Anda di halaman 1dari 10

Experimental Gerontology 115 (2019) 104–113

Contents lists available at ScienceDirect

Experimental Gerontology
journal homepage: www.elsevier.com/locate/expgero

The effects of a combined bodyweight-based and elastic bands resistance T


training, with or without protein supplementation, on muscle mass,
signaling and heat shock response in healthy older people

Mauricio Krausea,b,c, , Domenico Crognalea,c, Karl Cogana, Serena Contarellie, Brendan Egana,d,
⁎⁎
Philip Newsholmef, Giuseppe De Vitoa,c,
a
Institute for Sport & Health, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Ireland
b
Laboratory of Inflammation, Metabolism and Exercise Research (LAPIMEX) and Laboratory of Cellular Physiology, Department of Physiology, Institute of Basic Health
Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
c
Food for Health Ireland, University College Dublin, Ireland
d
School of Health & Human Performance, Dublin City University, Ireland
e
Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
f
CHIRI Biosciences Research Precinct, School of Biomedical Sciences, Curtin University, Australia

A R T I C LE I N FO A B S T R A C T

Section Editor: Christiaan Leeuwenburgh This investigation sought to determine the effects of twelve weeks of resistance exercise training in addition to
Keywords: protein supplementation on body composition, markers of muscle atrophy/hypertrophy and heat shock response
Resistance exercise training (HSR) in healthy older adults. Thirty-eight healthy sedentary participants (M/F, 18/20; age, 63.5 ± 4.4 y) were
Protein supplementation randomly assigned to four groups: I) PLACEBO: no training, receiving placebo sachets; II) NUTRITION: no
Muscle growth and function training, receiving protein supplementation sachets; III) EXERCISE PLACEBO: training, placebo sachets and IV)
Heat shock response EXERCISE NUTRITION: training, receiving protein sachets. The resistance training (using bodyweight and elastic
bands) consisted of 45 min supervised training sessions, 3×/week. Participants from both exercise groups in-
creased their total lean body mass (from 48.4 ± 8.7 to 49.2 ± 8.7 kg and from 44.9 ± 7.8 to 45.9 ± 8.1 kg,
average of gain ~0.8 and 1 kg, placebo and nutrition respectively) and improved results in physical tests.
Exercise nutrition group also reduced their body fat (from 34.8 ± 7.3 to 32.9 ± 7.4%), increased the expression
of proteins/gene involved on the HSR, S6 and eEF2, while FOXO3 and Murf1 were reduced. Expression of MHC-I
was reduced in both exercise groups while MHC-IIa increased, with no effect of protein supplementation alone.
Body-weight and elastic bands based resistance exercise prompted, in healthy older people, improvements in
body composition and muscle function. When protein supplementation was added to the people engaged in
resistance training, improvements in fat mass and changes in skeletal muscle signaling were detected, favoring
protein synthesis pathways and the protective heat shock response.

1. Introduction These age-related losses, affecting physical function, can be further


exacerbated by reduced physical activity levels and a suboptimal nu-
According to the most recent projections in 2030 the number of tritional profile (Naseeb and Volpe, 2017).
older citizens (80+ years) will represent up to 30% of the industrialised In this context, to preserve muscle function is crucial in aging for
countries population and 12% of the under development ones maintaining mobility so reducing the economic and social cost asso-
(Knickman and Snell, 2002). Aging unfortunately, is associated with a ciated with possible loss of independence. It is now unanimously re-
generalized deterioration of physiological function including a pro- cognized that the practice of regular physical exercise is an effective
gressive decline in skeletal muscle mass and muscular strength and non-pharmacological therapeutic and preventive intervention delaying
power which progressively translate into functional impairment, in- or attenuating the negative effects that aging operates on functional and
creased rate of disability and dependency (Naseeb and Volpe, 2017). metabolic parameters (Leite et al., 2015).


Correspondence to: M. Krause, Department of Physiology, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil.
⁎⁎
Correspondence to: G. De Vito, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Belfield, Dublin 4, Ireland.
E-mail addresses: mauricio.krause@ufrgs.br (M. Krause), giuseppe.devito@ucd.ie (G. De Vito).

https://doi.org/10.1016/j.exger.2018.12.004
Received 4 September 2018; Received in revised form 28 November 2018; Accepted 4 December 2018
Available online 05 December 2018
0531-5565/ © 2018 Elsevier Inc. All rights reserved.
M. Krause et al. Experimental Gerontology 115 (2019) 104–113

Table 1
General group characteristics.
PLACEBO NUTRITION EXERCISE + PLACEBO EXERCISE + NUTRITION
(5♂/5♀) (4♂/3♀) (5♂/5♀) (4♂/7♀)

Age (y) 63.8 ± 4.5 62.3 ± 5.3 63.9 ± 3.9 63.9 ± 4.3
Body mass (kg) 73.3 ± 11.5 78.4 ± 13.4 73.6 ± 12.5 71.5 ± 6.1
Height (m) 1.66 ± 0.09 1.69 ± 0.1 1.71 ± 0.07 1.68 ± 0.08
BMI (kg·m−2) 26.3 ± 2.5 27.2 ± 3.3 25 ± 2.8 25.4 ± 2.3

Resistance exercise (RE), represents a well-established counter- (Greig et al., 1994). Participants were excluded if they reported a his-
measure to lessen the decline in lean body mass (LBM) and muscle tory of myocardial infarction, cardiac illness, vascular disease, un-
function that occurs with aging (Cruz-Jentoft et al., 2010; Krause et al., controlled metabolic disease, stroke, sarcopenia [according to the
2015a). However, stable-isotope-based methodologies have demon- European Working Group on Sarcopenia in Older People (EWGSOP)
strated that muscle protein synthesis (MPS) in older adults, in response criteria (Cruz-Jentoft et al., 2010)], hormonal replacement, major sys-
to exercise or protein supplementation, appear to be reduced or de- temic disease or any condition that would prevent them from engaging
layed, indicating anabolic resistance (AR) (Burd et al., 2013; in an exercise study; or if they were already engaging in two or more
Drummond et al., 2008). Despite the presence of AR, present evidence planned and structured exercise sessions per week. All subjects were
suggests that protein supplementation may be able to overcome these reported as non-diabetic after interview with an experienced physician
issues, particularly when combined with RE programmes (Murton, who considered their fasting glycaemia as the main criteria for diabetes
2015). exclusion.
Muscle protein synthesis is stimulated by RE and the time course
and the overall MPS response to this form of exercise have been ex- 2.2. Study design
tensively investigated (Burd et al., 2013), but the effects of RE training,
on markers of muscle atrophy and heat shock response (HSR) in older This study was a randomized, single blinded, controlled trial of
adults is at present under reported. The HSR involves the expression of bodyweight-based and elastic bands resistance exercise training, com-
heat shock proteins (particularly HSP70) in response to non-lethal bined with protein supplementation, performed for 12 weeks (for de-
stress to the cells (Krause et al., 2015b). The 72 kDa member of the tails please consult flow chart of patients at the Supplemental material).
70 kDa family of heat shock proteins, HSP70 (encoded by the HSPA1A Participants were randomly assigned to four groups (Table 1): I) PLA-
gene in humans) is the most abundant of all HSPs accounting for 1–2% CEBO: no exercise training, receiving placebo sachets; II) NUTRITION:
of cellular protein, and is plentiful in skeletal muscle (Noble et al., no exercise training, receiving protein supplementation sachets; III)
2008). As molecular chaperones, the intracellular HSP70 proteins can EXERCISE PLACEBO: exercise training, receiving placebo sachets and
interact with other proteins (unfolded, in non-native state and/or stress- IV) EXERCISE NUTRITION: exercise training, receiving protein sup-
denatured conformations) to avoid inappropriate interactions, forma- plementation sachets. After completion of baseline assessments, the
tion of protein aggregates and degradation of damaged proteins, as well randomization procedure was stratified by gender, based on equal
as helping the correct refolding of nascent proteins (Krause et al., distribution of males and females, but randomized to respective inter-
2015b). Other functions include protein translocation (Chirico et al., vention groups thereafter, without blocking. The control group was
1988), anti-apoptosis (Garrido et al., 2001) and anti-inflammatory re- instructed to maintain their usual physical activity and dietary habits
sponses (Krause et al., 2015c), control of cell signaling (Calderwood for the duration of the study. The exercise training intervention con-
et al., 2007), modulation of immune response (Johnson and Fleshner, sisted of 45 min supervised group training sessions performed three
2006), and modulation of chronic disease such as diabetes, obesity and times per week for 12 weeks, with each session consisting of body-
insulin resistance (Chung et al., 2008). weight-based resistance exercise and elastic band exercises. Assess-
Recently, evidences for a crosstalk between skeletal muscle protein ments of body composition and physical function took place at baseline
synthesis, degradation and HSR signaling has been described (Senf, (PRE) approximately one week before commencing the intervention,
2013; Wiggs, 2016). However, HSR may be blunted in older in- and after 12 weeks of intervention (POST). This study was part of a
dividuals, but can be potentially restored by RE (de Lemos Muller et al., previous joint project between two research institutions were the pri-
2018). Thus, the aim of the present study was to investigate the effects mary outcome variable was change in lean body mass (Francis et al.,
of twelve weeks of bodyweight and elastic bands based resistance ex- 2016). The estimated sample size was calculated for a larger study,
ercise training, combined or not with protein supplementation, on body using the data from Tieland et al. (2012), based on an increase in LBM
composition, physical function, markers of muscle atrophy/hyper- of 1.1 ± 1.4 kg (mean ± SD). A minimum of 20 participants per
trophy and HSR in healthy older adults. group was required to detect a difference (α = 0.05 and β = 0.8). In the
present study, however, we analyzed a different sub-group of people
2. Materials and methods (no additional sample size calculation was performed; n = 38, ~10 per
group;) where skeletal muscle biopsy samples were obtained to eval-
2.1. Participants uate markers of gene/protein expression at PRE and POST, looking at
genes and proteins involved on protein synthesis/degradation, meta-
A total of 38 sedentary non-smoking participants (M/F, 18/20; age, bolism, inflammation and heat shock response.
63.5 ± 4.4 y; body mass, 74.3 ± 10.9 kg; BMI, 25.5 ± 2.7 kg·m−2)
volunteered for the present study. Participants were recruited using a 2.3. Bodyweight-based and elastic bands resistance training intervention
wide variety of techniques, including newspaper advertisements, pos-
ters, distribution of information flyers, and by word of mouth. Written Supervised (by qualified sport and exercise scientists) exercise was
informed consent was obtained from all participants prior to com- performed in a gym on three non-consecutive days of the week. Each
mencing the study after written and verbal explanation of the study session lasted 45–60 min and consisted of a warm up, the bodyweight-
design approved by the University College Dublin Research Ethics based resistance exercise training and a cool down. Compliance to the
Committee (RCT number LS-11-212-DeVito). The medical history of the exercise program was monitored using a weekly exercise log which was
participants was evaluated, and all were defined as medically-stable checked by the trainers administering the exercise session. The first

105
M. Krause et al. Experimental Gerontology 115 (2019) 104–113

3 weeks of the program had an emphasis on ensuring the correct ex- repetition chair rise test and by counting the number of chair rises
ercise technique and monitoring, for everyone, the appropriate amount completed in 30 s (Forte et al., 2013). Lower body flexibility was as-
of exercise and rest intervals. Between weeks 3 and 12, the program sessed using the chair version of the sit & reach test (Forte et al., 2013).
was designed to promote muscle hypertrophy (4–6 sets, 8–15 repeti- Functional ability assessment included walking speed measurement
tions) as recommended by Bird et al. (Bird et al., 2005). The training performed on a 10 m indoor course using measuring gates (Smartspeed,
consisted of a combination of upper and lower body exercises using Fusion Sport, Coopers Plains, Australia). Participants were asked to
thera-bands (Therabands resistance loop, USA) and body weight as the walk “as fast as possible without running” for maximal walking speeds
primary resistance. The primary exercises used throughout the program assessment. Isometric hand grip strength of both hands was measured
included squats, lunges, hip abduction, shoulder press, latissimus dorsi after adjustment for hand size, with the participant standing and re-
pull-down, bicep curls, calf raises, push-ups, triceps dips and lumbo- laxing their arm along their body (Baseline Hydraulic Hand Dynam-
pelvic stabilisation exercises. Thera-band exercises were progressed by ometer Fabrication Enterprise Inc. Irvington NY). It is advisable to
increasing the resistance of the band in ascending order: red, green, measure hand grip in both arms, since significant differences between
blue and black respectively. Progression was determined based on the the dominant and the non-dominant side have been observed for
participants post exercise rate of perceived exertion (RPE) in con- handgrip strength, in both sexes (Ditroilo et al., 2010). Two trials were
sultation with the exercise trainer. Participants received at least 30 s performed with 1 min rest in between and the highest score used for
rest between sets. The training program was set out in 4 × 3 week statistical analysis. All measurements were carried out by the same
progressive cycles (Cycle 1: 3 sets of 8–12 reps; Cycle 2: 3 sets of 14 exercise scientist to exclude issues with inter-tester reliability.
reps; Cycle 3: 4 sets of 10–12 reps; Cycle 4: 4 sets of 12–14 reps). A full
description of the training program can be found elsewhere (Francis 2.7. Blood pressure and biochemistry
et al., 2016).
Systemic arterial blood pressure (BP) was measured in the brachial
2.4. Protein supplementation and placebo artery using an Omron M5-1 fully automated BP monitor (HEM 907,
Omron Healthcare, Kyoto, Japan). The mean of two measurements
Participants were instructed to take a supplement at their two lower taken at 2 minute intervals of supine rest was recorded. Venous blood
protein containing meals of the day (typically breakfast and lunch). samples were taken after fasting from an antecubital vein in heparin
Sachets were provided in powder format and could be mixed with water coated and gel-clot vacutainerTM tubes using standard aseptic techni-
to make-up a powdered beverage. Supplements were prescribed relative ques. Samples were immediately centrifuged (at 4 °C and 1000 g for
to the median 4 levels of participants' body mass (BM) (i.e. 45–59.9 kg, 15 min), after which plasma and serum was removed and stored at
median of 52.5 kg; 60–74.9 kg, median of 67.5 kg; 75–89.9 kg, median −80 °C for further analysis.
of 82.5 kg and 90–105 kg, median of 97.5 kg). Each protein supplement
dose provided 0.165 g protein kg−1 BM day−1 of median BM. Five fla- 2.8. Plasma IGF-1 and IGFBP3 quantification
vors were available to off-set flavor fatigue and to improve compliance.
All used or unused sachets were returned by the subjects for intake A highly sensitive, enzyme-linked immunosorbent assay method
control. Subjects were blinded to the supplement composition assigned was used to determine the levels of IGF-1 and IGFBP3 (Quantikine
to them. Compliance was monitored by count-back at the end of each ELISA Kit, R&D Systems, USA) in plasma samples. Absorbance was
month. Placebo consisted of an isoenergetic, non-nitrogenous mal- measured at 450 nm, and a standard curve constructed from known
todextrin sachet that was ingested at breakfast and midday meals. dilutions of the proteins. Quantification was made using a microplate
The milk protein matrix was composed of a 9:2:1 ratio of milk reader (Molecular Devices SpectraMax Plus 384, Sunnyvale, California,
protein concentrate [80% (wt:wt protein); Glanbia Ingredients Ireland, United States). The intra- and inter-assay CV, respectively, ranged be-
Kilkenny Ireland]; whey protein concentrate hydrolysate, degree of tween 4.3 and 5% for IGF-1 and 3.2 and 7% for IGFBP3.
hydrolysis 32% [WPC DH 32. 78% (wt:wt protein); Carbery Food
Ingredients, Cork, Ireland; and whey protein isolate hydrolysate, degree 2.9. Muscle biopsy
of hydrolysis 45% [WPC DH 45, 75% (wt:wt protein); Glanbia
Ingredients Ireland]. The total protein concentration was 72.7 g/100 g Skeletal muscle biopsies were obtained from the vastus lateralis
powder. The protein matrix was supplemented with 2187 mg/100 g muscle before (PRE) and after (POST) the 12-week intervention. The
powder of milk-based calcium (Trucal; Glanbia Ingredients Ireland) and POST biopsy was taken between 48 and 72 h after the final training
57.3 mg/100 g powder cholecalciferol. A full breakdown of the sup- session. Muscle biopsies were obtained from separate sites (2 cm apart)
plement composition has recently been published (please consult from the lateral portion of the muscle. Biopsies were obtained using a
(Norton et al., 2016) and Supplementary Table 1). semi-automatic spring-loaded biopsy system (MEDAX/BF14100-C0;
Bio-Feather with coaxial 14 g 10 cm). Samples were obtained (about
2.5. Anthropometry and body composition 30–50 mg) under local anesthesia (0.2% lidocaine) and immediately
frozen in liquid nitrogen and stored at −80 °C for later analysis.
Standing height was measured using a Stainless Steel Harpenden
Stadiometer (Holtain Ltd, Pembrokeshire, United Kingdom), with the 2.10. Protein quantification and Western blot
participants' shoes off and head at the Frankfort horizontal plane. Body
mass was measured using a Seca 888 weighing scale, Birmingham, Briefly, the tissues were homogenized (still frozen) in 0.1% (w/v)
United Kingdom. Body composition was assessed in the overnight SDS buffer containing protease and phosphatase inhibitor cocktail
fasted state by dual energy X-ray absorptiometry (DEXA) (Lunar iDXA, (Sigma). Afterwards, the homogenates were centrifuged at 15000 ×g
GE Healthcare, Buckinghamshire, United Kingdom). DXA quality as- for 5 min at room temperature and the supernatant fractions were saved
surance measurements and calibrations were performed daily to ensure for protein determination by using bovine serum albumin (BSA, Sigma)
scanner reliability, and identical patient scan protocols were performed as standard. Then, equivalent amounts of protein from each sample (~
for all participants. 40 μg) were mixed with Laemmli's gel loading buffer in a ratio of 1:1,
boiled for 5 min and electrophoresed. The samples were separated by
2.6. Assessments of physical function SDS-PAGE (4–15% MiniPROTEAN TGX™ Precast Protein Gels, Bio-Rad)
and transferred onto nitrocellulose membranes (GE Healthcare Life
The ability to rise from a chair was assessed using a timed 5 Sciences “Amersham Protran 0.2 NC”). Non-specific binding was

106
M. Krause et al. Experimental Gerontology 115 (2019) 104–113

blocked in a 7.5% milk/2.5% BSA/TBS-t (10 mM Tris pH 7.5, 100 mM reduced whereas the number of chair rises completed in 30 s increased
NaCl, 0.4% Tween20) for 1 h at room temperature. Membranes were (Cohen's D effect size: 0.58 and 0.32, respectively, Fig. 1D and E). No
incubated overnight with primary antibodies directed towards the fol- changes were found in the 10 m walk test (Fig. 1F), however, both
lowing primary antibodies: eEF2 (1:1000; Cell Signaling Technology, exercised groups increased their flexibility, based on the sit and reach
Beverly, MA; no. 2332), p70 S6 Kinase (1:1000; Cell Signaling; no. test results (Cohen's D effect size: 0.355 and 0.05, respectively, Fig. 1G).
9202), S6 Ribosomal Protein (5G10) (1:1000; Cell Signaling; no. 2217), As expected, considering that the training was not specific for upper
HSF1 (1:750; Cell Signaling; no. 4356), GAPDH (1:7500; Cell Signaling; body, no changes were detected for handgrip strength (Fig. 1C).
no. 2118), HSP70 (1:2000; Sigma-Aldrich (St. Louis,MO, USA; no.
H5147). Membranes were washed in TBS-t and incubated with appro- 3.3. General blood biochemistry
priate secondary HRP-conjugated antibodies (Bio-Rad), visualized by
ECL (Pierce™ ECL, Thermo Fisher Scientific) and quantified on ImageJ No changes were found for any of the considered parameters
software. GAPDH were used for normalization. (Table 2).

2.11. Quantitative polymerase chain reaction (qPCR) analysis of gene 3.4. Gene and protein expression
expression
Skeletal muscle samples were taken after 48–72 h to minimize any
Total RNA was isolated from approximately 20 mg of crude muscle residual effect of the last training session. After 12 weeks of interven-
using TRIzol reagent (Sigma-Aldrich, UK) as per the manufacturer's tion exercise nutrition group increased the levels of HSF-1 (p < 0.0001),
instructions. Total RNA concentration was quantified spectro- S6 (p < 0.05), HSP72 (p < 0.05) and eEF2 (p < 0.01). No changes
photometrically at an absorbance of 260 nm (NanoDrop ND-1000 on P70 levels were found (Fig. 2). FOXO3 (p < 0.05) and Murf1 were
Spectrophotometer, ThermoFisher Scientific, Waltham, MA, USA). The reduced only in exercise nutrition group and no changes on HSP72 or
integrity and purity of each RNA sample was verified by measuring the MAFbx mRNA levels were observed (Fig. 3A, B, C and D). Interestingly,
spectrophotometric A260/A280 (> 1.8) and A260/A230 (> 1.5) ra- levels of MHC-I reduced (p < 0.01 for exercise nutrition group and
tios. RNA (1 μg) was reverse transcribed to cDNA using the High p < 0.05 for exercise placebo group) and MHC-IIa increased in both
Capacity cDNA Reverse Transcription Kit (Applied Biosystems) as per exercise groups after the intervention (p = 0.019 for exercise nutrition
the manufacturer's instructions. The cDNA template was stored at room group and p < 0.01 for exercise placebo group) without any additional
temperature until subsequent analysis. Relative mRNA expression effect of protein supplementation (Fig. 3F, G and H). Exercise nutrition
(20 ng cDNA template per reaction) was determined using quantitative group also reduced MHC-IIx and MyoD expression after intervention
real-time PCR (ABI Prism 7900HT, Applied Biosystems, Foster City, CA, (p < 0.01). No significant differences were found for gene expression
USA) using Assay-On-Demand primer pairs and probes (P/N 4331182, of KLF15, PGC-1α, IL1-β, CCL2, IGF1 or MSTN (data not shown).
TaqmanH Gene Expression Assays, Applied Biosystems) and TaqmanH
Universal PCR MasterMix (Applied Biosystems). Assay IDs for specific 4. Discussion
mRNA targets are listed in Table S1. GAPDH mRNA expression
(4333764F, Applied Biosystems) was stable across time points during 4.1. Bodyweight-based resistance exercise training increases muscle mass
the training intervention and used as the housekeeping gene to which and function in healthy older people, independently of protein
target mRNA expression was normalized. supplementation

2.12. Statistical analysis Progressive resistance training (PRT) has been clearly shown ef-
fective for both strength and muscle mass enhancement in older in-
The GraphPad Prism (v6.0; IBM Corp.) statistics and graphical dividuals and has been widely used in this population. However, PRT
software package was used for all analyses. Descriptive data are re- may not represent the first choice of exercise for the older, especially
ported as mean ± SD. Two-way (time x group) repeated measures women, who may prefer other kind of activities (Cadore et al., 2014;
analysis of variance (ANOVA) was used to detect differences, with Rodrigues-Krause et al., 2016). In the present work, it has been shown
pairwise comparisons performed using Bonferroni post-hoc test when a that a bodyweight-based resistance exercise intervention [inexpensive
significant main or interaction effect was detected. For all analyses, and with a good adherence (compliance of the exercise groups ~85%)],
statistical significance was accepted at p < 0.05. can induce, in healthy older people, improvements in body composition
(mainly lean mass) and muscle function. When protein supplementa-
3. Results tion was added to the diet of the people engaged on the resistance
training, additional gains could be detected, including lower fat mass
3.1. Body composition and changes in skeletal muscle signaling (protein synthesis pathways
and heat shock response).
Table 1 displays baseline participants' characteristics. There were no These results are even more important because were obtained in
differences at baseline between groups in terms of age, BMI, body mass relatively young older individuals and with a proper information and
and height. After 12 weeks of intervention, participants from both ex- access to healthy food, especially in respect to the protein intake. So far,
ercise groups, protein supplementation or placebo (p = 0.001 and evidence from several studies shows no clear benefit of protein sup-
p = 0.0058; Cohen's D effect size: 0.26 and 0.08, respectively), increased plementation on skeletal muscle mass in older people, since some stu-
their total lean body mass in a similar manner while the 2 non-exercise dies found positive effects (Bonnefoy et al., 2003; Borsheim et al., 2008)
groups did not change (Fig. 1A). No differences were found for total and others did not (Candow and Chilibeck, 2008; Verdijk et al., 2009;
lean mass gain between the exercise groups (nutrition vs. placebo), Verhoeven et al., 2009). Despite that, long-term interventions have
however, only in the exercise nutrition group a significant reduction in % shown that protein supplementation improves physical performance,
of body fat was observed (p < 0.0001; Cohen's D effect size: 0.139) but does not necessarily increase skeletal muscle mass in frail older
(Fig. 1B). No changes were found in bone mineral density (Table 2). people (Tieland et al., 2012), indicating that protein supplementation
(along with exercise) may induce improvements in neuromuscular
3.2. Functional physical tests control and skeletal muscle only when combined with an appropriate
exercise training. Interestingly, in our twelve-week intervention, the
In both exercise groups the time taken to complete 5 chair rises was increased anabolic signaling, induced only in people engaged on

107
M. Krause et al. Experimental Gerontology 115 (2019) 104–113

Fig. 1. Changes in whole-body lean body mass and body fat mass assessed by DXA (A and B). Pre and post results of Tests of physical performance: Handgrip strength
(C), chair test (D and E), 10 meters walk test (F) and sit and reach test (G). Data are mean ± SD. Data for body composition are shown as box-and-whisker plots, and
error bars represent Tukey confidence intervals. *p < 0.05.

exercise and protein supplementation (Exercise Nutrition Group), was (Koopman and van Loon, 2009), the majority of studies have found that
not translated into differences in skeletal muscle mass or in physical the rates of muscle protein turnover are not abnormal in older in-
function. In fact, both exercise groups increased their muscle mass and dividuals under basal conditions (Markofski et al., 2015), indicating
improve their functional test results. However, these changes in protein that other factors that influence muscle protein turnover may be re-
signaling may be important, in long term, for exercise-induced adap- sponsible for muscle atrophy during aging. Physical training has been
tations (e.g. skeletal muscle mass and function maintenance, insulin shown to reduce the age-associated loss of muscle strength and the
sensitivity, inflammatory control, etc.) that will be translated in main- increased muscle fat infiltration in elderly people (Goodpaster et al.,
tenance of muscle size and function. 2008). Moreover, MPS can be stimulated by both resistance exercise
Regarding the functional tests results, it is important, however, to and nutrition in older people (Drummond et al., 2008). However, the
consider the data with caution, since the Exercise Placebo Group started MPS response to different exercise stimuli is reduced, indicating ana-
from a slightly lower functional capacity (considering the chair test bolic resistance (AR).
results). Whey proteins are a rich source of essential amino acids and rapidly
elevate plasma amino acids, thus providing the foundations for pre-
4.2. Protein supplementation increases skeletal muscle anabolic signaling servation of muscle mass. Previous studies involving supplementation
only when associated with exercise training with whey protein have been shown to be effective in augmenting the
effects of resistance exercise, particularly when supplementation occurs
Although early evidence suggests a reduced basal MPS rate (i.e. in the hours surrounding the exercise training (Hayes and Cribb, 2008).
following an overnight fast) in the older compared with young subjects Not only leucine, but other amino acids may be involved on the

108
M. Krause et al. Experimental Gerontology 115 (2019) 104–113

Table 2
Body composition and general biochemistry responses.
PLACEBO NUTRITION EXERCISE + PLACEBO EXERCISE + NUTRITION
(5♂/5♀) (4♂/3♀) (5♂/5♀) (4♂/7♀)

PRE POST PRE POST PRE POST PRE POST

Urea (mmol/L) 6.12 ± 0.9 4.78 ± 0.8 5.13 ± 0.64 5.39 ± 1.04 5.86 ± 1.87 4.93 ± 1.61 5.5 ± 1.24 5.82 ± 1.88
Creatinine (μmol/L) 71.70 ± 11.94 74.00 ± 12.35 67.00 ± 13.17 68.25 ± 11.99 75.20 ± 23.18 76.30 ± 21.47 74.09 ± 17.52 72.91 ± 8.85
Albumin (mmol/L) 39.40 ± 1.58 37.50 ± 1.65 35.39 ± 12.78 37.00 ± 1.69 36.12 ± 11.37 36.60 ± 2.12 38.45 ± 3.56 36.80 ± 2.25
Total Cholesterol (mmol/L) 5.11 ± 0.69 4.35 ± 0.45 4.88 ± 0.41 4.78 ± 0.81 5.03 ± 0.72 5.01 ± 0.87 5.03 ± 1.04 4.70 ± 0.96
Tryglicerides (mmol/L) 1.48 ± 1.13 1.08 ± 0.75 1.17 ± 0.63 1.47 ± 0.96 1.00 ± 0.28 1.05 ± 0.37 1.08 ± 0.62 1.07 ± 0.68
HDL (mmol/L) 1.24 ± 0.32 1.18 ± 0.29 1.48 ± 0.40 1.21 ± 0.44 1.27 ± 0.34 1.28 ± 0.41 1.56 ± 0.48 1.42 ± 0.46
LDL (mmol/L) 3.16 ± 0.60 2.32 ± 0.46 2.61 ± 0.70 2.53 ± 0.86 2.94 ± 0.50 2.83 ± 0.56 2.69 ± 0.92 2.37 ± 0.95
Calcium (mmol/L) 2.26 ± 0.09 2.10 ± 0.13 2.32 ± 0.07 2.04 ± 0.06 2.34 ± 0.08 2.08 ± 0.08 2.32 ± 0.15 2.10 ± 0.09
Vitamin D (nmol/L) 74.20 ± 21.16 61.70 ± 21.37 69.25 ± 23.42 63.50 ± 22.56 76.20 ± 17.55 61.40 ± 18.91 68.27 ± 25.02 80.27 ± 17.32
BMD (g/cm2) 1.1 ± 0.1 1.1 ± 0.1 1.1 ± 0.1 1.1 ± 0.1 1.1 ± 0.2 1.1 ± 0.2 1.1 ± 0.1 1.1 ± 0.1
Total Lean Body Mass (Kg) 46.7 ± 10.1 46.7 ± 10 48.4 ± 11.6 48.9 ± 11.5 48.4 ± 8.7 49.2 ± 8.7 44.9 ± 7.8 45.9 ± 8.1
Body Fat (%) 34.4 ± 6.3 33.9 ± 6.9 36.8 ± 13.17 35.6 ± 8.1 32.8 ± 4.9 31.9 ± 5 34.8 ± 7.3 32.9 ± 7.4
Glucose (mmol/L) 5.54 ± 0.43 5.26 ± 0.31 5.68 ± 0.63 5.4 ± 0.5 5.36 ± 0.35 5.06 ± 0.64 5.4 ± 0.37 5.05 ± 0.88
Insulin (mlU/L) 13.71 ± 8.8 11.09 ± 5.09 10.04 ± 3.28 12.4 ± 11.9 7.63 ± 5.35 10.58 ± 6.48 7.49 ± 4.12 10.41 ± 6.5
HOMA-IR (AU) 3.44 ± 2.45 2.61 ± 1.25 2.57 ± 1 3 ± 3.1 1.87 ± 1.4 2.42 ± 1.6 1.78 ± 0.95 2.41 ± 1.64
IGF-1 (ng/mL) 118.2 ± 28.9 161.2 ± 47.7 155.5 ± 92.5 144.1 ± 45.1 129.7 ± 38.3 145.8 ± 61.3 113.1 ± 44.5 139.1 ± 58.1
IGFBP3 (ng/mL) 71.2 ± 18.9 73.4 ± 21.3 66.7 ± 10.4 76.7 ± 10.7 65.7 ± 13.3 74.34 ± 15.2 72.5 ± 21.4 78.3 ± 3
IGF-1/IGFBP3 1.78 ± 0.6 2.39 ± 1.06 2.33 ± 1.33 1.91 ± 0.63 2.01 ± 0.54 1.98 ± 0.68 1.55 ± 0.3 1.82 ± 0.68

With the except of lean body mass and fat mass (* different from baseline, p < 0.05), no other differences were found.

anabolic effects of whey protein supplementation, such as arginine, 4.3. Resistance training along with protein supplementation increases heat
cysteine and glutamine. Although the plasma concentration of amino shock response signaling in skeletal muscle and reduces key catabolic factors
acids has not been measured in the present study, based on previous
studies (Farup et al., 2016), it is reasonable to expect that, these amino To the best of our knowledge, this is the first published work that
acids, especially leucine, arginine, cysteine and glutamine should be evaluates how exercise (resistance), alone or combined with protein
increased after the supplement ingestion (please consult Supplementary supplementation, modifies HSP70 expression and HS response in older
Table 1 which describes the contents of the supplement drink). In fact, individuals. Induction of HSP gene expression is regulated by the in-
at least when consumed after exercise (recovery period), whey protein teraction between the heat shock transcription factors (mainly HSF1)
and leucine supplementation, resulted in significant increments on and the regulatory heat shock elements (Krause et al., 2015a). Based on
plasma glycine, arginine, glutamine, and leucine (Nelson et al., 2013). its functions, it is predictable that HSP70 may protect against muscle
Our results indicate that protein supplementation can potentiate the atrophy through several different mechanisms (Wiggs, 2016); by its
effects of exercise on skeletal muscle signaling, particularly increasing ability to serve as a chaperone; by acting to maintain mitochondrial
anabolic and decreasing catabolic pathways. The additional effects of integrity; by prevent disuse atrophy induced-proteolysis (through in-
protein supplementation over this signaling may be explained by the hibiting of the activation of FOXO3 and NF-kB signaling pathways)
increased availability of specific amino acids, such as leucine, and di- (Senf, 2013; Senf et al., 2010); and finally, by maintaining the rate of
peptides, including glutamine dipeptides. In fact, whey protein hydro- translation elongation of nascent polypeptides (Ku et al., 1995; Nelson
lysate, is a rich source of these components. In the case of leucine, the et al., 1992). Thus, strategies to increase HSP70 and HSF-1 may induce
manner in which this amino acid elicits these anabolic effects within the beneficial effects for skeletal muscle development in the older in-
muscle cell appears through stimulation of the mammalian target of dividuals.
rapamycin complex 1 (mTORc1) signaling cascade (Murton, 2015). Glutamine (and its precursors, e.g. glutamate) availability, for ex-
This is achieved by mTORc1 inducing two main effects. Firstly, through ample, can improve cellular heat shock response (HS response - capacity
the activation of p70 ribosomal S6 kinase, and secondly, by inactivation to increase HSP70 in response to non-lethal stress) by enhancing HSF1
of the eukaryotic initiation factor 4E binding protein 1 (eIF4B), a pro- activation and expression in cultured cells and also in skeletal muscle of
tein that is known to repress protein translation. Other key downstream trained rats (Leite et al., 2016; Petry et al., 2014; Xue et al., 2012). This
effectors of mTORC1 signaling are the ribosomal protein S6 (S6) and can be done mainly by glutamine role on the activation of the hex-
eukaryotic elongation factor 2 (eEF2), where the activation of these osamine biosynthetic pathway (HBP), that will results in activation of
proteins will finally lead to protein synthesis. In the present interven- HSF-1, enhancing HSP70 expression (Krause et al., 2015b). Thus, the
tion, protein supplementation, to people engaged in the resistance increased availability of glutamine and/or glutamine dipeptides to
training, resulted in increased expression of S6 and eEF2, indicating a people under the exercise protocol, may have resulted in improvements
signaling favoring protein synthesis that may be stimulated by the in- of the HS response, explaining our findings with regard to HSF-1 and
creased leucine delivered to the skeletal muscle. HSP70 expression. In this regard, it is important to consider, that our
The anabolic signaling for adaptive hypertrophy induced by ex- population was composed by healthy older adults that ingest an average
ercise can involve the rise in blood anabolic hormones such as growth of 1.0–1.2 g protein/kg/day (Candow et al., 2006; Iglay et al., 2007;
hormone (GH) and insulin-like growth factor (IGF)-I, and consequent Verdijk et al., 2009), a protein intake that would result in sufficient
activation of a signaling cascade through phosphatidylinositol 3-kinases glutamine to induce a heat shock response. Moreover, recent evidences
(PI3K)-Akt-mTOR by IGF-I (Egan and Zierath, 2013). However, there have shown that increments in glutamine availability, combined with
were no changes on plasma levels of neither insulin, IGF-1 or IGFBP3, exercise (a mandatory stress), can induce further increments on HSP70
indicating that local modulators may be involved in the activation of expression through higher HBP flux (Leite et al., 2016).
the studied pathways, thus potentially involving IGF-I-independent In the present study, exercise without protein supplementation did
mechanisms of mTOR activation through mechano-sensory regulation not induce changes in muscle HSP70 content. Despite previous studies
(Philp et al., 2011). reporting that resistance training increases HSP70 in muscle, it is im-
portant to note that the majority of this previous research was

109
M. Krause et al. Experimental Gerontology 115 (2019) 104–113

Fig. 2. Changes in muscle protein expression by Western blot. A. Representative gels; Expression of B. HSF-1, C. HSP70, D. eEF2, E. P70 and F. S6. Data are
mean ± SD. *p < 0.05.

conducted on active healthy young people (Liu et al., 2000; Paulsen reduced after heat shock. In human studies, the heat shock-induced
et al., 2012). Some studies involving older people even showed de- HSP72 was blunted in older people in comparison with their younger
crease of HSP70 after resistance exercise in lymphocytes (Bautmans counterparts (Njemini et al., 2002; Njemini et al., 2004). Despite that, it
et al., 2005). In addition, in animal models and isolated fibroblasts and cannot be excluded that a longer exercise intervention may induce in-
lymphocytes from elderly human donors, it has been demonstrated that crements in HSP70 content even in the absence of any supplementation.
aging may induce declines in the HSR (as measured by their capacity to As previously discussed, HSP70 prevent disuse atrophy induced-
express iHSP72) (Blake et al., 1991; Deguchi et al., 1988; Njemini et al., proteolysis (through inhibiting of the activation of FOXO3 and NF-kB
2002; Njemini et al., 2004). In aged rats (Blake et al., 1991), for ex- signaling pathways) (Senf, 2013; Senf et al., 2010). Another point of
ample, the expression of HSP72 (in brain, lung and skin tissues), is crosstalk between HS response and protein synthesis may occur at the

110
M. Krause et al. Experimental Gerontology 115 (2019) 104–113

Fig. 3. Changes in transcript abundance for markers of muscle atrophy, regulators of growth, HSP70 and myosin heavy chain isoform. Relative mRNA expression was
determined using qPCR and normalized to the housekeeping gene β-actin. A, HSP70. B, FOXO3. C, MuRF1. D, MAFbx. E, MyoD. F, MHC-I. G, MHC-IIA. H, MHC-IIX.
Data are mean ± SD. *p < 0.05.

level of eIF2 protein (a downstream protein of the mTOR pathway). In FOXO3a-induced promoter activation of atrogin-1(MAFbx) (Senf et al.,
fact, eIF2 promotes the activation of HSF1 (Dokladny et al., 2015), 2010). In the present work, the group taking the supplementation and
leading to the expression of HSPs under stress conditions. It has been performing exercise, increased their HS response protein expression and
recently demonstrated, that HSP70 inhibits FOXO3-dependent tran- this co-occur with a higher expression of eIF2, lower gene expression of
scription in a gene-specific manner. Specifically, Hsp70 inhibited FOXO3 and lower MAFbx expression. As a result, protein

111
M. Krause et al. Experimental Gerontology 115 (2019) 104–113

supplementation, only in the trained group, seems to sensitize skeletal R663–R667.


muscle cells to respond to anabolic stimulus through increased HS re- Bonnefoy, M., Cornu, C., Normand, S., Boutitie, F., Bugnard, F., Rahmani, A., Lacour, J.R.,
Laville, M., 2003. The effects of exercise and protein-energy supplements on body
sponse and protein synthesis signaling. composition and muscle function in frail elderly individuals: a long-term controlled
randomised study. Br. J. Nutr. 89, 731–739.
4.4. Bodyweight-based resistance exercise training induce changes in Borsheim, E., Bui, Q.U., Tissier, S., Kobayashi, H., Ferrando, A.A., Wolfe, R.R., 2008.
Effect of amino acid supplementation on muscle mass, strength and physical function
skeletal muscle myosin heavy chain isoforms in elderly. Clin. Nutr. 27, 189–195.
Burd, N.A., Gorissen, S.H., van Loon, L.J., 2013. Anabolic resistance of muscle protein
Our intervention, in both groups of exercise, induced a change in synthesis with aging. Exerc. Sport Sci. Rev. 41, 169–173.
Cadore, E.L., Pinto, R.S., Bottaro, M., Izquierdo, M., 2014. Strength and endurance
the skeletal muscle Myosin heavy chain isoforms (MHCs expression). training prescription in healthy and frail elderly. Aging Dis. 5, 183–195.
MHC expression was decreased for type I (both placebo and protein Calderwood, S.K., Mambula, S.S., Gray Jr., P.J., Theriault, J.R., 2007. Extracellular heat
group) and type IIX (only in the protein group) and increased for type shock proteins in cell signaling. FEBS Lett. 581, 3689–3694.
Candow, D.G., Chilibeck, P.D., 2008. Timing of creatine or protein supplementation and
IIA muscle after the exercise intervention. Similar results were identi-
resistance training in the elderly. Appl. Physiol. Nutr. Metab. 33, 184–190.
fied previously, confirming that skeletal muscle plasticity, in response Candow, D.G., Chilibeck, P.D., Facci, M., Abeysekara, S., Zello, G.A., 2006. Protein sup-
to resistance training, is still present, even at older ages (Sharman et al., plementation before and after resistance training in older men. Eur. J. Appl. Physiol.
2001). It is commonly reported that in aging muscles the reduction in 97, 548–556.
Chirico, W.J., Waters, M.G., Blobel, G., 1988. 70K heat shock related proteins stimulate
individual fiber size is mainly confined to fast type II fibers, leading to a protein translocation into microsomes. Nature 332, 805–810.
progressive decrease in the type II-to-type I fiber area ratio, indicating a Chung, J., Nguyen, A.K., Henstridge, D.C., Holmes, A.G., Chan, M.H., Mesa, J.L.,
selective atrophy of type II fibers (Korhonen et al., 2006). Muscle Lancaster, G.I., Southgate, R.J., Bruce, C.R., Duffy, S.J., Horvath, I., Mestril, R., Watt,
M.J., Hooper, P.L., Kingwell, B.A., Vigh, L., Hevener, A., Febbraio, M.A., 2008.
wasting depends on the activation of two major pathways, the protea- HSP72 protects against obesity-induced insulin resistance. Proc. Natl. Acad. Sci. U. S.
somal and the autophagic-lysosomal systems, both controlled by A. 105, 1739–1744.
FOXO3. As discussed, other pathways, such as NF-κB, are also involved. Cruz-Jentoft, A.J., Baeyens, J.P., Bauer, J.M., Boirie, Y., Cederholm, T., Landi, F., Martin,
F.C., Michel, J.P., Rolland, Y., Schneider, S.M., Topinkova, E., Vandewoude, M.,
FOXO3 activity is downregulated by several factors such as Akt, HSP70 Zamboni, M., 2010. Sarcopenia: European consensus on definition and diagnosis:
and the transcriptional co-activator PGC-1α, where the last being more report of the European Working Group on Sarcopenia in Older People. Age Ageing
abundant in slow oxidative than in fast glycolytic muscle fibers. The 39, 412–423.
Deguchi, Y., Negoro, S., Kishimoto, S., 1988. Age-related changes of heat shock protein
fact that our resistance exercise training resulted in, perhaps (at least gene transcription in human peripheral blood mononuclear cells. Biochem. Biophys.
concerning the genes expression), a shift of muscle fiber type, from Res. Commun. 157, 580–584.
oxidative type I to more fast type IIA may represent a positive adap- Ditroilo, M., Forte, R., Benelli, P., Gambarara, D., De Vito, G., 2010. Effects of age and
limb dominance on upper and lower limb muscle function in healthy males and fe-
tation to preserve muscle function, muscle size and functional activity.
males aged 40–80 years. J. Sports Sci. 28, 667–677.
However, it is important to analyze the data with caution, since no Dokladny, K., Myers, O.B., Moseley, P.L., 2015. Heat shock response and autophagy–-
histological measurements were performed. cooperation and control. Autophagy 11, 200–213.
Drummond, M.J., Dreyer, H.C., Pennings, B., Fry, C.S., Dhanani, S., Dillon, E.L., Sheffield-
Moore, M., Volpi, E., Rasmussen, B.B., 2008. Skeletal muscle protein anabolic re-
5. Conclusions and perspectives sponse to resistance exercise and essential amino acids is delayed with aging. J Appl
Physiol (1985) 104, 1452–1461.
Herein we have shown that twelve weeks of an inexpensive body- Egan, B., Zierath, J.R., 2013. Exercise metabolism and the molecular regulation of ske-
letal muscle adaptation. Cell Metab. 17, 162–184.
weight and elastic bands based resistance exercise intervention, was Farup, J., Rahbek, S.K., Storm, A.C., Klitgaard, S., Jorgensen, H., Bibby, B.M., Serena, A.,
able to induce, in healthy older persons, improvements in body com- Vissing, K., 2016. Effect of degree of hydrolysis of whey protein on in vivo plasma
position and muscle function. When protein supplementation was amino acid appearance in humans. Springerplus 5 (382).
Forte, R., Costaleite, J., Ditroilo, M., Rodrigues-Krause, J., Brennan, L., Gibney, E., De
added to the diet of the people engaged in the resistance training, ad- Vito, G., Boreham, C.A., 2013. Functional and muscular gains in older adults: mul-
ditional improvements in fat mass (↓) and changes in skeletal muscle ticomponent vs. resistance exercise. J. Aging Res. Clin. Pract. 2, 242–248.
signaling, favoring protein synthesis pathways and the protective heat Francis, P., Mc Cormack, W., Toomey, C., Norton, C., Saunders, J., Kerin, E., Lyons, M.,
Jakeman, P., 2016. Twelve weeks' progressive resistance training combined with
shock response were detected.
protein supplementation beyond habitual intakes increases upper leg lean tissue
mass, muscle strength and extended gait speed in healthy older women.
Conflict of interest Biogerontology 18 (6), 881–891.
Garrido, C., Gurbuxani, S., Ravagnan, L., Kroemer, G., 2001. Heat shock proteins: en-
dogenous modulators of apoptotic cell death. Biochem. Biophys. Res. Commun. 286,
The authors declare that there is no conflict of interest that could be 433–442.
perceived as prejudicing the impartiality of the research reported. Goodpaster, B.H., Chomentowski, P., Ward, B.K., Rossi, A., Glynn, N.W., Delmonico, M.J.,
Kritchevsky, S.B., Pahor, M., Newman, A.B., 2008. Effects of physical activity on
strength and skeletal muscle fat infiltration in older adults: a randomized controlled
Funding trial. J. Appl. Physiol. 105, 1498–1503 (1985).
Greig, C.A., Young, A., Skelton, D.A., Pippet, E., Butler, F.M., Mahmud, S.M., 1994.
This work was supported by Food for Health Ireland and Enterprise Exercise studies with elderly volunteers. Age Ageing 23, 185–189.
Hayes, A., Cribb, P.J., 2008. Effect of whey protein isolate on strength, body composition
Ireland Grant CC/2008/001. and muscle hypertrophy during resistance training. Curr. Opin. Clin. Nutr. Metab.
Care 11, 40–44.
Appendix A. Supplementary data Iglay, H.B., Thyfault, J.P., Apolzan, J.W., Campbell, W.W., 2007. Resistance training and
dietary protein: effects on glucose tolerance and contents of skeletal muscle insulin
signaling proteins in older persons. Am. J. Clin. Nutr. 85, 1005–1013.
Supplementary data to this article can be found online at https:// Leite, Jaqueline Santos Moreira, Cruzat, Vinicius Fernandes, Krause, Mauricio, de
doi.org/10.1016/j.exger.2018.12.004. Bittencourt Jr., Paulo Ivo Homem, 2016. Physiological regulation of the heat shock
response by glutamine: implications for chronic low-grade inflammatory diseases in
age-related conditions. Nutrire 41, 1–12.
References Johnson, J.D., Fleshner, M., 2006. Releasing signals, secretory pathways, and immune
function of endogenous extracellular heat shock protein 72. J. Leukoc. Biol. 79,
425–434.
Bautmans, I., Njemini, R., Vasseur, S., Chabert, H., Moens, L., Demanet, C., Mets, T., 2005.
Knickman, J.R., Snell, E.K., 2002. The 2030 problem: caring for aging baby boomers.
Biochemical changes in response to intensive resistance exercise training in the el-
Health Serv. Res. 37, 849–884.
derly. Gerontology 51, 253–265.
Koopman, R., van Loon, L.J., 2009. Aging, exercise, and muscle protein metabolism. J.
Bird, S.P., Tarpenning, K.M., Marino, F.E., 2005. Designing resistance training pro-
Appl. Physiol. 106, 2040–2048 (1985).
grammes to enhance muscular fitness: a review of the acute programme variables.
Korhonen, M.T., Cristea, A., Alen, M., Hakkinen, K., Sipila, S., Mero, A., Viitasalo, J.T.,
Sports Med. 35, 841–851.
Larsson, L., Suominen, H., 2006. Aging, muscle fiber type, and contractile function in
Blake, M.J., Fargnoli, J., Gershon, D., Holbrook, N.J., 1991. Concomitant decline in heat-
sprint-trained athletes. J. Appl. Physiol. 101, 906–917 (1985).
induced hyperthermia and HSP70 mRNA expression in aged rats. Am. J. Phys. 260,
Krause, M., Bock, P.M., Takahashi, H.K., Homem De Bittencourt Jr., P.I., Newsholme, P.,

112
M. Krause et al. Experimental Gerontology 115 (2019) 104–113

2015a. The regulatory roles of NADPH oxidase, intra- and extra-cellular HSP70 in determinant of heat shock protein 70 serum concentrations during aging.
pancreatic islet function, dysfunction and diabetes. Clin. Sci. (Lond.) 128, 789–803. Biogerontology 5, 31–38.
Krause, M., Heck, T.G., Bittencourt, A., Scomazzon, S.P., Newsholme, P., Curi, R., Homem Noble, E.G., Milne, K.J., Melling, C.W., 2008. Heat shock proteins and exercise: a primer.
de Bittencourt Jr., P.I., 2015b. The chaperone balance hypothesis: the importance of Appl. Physiol. Nutr. Metab. 33, 1050–1065.
the extracellular to intracellular HSP70 ratio to inflammation-driven type 2 diabetes, Norton, C., Toomey, C., McCormack, W.G., Francis, P., Saunders, J., Kerin, E., Jakeman,
the effect of exercise, and the implications for clinical management. Mediat. Inflamm. P., 2016. Protein supplementation at breakfast and lunch for 24 weeks beyond ha-
249205, 2015. bitual intakes increases whole-body lean tissue mass in healthy older adults. J. Nutr.
Krause, M., Ludwig, M.S., Heck, T.G., Takahashi, H.K., 2015c. Heat shock proteins and 146, 65–69.
heat therapy for type 2 diabetes: pros and cons. Curr. Opin. Clin. Nutr. Metab. Care Paulsen, G., Hanssen, K.E., Ronnestad, B.R., Kvamme, N.H., Ugelstad, I., Kadi, F., Raastad,
18, 374–380. T., 2012. Strength training elevates HSP27, HSP70 and alphaB-crystallin levels in
Ku, Z., Yang, J., Menon, V., Thomason, D.B., 1995. Decreased polysomal HSP-70 may musculi vastus lateralis and trapezius. Eur. J. Appl. Physiol. 112, 1773–1782.
slow polypeptide elongation during skeletal muscle atrophy. Am. J. Phys. 268, Petry, E.R., Cruzat, V.F., Heck, T.G., Leite, J.S., Homem de Bittencourt Jr., P.I., Tirapegui,
C1369–C1374. J., 2014. Alanyl-glutamine and glutamine plus alanine supplements improve skeletal
Leite, J.C., Forte, R., de Vito, G., Boreham, C.A., Gibney, M.J., Brennan, L., Gibney, E.R., redox status in trained rats: involvement of heat shock protein pathways. Life Sci. 94,
2015. Comparison of the effect of multicomponent and resistance training programs 130–136.
on metabolic health parameters in the elderly. Arch. Gerontol. Geriatr. 60, 412–417. Philp, A., Hamilton, D.L., Baar, K., 2011. Signals mediating skeletal muscle remodeling by
de Lemos Muller, C.H., Rech, A., Botton, C.E., Schroeder, H.T., Bock, P.M., Farinha, J.B., resistance exercise: PI3-kinase independent activation of mTORC1. J Appl Physiol
Lopez, P., Scholer, C.M., Grigolo, G.B., Coelho, J., Kowalewski, L.S., Rodrigues, (1985) 110, 561–568.
M.I.L., de Azevedo, M.A., Quincozes-Santos, A., Rodrigues-Krause, J., Reischak- Rodrigues-Krause, J., Farinha, J.B., Krause, M., Reischak-Oliveira, A., 2016. Effects of
Oliveira, A., Pinto, R.S., De Vito, G., de Bittencourt Junior, P.I.H., Krause, M., 2018. dance interventions on cardiovascular risk with ageing: systematic review and meta-
Heat-induced extracellular HSP72 release is blunted in elderly diabetic people analysis. Complement. Ther. Med. 29, 16–28.
compared with healthy middle-aged and older adults, but it is partially restored by Senf, S.M., 2013. Skeletal muscle heat shock protein 70: diverse functions and therapeutic
resistance training. Exp. Gerontol. 111, 180–187. potential for wasting disorders. Front. Physiol. 4 (330).
Liu, Y., Lormes, W., Baur, C., Opitz-Gress, A., Altenburg, D., Lehmann, M., Steinacker, Senf, S.M., Dodd, S.L., Judge, A.R., 2010. FOXO signaling is required for disuse muscle
J.M., 2000. Human skeletal muscle HSP70 response to physical training depends on atrophy and is directly regulated by Hsp70. Am. J. Physiol. Cell Physiol. 298,
exercise intensity. Int. J. Sports Med. 21, 351–355. C38–C45.
Markofski, M.M., Dickinson, J.M., Drummond, M.J., Fry, C.S., Fujita, S., Gundermann, Sharman, M.J., Newton, R.U., Triplett-McBride, T., McGuigan, M.R., McBride, J.M.,
D.M., Glynn, E.L., Jennings, K., Paddon-Jones, D., Reidy, P.T., Sheffield-Moore, M., Hakkinen, A., Hakkinen, K., Kraemer, W.J., 2001. Changes in myosin heavy chain
Timmerman, K.L., Rasmussen, B.B., Volpi, E., 2015. Effect of age on basal muscle composition with heavy resistance training in 60- to 75-year-old men and women.
protein synthesis and mTORC1 signaling in a large cohort of young and older men Eur. J. Appl. Physiol. 84, 127–132.
and women. Exp. Gerontol. 65, 1–7. Tieland, M., van de Rest, O., Dirks, M.L., van der Zwaluw, N., Mensink, M., van Loon, L.J.,
Murton, A.J., 2015. Muscle protein turnover in the elderly and its potential contribution de Groot, L.C., 2012. Protein supplementation improves physical performance in frail
to the development of sarcopenia. Proc. Nutr. Soc. 74, 387–396. elderly people: a randomized, double-blind, placebo-controlled trial. J. Am. Med. Dir.
Naseeb, M.A., Volpe, S.L., 2017. Protein and exercise in the prevention of sarcopenia and Assoc. 13, 720–726.
aging. Nutr. Res. 40, 1–20. Verdijk, L.B., Jonkers, R.A., Gleeson, B.G., Beelen, M., Meijer, K., Savelberg, H.H.,
Nelson, R.J., Ziegelhoffer, T., Nicolet, C., Werner-Washburne, M., Craig, E.A., 1992. The Wodzig, W.K., Dendale, P., van Loon, L.J., 2009. Protein supplementation before and
translation machinery and 70 kd heat shock protein cooperate in protein synthesis. after exercise does not further augment skeletal muscle hypertrophy after resistance
Cell 71, 97–105. training in elderly men. Am. J. Clin. Nutr. 89, 608–616.
Nelson, A.R., Jackson, L., Clarke, J., Stellingwerff, T., Broadbent, S., Rowlands, D.S., Verhoeven, S., Vanschoonbeek, K., Verdijk, L.B., Koopman, R., Wodzig, W.K., Dendale, P.,
2013. Effect of post-exercise protein-leucine feeding on neutrophil function, im- van Loon, L.J., 2009. Long-term leucine supplementation does not increase muscle
munomodulatory plasma metabolites and cortisol during a 6-day block of intense mass or strength in healthy elderly men. Am. J. Clin. Nutr. 89, 1468–1475.
cycling. Eur. J. Appl. Physiol. 113, 2211–2222. Wiggs, M.P., 2016. Can endurance exercise preconditioning prevention disuse muscle
Njemini, R., Abeele, M.V., Demanet, C., Lambert, M., Vandebosch, S., Mets, T., 2002. Age- atrophy? Front. Physiol. 6, 63.
related decrease in the inducibility of heat-shock protein 70 in human peripheral Xue, H., Slavov, D., Wischmeyer, P.E., 2012. Glutamine-mediated dual regulation of heat
blood mononuclear cells. J. Clin. Immunol. 22, 195–205. shock transcription factor-1 activation and expression. J. Biol. Chem. 287,
Njemini, R., Demanet, C., Mets, T., 2004. Inflammatory status as an important 40400–40413.

113

Anda mungkin juga menyukai