 2012;11(1):105.
10.CermakNM,deGrootLC,SarisWH,vanLoonLJ.Protein
supplementation augments the adaptive response of skeletal muscle
to resistance-type exercise training: a meta-analysis. Am J Clin Nutr
2012;96(6):1454–64.
11. Wilkinson SB, Tarnopolsky MA, MacDonald MJ, MacDonald JR,
Armstrong D, Phillips SM. Consumption of uid skim milk promotes
greater muscle protein accretion after resistance exercise than does
consumption of an isonitrogenous and isoenergetic soy-protein
beverage. Am J Clin Nutr 2007;85(4):1031–40.
12. Hidayat K, Chen GC, WangY, Zhang Z, Dai X, Szeto IM, Qin LQ. Eects
of milk proteins supplementation in older adults undergoing resistance
training: a meta-analysis of randomized control trials. J Nutr Health
Aging 2018;22(2):237–45.
13. PRISMA GroupMoher D, Liberati A, Tetzla J, Altman DG;. Preferred
Reporting Items for Systematic Reviews and Meta-Analyses: the
PRISMA statement. PLoS Med 2009;6(7):e1000097.
14. Oxford English Dictionary [Internet]. 2017. [cited 2017 Jun 6]. Available
from: http://www.oed.com/.
15. Raguso CA, Kyle U, Kossovsky MP, Roynette C, Paoloni-Giacobino A,
Hans D, Genton L, Pichard C. A 3-year longitudinal study on body
composition changes in the elderly: role of physical exercise. Clin Nut
2006;25(4):573–80.
16. Meredith CN, Frontera WR, O’Reilly KP, Evans WJ. Body composition
in elderly men: eect of dietary modication during strength training. J
Am Geriatr Soc 1992;40(2):155–62.
17. Guralnik JM, Ferrucci L, Pieper CF, Leveille SG, Markides KS, Ostir GV,
Studenski S, Berkman LF, Wallace RB. Lower extremity function and
subsequent disability: consistency across studies, predictive models, and
value of gait speed alone compared with the short physical performance
battery. J Gereontol 2000;55(4):M221–31.
18. Higgins JP, Altman DG, Gøtzsche PC, Jüni P, Moher D, Oxman AD,
Savovi´
cJ,SchulzKF,WeeksL,SterneJA.TheCochraneCollaborations
tool for assessing risk of bias in randomised trials. BMJ 2011;343:d5928.
19. Higgins JP, Green S, editors. Cochrane Handbook for Systematic
Reviews of Interventions. London: John Wiley & Sons; 2011.
20. Alemán-Mateo H, Macías L, Esparza-Romero J, Astiazaran-García H,
Blancas AL. Physiological eects beyond the signicant gain in muscle
mass in sarcopenic elderly men: evidence from a randomized clinical
trial using a protein-rich food. Clin Interv Aging 2012;7:225.
21. Bauer JM, Verlaan S, Bautmans I, Brandt K, Donini LM, Maggio M,
McMurdo ME, Mets T, Seal C, Wijers SL, et al. Eects of a vitamin D
and leucine-enriched whey protein nutritional supplement on measures
of sarcopenia in older adults, the PROVIDE study: a randomized,
double-blind, placebo-controlled trial. J Am Med Dir Assoc 2015;16(9):
740–7.
22. Rondanelli M, Klersy C, Terracol G, Talluri J, Maugeri R, Guido D,
Faliva MA, Solerte BS, Fioravanti M, Lukaski H, et al. Whey protein,
amino acids, and vitamin D supplementation with physical activity
increases fat-free mass and strength, functionality, and quality of life
and decreases inammation in sarcopenic elderly. Am J Clin Nutr
2016;103(3):830–40.
23. Björkman MP, Pilvi TK, Kekkonen RA, Korpela R, Tilvis RS. Similar
eects of leucine rich and regular dairy products on muscle mass
and functions of older polymyalgia rheumatica patients: a randomized
crossover trial. J Nutr Health Aging 2011;15(6):462–7.
24. Tieland M, van de Rest O, Dirks ML, van der Zwaluw N, Mensink
M, van Loon LJ, de Groot LC. Protein supplementation improves
physical performance in frail elderly people: a randomized, double-
blind, placebo-controlled trial. J Am Med Dir Assoc 2012;13(8):
720–6.
25. Chalé A, Cloutier GJ, Hau C, Phillips EM, Dallal GE, Fielding RA.
Ecacy of whey protein supplementation on resistance exercise–
induced changes in lean mass, muscle strength, and physical function
in mobility-limited older adults. J Gereontol 2012;68(6):682–90.
26. Alemán-Mateo H, Carreón VR, Macías L, Astiazaran-García H,
Gallegos-Aguilar AC, Enríquez JR. Nutrient-rich dairy proteins
improve appendicular skeletal muscle mass and physical performance,
and attenuate the loss of muscle strength in older men and women
subjects: a single-blind randomized clinical trial. Clin Int Aging
2014;9:1517.
27. Arnarson A, Gudny Geirsdottir O, Ramel A, Briem K, Jonsson PV,
Thorsdottir I. Eects of whey proteins and carbohydrates on the ecacy
of resistance training in elderly people: double blind, randomised
controlled trial. Eur J Nutr 2013;67(8):821–6.
28. Verreijen AM, Verlaan S, Engberink MF, Swinkels S, de Vogel-van den
Bosch J, Weijs PJ. A high whey protein–, leucine-, and vitamin D–
enriched supplement preserves muscle mass during intentional weight
loss in obese older adults: a double-blind randomized controlled trial.
Am J Clin Nutr 2015;101(2):279–86.
10 Hanach et al.
29. Karelis A, Messier V, Suppère C, Briand P, Rabasa-Lhoret R.
Eect of cysteine-rich whey protein (Immunocal) supplementation
in combination with resistance training on muscle strength and lean
body mass in non-frail elderly subjects: a randomized, double-blind
controlled study. J Nutr Health Aging 2015;19(5):531.
30. Zhu K, Kerr DA, Meng X, Devine A, Solah V, Binns CW, Prince RL.
Two-year whey protein supplementation did not enhance muscle mass
and physical function in well-nourished healthy older postmenopausal
women. J Nutr 2015;145(11):2520–6.
31. Leenders M, Verdijk LB, Van der Hoeven L, Van Kranenburg J,
Nilwik R, Wodzig WK, Senden JM, Keizer HA, Van Loon LJ. Protein
supplementation during resistance-type exercise training in the elderly.
Med Sci Sports 2013;45(3):542–52.
32. Verdijk LB, JonkersRA, Gleeson B G, Beelen M, MeijerK, Savelberg HH,
Wodzig WK, Dendale P, van Loon LJ. Protein supplementation before
and after exercise does not further augment skeletal muscle hypertrophy
after resistance training in elderly men. Am J Clin Nutr 2009;89(2):608–
16.
33. Norton C, Toomey C, McCormack WG, Francis P, Saunders J, Kerin
E, Jakeman P. Protein supplementation at breakfast and lunch for 24
weeks beyond habitual intakes increases whole-body lean tissue mass
in healthy older adults. J Nutr 2016;146(1):65–9.
34. Hayashida I, Tanimoto Y, Takahashi Y, Kusabiraki T, Tamaki
J. Correlation between muscle strength and muscle mass, and
their association with walking speed, in community-dwelling elderly
Japanese individuals. PloS One 2014;9(11):e111810.
35. Goodpaster BH, Park SW, Harris TB, Kritchevsky SB, Nevitt M,
Schwartz AV, Simonsick EM, Tylavsky FA, Visser M, Newman AB.
The loss of skeletal muscle strength, mass, and quality in older
adults: the Health, Aging and Body Composition Study. J Gereontol
2006;61(10):1059–64.
36. Burd NA, Gorissen SH, van Loon LJ. Anabolic resistance of muscle
protein synthesis with aging. Exec Sport Sci Rev 2013;41(3):169–73.
37.HardyR,CooperR,ShahI,HarridgeS,GuralnikJ,KuhD.Ischair
rise performance a useful measure of leg power?. Aging Clin Exp Res
2010;22(5–6):412.
38. Cesari M, Onder G, Zamboni V, Manini T, Shorr RI, Russo A, Bernabei
R, Pahor M, Landi F. Physical function and self-rated health status
as predictors of mortality: results from longitudinal analysis in the
ilSIRENTE study. BMC Geriatr 2008;8(1):34.
39. Campbell WW, Leidy HJ. Dietary protein and resistance training eects
on muscle and body composition in older persons. J Am Coll Nutr
2007;26(6):696S–703S.
40.BauerJ,BioloG,CederholmT,CesariM,Cruz-JentoftAJ,Morley
JE, Phillips S, Sieber C, Stehle P, Teta D, et al. Evidence-based
recommendations for optimal dietary protein intake in older people: a
position paper from the PROT-AGE Study Group. J Am Med Dir Assoc
2013;14(8):542–59.
41.HamarslandH,NordengenAL,AasSN,HolteK,GartheI,PaulsenG,
Cotter M, Børsheim E, Benestad HB, Raastad T. Native whey protein
with high levels of leucine results in similar post-exercise muscular
anabolic responses as regular whey protein: a randomized controlled
trial. J Int Soc Sports Nut 2017;14(1):43.
42. Beaudart C, Buckinx F, Rabenda V, Gillain S, Cavalier E, Slomian J,
Petermans J, Reginster JY, Bruyère O. The eects of vitamin D on
skeletal muscle strength, muscle mass, and muscle power: a systematic
review and meta-analysis of randomized controlled trials. J Endocrinol
Metab 2014;99(11):4336–45.
Dairy protein intake and sarcopenia: a review 11
... Surgical stress and immobilization, which promote catabolism or decreased anabolism, often lead to a prolonged decrease in muscle strength following lower limb surgery [2,[48][49][50]. Protein and/or EAA supplementation has been shown to stimulate muscle protein anabolism, suggesting a possibility to improve muscle strength [51]. ...
... In previous SRs focusing on older adults, the effects of protein and/or EAA supplementation on muscle mass have yet to reach a consensus, with results showing various effects [14,51,52]. Nevertheless, it is essential to note that the studies on muscle strength and mass involved different surgeries. ...
Article
Full-text available
This study aimed to examine the efficacy and safety of protein and/or essential amino acid (EAA) supplementation in all lower limb surgeries using systematic reviews and meta-analysis of randomized controlled trials (RCTs). We included RCTs that assessed the efficacy of protein and/or EAA supplementation in lower limb surgeries. On June 2, 2023, we searched EMBASE, MEDLINE, the Cochrane Central Register of Controlled Trials, the World Health Organization International Clinical Trials Registry Platform, and ClinicalTrials.gov. The primary outcomes were mobility, patient-reported outcomes (PRO), and acute kidney injury (AKI). The secondary outcomes were exercise capacity, muscle strength, muscle mass, and all adverse events. We performed meta-analyses using the random-effects model. We assessed the risk of bias using the Cochrane risk-of-bias tool and the certainty of evidence using the Grading of Recommendations, Assessment, Development, and Evaluation approach. We included 12 RCTs (622 patients). These studies included four on hip fracture surgery, three on total hip arthroplasty, and five on total knee arthroplasty. Protein and/or EAA supplementation may slightly improve PRO (standard mean difference 0.51, 95% confidence interval (CI): 0.22 to 0.80, low certainty of evidence). Nevertheless, it may not improve mobility (mean difference 0.07 m/s, 95% CI: -0.01 to 0.16, low certainty of evidence). No adverse events including AKI were reported. Muscle strength may have increased (standard mean difference 0.31, 95% CI: 0.02 to 0.61, very low certainty of evidence). However, exercise capacity (mean difference 5.43 m, 95% CI: -35.59 to 46.45, very low certainty of evidence) and muscle mass (standard mean difference -0.08, 95% CI: -0.49 to 0.33, very low certainty of evidence) were not improved. While protein and/or EAA supplementation in lower limb surgeries may improve PRO, it is unlikely to affect mobility. Despite this, the medical team and patients might still consider protein and/or EAA supplementation a useful option.
... Surgical stress and immobilization, which promote catabolism or decreased anabolism, often lead to a prolonged decrease in muscle strength following lower limb surgery [2,[48][49][50]. Protein and/or EAA supplementation has been shown to stimulate muscle protein anabolism, suggesting a possibility to improve muscle strength [51]. ...
... In previous SRs focusing on older adults, the effects of protein and/or EAA supplementation on muscle mass have yet to reach a consensus, with results showing various effects [14,51,52]. Nevertheless, it is essential to note that the studies on muscle strength and mass involved different surgeries. ...
... In addition, bovine milk has been studied extensively for its ability to stimulate muscle protein synthesis [117,118], a process critical for preserving muscle mass in older adults who often experience sarcopenia, the progressive loss of muscle mass and function with age [119]. While findings from previous studies are largely promising, they tend to focus on isolated milk proteins rather than whole dairy products [118,120]. In our study, we will not directly assess the effects of milk consumption on muscle protein synthesis but will explore the impacts of incorporating whole milks in the daily diets of older women on their body composition, including lean muscle mass, physical function, and physical activity levels. ...
Article
Full-text available
Background: Age-related changes can lead to dietary insufficiency in older adults. The inclusion of high-quality, nutrient-dense foods such as ruminant milks can significantly improve health outcomes. However, many older adults worldwide do not meet daily milk intake recommendations because of digestive discomfort and health concerns. Ovine and caprine milks are increasingly popular for their perceived digestive and nutritional benefits. While preclinical studies suggest differences in milk digestion, human studies investigating acute postprandial responses remain inconclusive, and the impacts of sustained milk consumption remain uncertain. Objectives: Hence, we present a randomized controlled trial investigating how the sustained consumption of bovine, caprine, or ovine milk influences digestion, nutrition, and metabolism in older women. Methods: A total of 165 healthy older women were randomized to receive bovine, caprine, or ovine milk, or no milk, twice daily for 12 weeks. The primary outcome is the impact of milk consumption on digestive comfort assessed via the Gastrointestinal Syndrome Rating Scale (GSRS). Secondary outcomes include changes in nutrient intake, plasma amino acid and lipid appearance, bowel habits, the gut microbiota, cardiometabolic health, physical function, physical activity, sleep, mood, sensory perception, and emotional response. Conclusions: The findings could inform dietary recommendations for older women and facilitate the development of targeted functional food products.
... Reduced skeletal muscle mass not only decreases an individual's motor function but also increases the risk of several adverse outcome diseases such as diabetes mellitus, cardiovascular disease, osteoporosis, and many other diseases, ultimately leading to a decreased quality of life (5,6). Moreover, the impact of sarcopenia is gradually extending beyond the elderly population to younger age groups, possibly attributed to a lack of physical activity, poor dietary habits, and the increasing prevalence of chronic disorders in daily life (7)(8)(9). ...
Preprint
Full-text available
Background The impact of diet on people's health is indisputable. While animal and cell experiments may suggest a link between coffee intake and increased skeletal muscle mass, translating these findings to humans requires careful investigation. The aim of this research is to evaluate the correlation between adult American skeletal muscle mass and caffeine consumption. Methods This study was conducted among persons 20 years of age and above between 2011 and 2018, using information from the National Health and Nutrition Examination Survey (NHANES). We investigated the connection between skeletal muscle mass and caffeine intake using three multiple linear regression models. Afterwards, To look into variations in the correlation between caffeine consumption and skeletal muscle mass across several demographic attributes, such as gender, age, race, and body mass index (BMI) categories, subgroup analyses were conducted. Result A total of 8,125 participants met the inclusion criteria. All three multiple linear regression models indicated a positive correlation between caffeine intake and skeletal muscle mass. Age-stratified analysis showed significant positive correlations for participants aged 30 to 39 and 40 to 49 years old. BMI-stratified analysis revealed a significant positive correlation between caffeine intake and muscle mass among normal and overweight individuals Conclusions Our study results indicate a positive correlation between caffeine intake and muscle mass. Individuals aged 30–49 years and those with a normal or overweight BMI may potentially benefit more. Future cohort studies are necessary to confirm these conclusions and to explore the underlying mechanisms.
... Various approaches, including exercise, drug and nutritional supplementation, and hormone therapy, have been explored to mitigate muscle atrophy associated with sarcopenia [16]. Among these, nutritional therapy via supplementation with polyunsaturated fatty acids, high-quality protein, vitamins, and essential amino acids has been shown to prevent muscle weakness in elderly people by stimulating protein synthesis [17][18][19]. Soy foods are traditional Asian foods that alleviate muscle weakness by stimulating muscle protein synthesis and increasing antioxidant capacity [20]. Isoflavone, an abundant soybean flavonoid, has been studied in the context of preventing obesity, elevated blood sugar levels, osteoporosis, breast cancer, and its antioxidant properties in recent years [2]. ...
Article
Full-text available
Background: Sarcopenic obesity, which is associated with a poorer prognosis than that of sarcopenia alone, may be positively affected by soy isoflavones, known inhibitors of muscle atrophy. Herein, we hypothesize that these compounds may prevent sarcopenic obesity by upregulating the gut metabolites with anti-inflammatory effects. Methods: To explore the effects of soy isoflavones on sarcopenic obesity and its mechanisms, we employed both in vivo and in vitro experiments. Mice were fed a high-fat, high-sucrose diet with or without soy isoflavone supplementation. Additionally, the mouse C2C12 myotube cells were treated with palmitic acid and daidzein in vitro. Results: The isoflavone considerably reduced muscle atrophy and the expression of the muscle atrophy genes in the treated group compared to the control group (Fbxo32, p = 0.0012; Trim63, p < 0.0001; Foxo1, p < 0.0001; Tnfa, p = 0.1343). Elevated levels of daidzein were found in the muscles and feces of the experimental group compared to the control group (feces, p = 0.0122; muscle, p = 0.0020). The real-time PCR results demonstrated that the daidzein decreased the expression of the palmitate-induced inflammation and muscle atrophy genes in the C2C12 myotube cells (Tnfa, p = 0.0201; Il6, p = 0.0008; Fbxo32, p < 0.0001; Hdac4, p = 0.0002; Trim63, p = 0.0114; Foxo1, p < 0.0001). Additionally, it reduced the palmitate-induced protein expression related to the muscle atrophy in the C2C12 myotube cells (Foxo1, p = 0.0078; MuRF1, p = 0.0119). Conclusions: The daidzein suppressed inflammatory cytokine- and muscle atrophy-related gene expression in the C2C12 myotubes, thereby inhibiting muscle atrophy.
... On lean body mass, fat mass, handgrip strength, gait velocity, and chair-stand ability, however, the impact of extra protein supplementation during resistance exercise training was not shown to be superior to resistance training alone for older persons. These findings are consistent with several other meta-analyses that looked at how protein supplements affected handgrip strength [26,56e59], lean body mass, fat mass [55,57,60], and physical function [52,55,57,61,62]. However, there is a significant improvement in the handgrip strength of older adults performing RT in a meta-analysis conducted by Kirwan et al., 2021 [59]. ...
... 138,140,141 Furthermore, fortified dairy products have the potential to enhance skeletal muscle health as a nutrient-dense food. 142 These dairy products are fortified with vitamin D and rich in protein and various micronutrients, such as vitamin B12, calcium, riboflavin, and zinc, essential for maintaining muscle health and function. 143 It is worth noting that normalizing circulating vitamin D concentrations may be a critical factor in the efficacy of protein supplements. ...
Article
Full-text available
This article reviews the mechanisms and prevention strategies associated with vitamin D and sarcopenia in older adults. As a geriatric syndrome, sarcopenia is defined by a notable decline in skeletal muscle mass and strength, which increases the risk of adverse health outcomes such as falls and fractures. Vitamin D, an essential fat-soluble vitamin, is pivotal in skeletal muscle health. It affects muscle function through various mechanisms, including regulating calcium and phosphorus metabolism, promoting muscle protein synthesis, and modulation of muscle cell proliferation and differentiation. A deficiency in vitamin D has been identified as a significant risk factor for the development of sarcopenia in older adults. Many studies have demonstrated that low serum vitamin D levels are significantly associated with an increased risk of sarcopenia. While there is inconsistency in the findings, most studies support the importance of vitamin D in maintaining skeletal muscle health. Vitamin D influences the onset and progression of sarcopenia through various pathways, including the promotion of muscle protein synthesis, the regulation of mitochondrial function, and the modulation of immune and inflammatory responses. Regarding the prevention and treatment of sarcopenia, a combination of nutritional, exercise, and pharmacological interventions is recommended. Further research should be conducted to elucidate the molecular mechanism of vitamin D in sarcopenia, to study genes related to sarcopenia, to perform large-scale clinical trials, to investigate special populations, and to examine the combined application of vitamin D with other nutrients or drugs. A comprehensive investigation of the interconnection between vitamin D and sarcopenia will furnish a novel scientific foundation and productive strategies for preventing and treating sarcopenia. This, in turn, will enhance the senior people’s quality of life and health.
Article
Full-text available
Objective To compare nutritional value and aspects with environmental impact of high-protein (HP) and ‘normal-protein’ (NP) ultra-processed foods (UPF). Design 299 HP and 286 NP products were evaluated regarding aspects of nutritional value, energy density, Nutri-Score, number of additives as well as hyper-palatability and price. Environmental impact of HP UPF was addressed by analysing protein sources and the use of environmentally persistent non-nutritive artificial sweeteners. Setting Cross-sectional market analysis in German supermarkets and online shops. Participants 299 HP and 286 NP UPF products. Results HP compared to NP UPF had a lower energy density, a lower content of sugar, total and saturated fat, whereas fibre and protein content (62·2 % animal protein) were higher (all P < 0·001). HP products therefore had a higher prevalence of Nutri-Score A (67·2 % v . 21·7 %) and a lower prevalence of Nutri-Score E (0·3 % v . 11·2 %) labelling (both P < 0·001). By contrast, salt content and the number of additives (environmentally persistent sweeteners, sugar alcohols, flavourings) were higher in HP compared to NP UPF ( P < 0·001). When compared to HP products, twice as many NP were identified as hyper-palatable (82·5 % v . 40·5 %; P < 0·001). The price of HP was on average 132 % higher compared to NP UPF ( P < 0·001). Conclusions While major adverse aspects of UPF regarding nutritional profile and hyper-palatability are less pronounced in HP compared to NP products, higher salt content, increased number of additives and negative environmental effects from frequent use of animal protein and environmentally persistent sweeteners are major drawbacks of HP UPF.
Article
Full-text available
The emergence of immunotherapy, particularly immune checkpoint inhibitors (ICIs), represents a groundbreaking approach to treating gastric cancer (GC). However, the prognosis of GC patients receiving ICI treatment is influenced by various factors. This manuscript identified sarcopenia and myosteatosis as inde-pendent prognostic factors impacting the outcomes of GC patients treated with ICIs. Additionally, this study introduced a visual predictive model to estimate the prognosis of GC patients. If confirmed by further studies, this observation could provide valuable insights to propel the advancement of personalized clinical medicine and the integration of precision medicine practices.
Article
Full-text available
肌肉减少症是指随着年龄的增长,肌肉质量和力量(或两者)以及生理功能的丧失。及早发现肌肉无力可以更好地护理和干预老年人的饮食习惯和蛋白质摄入量。本研究的目的是调查巴基斯坦人群中肌肉减少症的患病率,并将饮食习惯和生活方式与肌肉减少症的患病率联系起来。使用的样本量为150名60岁及以上的男性和女性。心血管疾病和肾功能衰竭患者被排除在外。研究领域是拉合尔社区。使用握力测力计计算肌肉力量,并使用计步器计算步态速度。筛查后,使用生物电阻抗分析(bioelectrical impedance analysis,BIA)计算肌肉质量,由此诊断肌肉减少症。在60–65岁年龄组中,123人(82%)的肌肉力量较低,93人(83%)的肌肉质量较低。在66–70岁年龄组中,15人(83.3%)肌力较低。在71–75岁年龄组中,9人(90%)肌力较低。76岁以上年龄组的低肌力百分比为100%。60岁及以上人群中,重度肌少症的比例为6%,中度肌少症的比例约为10%。男性肌少症患病率为21.53%,女性肌少症患病率为11.76%。肌肉减少症是老年人中一个新出现的健康问题,早期发现和生活方式改变将带来更好的健康结果,并将饮食习惯和生活方式与肌肉减少症的患病率联系起来。
Article
Full-text available
Background: Resistance exercise leads to net muscle protein accretion through a synergistic interaction of exercise and feeding. Proteins from different sources may differ in their ability to support muscle protein accretion because of different patterns of postprandial hyperaminoacidemia. Objective: We examined the effect of consuming isonitrogenous, isoenergetic, and macronutrient-matched soy or milk beverages (18 g protein, 750 kJ) on protein kinetics and net muscle protein balance after resistance exercise in healthy young men. Our hypothesis was that soy ingestion would result in larger but transient hyperaminoacidemia compared with milk and that milk would promote a greater net balance because of lower but prolonged hyperaminoacidemia. Design: Arterial-venous amino acid balance and muscle fractional synthesis rates were measured in young men who consumed fluid milk or a soy-protein beverage in a crossover design after a bout of resistance exercise. Results: Ingestion of both soy and milk resulted in a positive net protein balance. Analysis of area under the net balance curves indicated an overall greater net balance after milk ingestion (P < 0.05). The fractional synthesis rate in muscle was also greater after milk consumption (0.10 ± 0.01%/h) than after soy consumption (0.07 ± 0.01%/h; P = 0.05). Conclusions: Milk-based proteins promote muscle protein accretion to a greater extent than do soy-based proteins when consumed after resistance exercise. The consumption of either milk or soy protein with resistance training promotes muscle mass maintenance and gains, but chronic consumption of milk proteins after resistance exercise likely supports a more rapid lean mass accrual.
Article
Full-text available
Abstract Background Protein intake is essential to maximally stimulate muscle protein synthesis, and the amino acid leucine seems to possess a superior effect on muscle protein synthesis compared to other amino acids. Native whey has higher leucine content and thus a potentially greater anabolic effect on muscle than regular whey (WPC-80). This study compared the acute anabolic effects of ingesting 2 × 20 g of native whey protein, WPC-80 or milk protein after a resistance exercise session. Methods A total of 24 young resistance trained men and women took part in this double blind, randomized, partial crossover, controlled study. Participants received either WPC-80 and native whey (n = 10), in a crossover design, or milk (n = 12). Supplements were ingested immediately (20 g) and two hours after (20 g) a bout of heavy-load lower body resistance exercise. Blood samples and muscle biopsies were collected to measure plasma concentrations of amino acids by gas-chromatography mass spectrometry, muscle phosphorylation of p70S6K, 4E–BP1 and eEF-2 by immunoblotting, and mixed muscle protein synthesis by use of [2H5]phenylalanine-infusion, gas-chromatography mass spectrometry and isotope-ratio mass spectrometry. Being the main comparison, differences between native whey and WPC-80 were analysed by a one-way ANOVA and comparisons between the whey supplements and milk were analysed by a two-way ANOVA. Results Native whey increased blood leucine concentrations more than WPC-80 and milk (P
Article
Full-text available
Sarcopenia is a major public health issue. To convince health policy makers of the emergency to invest in the sarcopenia field, it is of critical importance to produce reliable figures of the expected burden of sarcopenia in the coming years. Age- and gender-specific population projections were retrieved until 2045 from the Eurostat online database (28 European countries). Age- and gender-specific prevalences of sarcopenia were interpolated from a study that compared prevalence estimates according to the different diagnostic cutoffs of the EWGSOP proposed definition. The reported prevalence estimates were interpolated between 65 and 100 years. Interpolated age- and gender-specific estimates of sarcopenia prevalence were then applied to population projections until 2045. Using the definition providing the lowest prevalence estimates, the number of individuals with sarcopenia would rise in Europe from 10,869,527 in 2016 to 18,735,173 in 2045 (a 72.4% increase). This corresponds to an overall prevalence of sarcopenia in the elderly rising from 11.1% in 2016 to 12.9% in 2045. With the definition providing the highest prevalence estimates, the number of individuals with sarcopenia would rise from 19,740,527 in 2016 to 32,338,990 in 2045 (a 63.8% increase), corresponding to overall prevalence rates in the elderly of 20.2% and 22.3% for 2016 and 2045, respectively. We showed that the number of sarcopenic patients will dramatically increase in the next 30 years, making consequences of muscle wasting a major public health issue.
Article
Full-text available
Background: Interventions to attenuate the adverse effects of age-related loss of skeletal muscle and function include increased physical activity and nutritional supplementation. Objective: This study tested the hypothesis that nutritional supplementation with whey protein (22 g), essential amino acids (10.9 g, including 4 g leucine), and vitamin D [2.5 μg (100 IU)] concurrent with regular, controlled physical activity would increase fat-free mass, strength, physical function, and quality of life, and reduce the risk of malnutrition in sarcopenic elderly persons. Design: A total of 130 sarcopenic elderly people (53 men and 77 women; mean age: 80.3 y) participated in a 12-wk randomized, double-blind, placebo-controlled supplementation trial. All participants concurrently took part in a controlled physical activity program. We examined body composition with dual-energy X-ray absorptiometry, muscle strength with a handgrip dynamometer, and blood biochemical indexes of nutritional and health status, and evaluated global nutritional status, physical function, and quality of life before and after the 12 wk of intervention. Results: Compared with physical activity and placebo, supplementation plus physical activity increased fat-free mass (1.7-kg gain, P < 0.001), relative skeletal muscle mass (P = 0.009), android distribution of fat (P = 0.021), handgrip strength (P = 0.001), standardized summary scores for physical components (P = 0.030), activities of daily living (P = 0.001), mini nutritional assessment (P = 0.003), and insulin-like growth factor I (P = 0.002), and lowered C-reactive protein (P = 0.038). Conclusion: Supplementation with whey protein, essential amino acids, and vitamin D, in conjunction with age-appropriate exercise, not only boosts fat-free mass and strength but also enhances other aspects that contribute to well-being in sarcopenic elderly. This trial was registered at clinicaltrials.gov as NCT02402608.
Article
Full-text available
Sarcopenia, an age-related decline in muscle mass, is a burgeoning public health concern in the UK, with the number of people over the age of 65 expected to double by 2050. Resistance exercise is an effective intervention in its prevention and management. Increasing quantity and improving quality of dietary protein, by inclusion of high-availability leucine, are also purportedly beneficial. Leucine is a key anabolic amino acid, found in dairy foods. A number of studies have investigated dairy foods in prevention of sarcopenia. This paper reviews interventions of exercise, amino acids including leucine, dairy protein and foods for prevention of sarcopenia.
Article
Full-text available
Systematic reviews should build on a protocol that describes the rationale, hypothesis, and planned methods of the review; few reviews report whether a protocol exists. Detailed, well-described protocols can facilitate the understanding and appraisal of the review methods, as well as the detection of modifications to methods and selective reporting in completed reviews. We describe the development of a reporting guideline, the Preferred Reporting Items for Systematic reviews and Meta-Analyses for Protocols 2015 (PRISMA-P 2015). PRISMA-P consists of a 17-item checklist intended to facilitate the preparation and reporting of a robust protocol for the systematic review. Funders and those commissioning reviews might consider mandating the use of the checklist to facilitate the submission of relevant protocol information in funding applications. Similarly, peer reviewers and editors can use the guidance to gauge the completeness and transparency of a systematic review protocol submitted for publication in a journal or other medium.
Article
This presentation reflects on the origins of the term sarcopenia. The Greek roots of the word are sarx for flesh and penia for loss. The term actually describes important changes in body composition and related functions. Clearly defining sarcopenia will allow investigators to appropriately classify patients and examine underlying pathogenic mechanisms and will allow funding agencies to appropriately target research funds to a taxonomically distinct syndrome.
Article
Background Older adults experience age-related physiological changes that affect body weight and body composition. In general, nutrition and exercise have been identified as potent stimulators of protein synthesis in skeletal muscle. Milk proteins are excellent sources of all the essential amino acids and may represent an ideal protein source to promote muscle anabolism in older adults undergoing resistance training. However, several randomized control trials (RCTs) have yielded mixed results on the effects of milk proteins supplementation in combination with resistance training on body weight and composition. Methods PubMed, Web of Science and Cochrane databases were searched for literature that evaluated the effects of milk proteins supplementation on body weight and composition among older adults (age ≥ 60 years) undergoing resistance training up to September 2016. A random-effects model was used to calculate the pooled estimates and 95% confidence intervals (CIs) of effect sizes. ResultsThe final analysis included 10 RCTs involving 574 participants (mean age range from 60 to 80.8 years). Overall, the combination of milk proteins supplementation and resistance training did not have significant effect on fat mass (0.30, 95% CI -0.25, 0.86 kg) or body weight (1.02, 95% CI: -0.01, 2.04 kg). However, a positive effect of milk proteins supplementation paired with resistance training on fat-free mass was observed (0.74, 95% CI 0.30, 1.17 kg). Greater fat-free mass gains were observed in studies that included more than 55 participants (0.73, 95% CI 0.30, 1.16 kg), and in studies that enrolled participants with aging-related medical conditions (1.60, 95% CI 0.92, 2.28 kg). There was no statistical evidence of publication bias among the studies. Conclusion Our findings provide evidence that supplementation of milk protein, in combination with resistance training, is effective to elicit fat-free mass gain in older adults.