Vous êtes sur la page 1sur 32

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/301562143

Aerobic Exercise Augments Muscle


Transcriptome Profile of Resistance Exercise
Article in AJP Regulatory Integrative and Comparative Physiology April 2016
Impact Factor: 3.11 DOI: 10.1152/ajpregu.00035.2016

READS

61

5 authors, including:
Tommy Lundberg

Rodrigo Fernandez-Gonzalo

Karolinska Institutet

Karolinska Institutet

10 PUBLICATIONS 99 CITATIONS

25 PUBLICATIONS 159 CITATIONS

SEE PROFILE

SEE PROFILE

Per A Tesch

Thomas Gustafsson

Karolinska Institutet

Karolinska Institutet

211 PUBLICATIONS 8,556 CITATIONS

120 PUBLICATIONS 2,588 CITATIONS

SEE PROFILE

SEE PROFILE

Available from: Tommy Lundberg


Retrieved on: 30 April 2016

Articles in PresS. Am J Physiol Regul Integr Comp Physiol (April 13, 2016). doi:10.1152/ajpregu.00035.2016

Aerobic Exercise Augments Muscle Transcriptome Profile of

Resistance Exercise

Tommy R. Lundberg1, Rodrigo Fernandez-Gonzalo2, Per A. Tesch2, Eric Rullman1, 3, Thomas

Gustafsson1.

University Hospital, Stockholm, Sweden,

Stockholm, Sweden, 3 Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden.

Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet and Karolinska
2

Department of Physiology & Pharmacology, Karolinska Institutet,

8
9

Running Head: Gene Signature of Concurrent Exercise

10
11
12

Address for Correspondance:

13

Tommy Lundberg, PhD

14

Karolinska Institutet, Department of Laboratory Medicine

15

Division of Clinical Physiology

16

Karolinska University Hospital, Huddinge

17

14186 Stockholm, Sweden

18

E-mail: tommy.lundberg@ki.se

19

Phone: +46(0)8-585 867 71

20
21
22
23
24
1
Copyright 2016 by the American Physiological Society.

25

Abstract

26
27

Recent reports suggest that aerobic exercise may boost the hypertrophic response to short-

28

term resistance training. This study explored the effects of an acute aerobic exercise bout on

29

the transcriptional response to subsequent resistance exercise. Ten moderately trained men

30

performed ~45 min cycling on one leg followed by 4x7 maximal knee extensions for each leg,

31

15 min later. Thus, one limb performed aerobic and resistance exercise (AE+RE), while the

32

opposing leg did resistance exercise only (RE). Biopsies were obtained from m. vastus

33

lateralis of each leg 3-h after the resistance exercise bout. Using DNA microarray, we

34

analyzed differences (1.5-fold, FDR 10%) in gene expression profiles for the two modes of

35

exercise. There were 176 genes up- (127) or down-regulated (49) by AE+RE compared with

36

RE. Among the most significant differentially expressed genes were established markers for

37

muscle growth and oxidative capacity, novel cytokines, transcription factors and microRNAs.

38

The most enriched functional categories were those linked to carbohydrate metabolism and

39

transcriptional regulation. Upstream analysis revealed that VEGF, CREB, TET2 and mTOR

40

were regulators highly activated by AE+RE, whereas JnK, Nf, MAPK and several

41

miRNAs were inhibited. Thus, aerobic exercise alters the skeletal muscle transcriptional

42

signature of resistance exercise to initiate important gene programs promoting both myofiber

43

growth and improved oxidative capacity. These results provide novel insight into human

44

muscle adaptations to diverse exercise modes and offer the very first genomic basis

45

explaining how aerobic exercise may augment, rather than compromise muscle growth

46

induced by resistance exercise.

47
48

Key words: Affymetrix, endurance, gene expression, microarray, strength

49
2

50

Introduction

51
52

It is often put forth that aerobic endurance-type exercise (e.g. running and cycling) interferes

53

with muscle adaptations to strength/resistance exercise executed as part of the same training

54

program (11, 12). Conversely, we have reported that concurrent aerobic exercise may in fact

55

boost skeletal muscle hypertrophic response to resistance exercise, accompanied by molecular

56

signaling responses that could favor increased net protein turnover (25, 27). At the same time,

57

the idea that aerobic exercise alone, particularly cycle training, may produce hypertrophy, has

58

been given more attention (17, 32). These reports are supported by studies showing that low-

59

load exercise may produce muscle growth and increased strength (17, 31). However, the

60

mechanisms explaining how aerobic exercise could act synergistically with resistance

61

exercise facilitating muscle growth, remain largely unexplored.

62

Any change in muscle mass arises from an altered balance between protein synthesis and

63

breakdown, favoring either muscle hypertrophy or atrophy (19). However, the precise

64

molecular networks underpinning altered protein balance and changes in muscle mass in

65

response to diverse exercise modes such as aerobic and resistance exercise, are not completely

66

understood. Based on acute measurements, we (24) and others (7) reported that concurrent

67

exercise may produce a more anabolic molecular and protein synthetic response compared

68

with resistance exercise alone. Indeed, muscle hypertrophy is typically accompanied by

69

mTOR-activation and increased muscle protein synthesis (29), yet these markers appear to

70

have minute predictive value to explain large individual variations in exercise-induced

71

hypertrophy (8, 30). Thus, the reductionistic single marker approach from acute studies

72

remains somewhat unsatisfactory to explain mechanisms governing muscle adaptations to

73

chronic exercise.

74

As newly introduced high-throughput techniques allow for in-depth analysis of the mRNA

75

responses to exercise, the transcriptional regulation of muscle hypertrophy has received

76

increased attention lately. In particular, microarray analysis makes it possible to

77

simultaneously assess a large number of muscle transcripts involved in muscle regenerative

78

processes in response to acute and chronic exercise (1, 35, 38). Studies have shown that

79

young individuals and/or high-responders to resistance exercise may show a different

80

signature with regard to myogenic growth transcripts, microRNAs (miRNA) and key

81

transcription factors than old individuals and/or low responders (1, 16). Moreover, while it

82

appears that several hundred genes co-vary with gains in lean mass, there are apparent

83

distinctions between acute and chronic exercise, as well as between aerobic- and resistance-

84

type exercise (33). To date, no study has examined muscle gene expression profile to gain

85

insight about the molecular regulation responsible for adaptations to combined aerobic and

86

resistance exercise.

87

In an attempt to reveal the molecular basis of why concurrent aerobic and resistance exercise

88

may produce different phenotypic adaptations (e.g. greater hypertrophy) than resistance

89

exercise alone, we examined subjects who had undertaken 5 weeks of resistance exercise with

90

(AE+RE) or without (RE) concurrent aerobic exercise. The specific aim was to analyze the

91

acute transcriptome response to a novel exercise bout of either AE+RE or RE. Gene

92

expression profiles were assessed by means of microarray analysis. As 5 weeks training using

93

the current AE+RE regimen produced substantially greater muscle hypertrophy than RE (27),

94

we hypothesized that an acute AE+RE bout would elicit a gene signature indicative of greater

95

muscle mass accretion, compared with RE.

96
97
98
4

99

Methods

100

Experimental design

101

After familiarization with the protocols and exercise equipment, ten participants performed an

102

acute exercise bout of knee extensor AE+RE for one leg, and RE only for the contralateral

103

limb (Fig. 1). The leg assigned to AE+RE was randomized in a counter-balanced manner. A

104

DNA microarray analysis was performed in biopsies obtained from m. vastus lateralis of both

105

legs 3 h after the resistance exercise bout. Subsequently, and as we have reported previously

106

(27), the subjects completed a 5-week training program using these identical exercise modes

107

(AE+RE vs RE).

108

Subjects

109

Ten men (26 5 yrs, 183 7 cm and 77 9 kg) volunteered for the study. Subjects were

110

moderately trained college students performing recreational exercise 2-3 dwk-1, (e.g. running

111

and team sports). At the time of the study, they had not experienced any lower limb injury for

112

the last 6 months or performed structured/intense resistance training in the past year. After

113

being informed about study procedures, risks and discomforts, subjects gave their written

114

informed consent to participate. The study experiments were approved by the Regional

115

Ethical Review Board in Stockholm.

116

Exercise experiments

117

Exercise equipment, protocols, and their efficacy in stimulating robust muscle adaptations to

118

5 weeks training, have been described in detail elsewhere (25, 27). After a standardized

119

warm-up, the aerobic exercise bout was performed in the morning of the experimental day

120

(~8.00 AM). It consisted of a 40 min isolated and dynamic knee extensions on a modified

121

one-legged ergometer (model 828E, Monark Exercise AB, Varberg, Sweden) at ~70% of

122

maximal workload (previously determined by a maximal incremental test) with a cadence of


5

123

60 rpm. Rate of perceived exertion was assessed regularly to confirm strenuous effort. Upon

124

completion of the 40 min, the workload was increased (~20 W) until subject failed to

125

maintain target cadence (~2-4 min). After 15 min recovery, resistance exercise for both legs

126

(one leg at a time) was carried out. Four sets of 7 maximal repetitions were performed on a

127

knee extension iso-inertial flywheel ergometer (YoYo Technology Inc., Stockholm, Sweden

128

(37)), with 2 min rest between sets. This exercise device uses the inertia of a spinning

129

flywheel (inertia, 0.11 kg/m2) to offer unlimited resistance during coupled concentric and

130

eccentric muscle actions. The range of motion was from 90 knee flexion to almost full

131

extension (i.e. 180). Power was measured in all repetitions. During all tests, subjects received

132

strong verbal encouragement from research personnel.

133

Muscle biopsies and diet control

134

Three hours after completion of the resistance exercise bout, biopsies were obtained from m.

135

vastus lateralis of both legs. Under local anesthesia, a 5-mm Bergstrm needle with suction

136

was employed to obtain muscle tissue samples (2). After excess blood, connective tissue

137

and/or fat had been removed; the tissue was frozen in liquid nitrogen and stored at -80C until

138

further analysis. Subjects received a standardized dinner the night before the experiments

139

(pasta, tomato sauce, and juice; 2.21 g carbohydrates/kg body weight (bw), 0.22 g protein/kg

140

bw, and 0.04 g fat/kg bw). In addition, standardized breakfast 2 hr prior to the aerobic

141

exercise bout was provided to participants (1.01 g carbohydrates/kg bw, 0.31 g protein/kg bw,

142

and 0.24 g fat/kg bw; Ensure Plus, Abbott Laboratories, Maidenhead, UK). Subjects then

143

remained fasted during the 3 h resting time prior to biopsy collection.

144

RNA extraction and microarray analysis

145

Total RNA was extracted from ~20-mg wet muscle samples using a bead beater device

146

(BioSpec Products, Inc., Bartlesville, OK) and TRIzol (Invitrogen Life Technologies,

147

Carlsbad, CA). Total RNA was then purified using PureYield RNA Midiprep System
6

148

(Promega Corporation, Madison, WI). Each of the 20 samples were subjected to analysis on

149

the Affymetrix HuGene-2.1-st platform. Hybridization, washing, staining and scanning of the

150

arrays were performed according to the manufacturers instructions (Affymetrix). To control

151

the quality, in addition to the standard quality assessments including scaling factors and chip

152

housekeeper 5/3-ratios, all individual arrays were examined using hierarchical clustering

153

and Normalized Unscaled Standard Error (NUSE, a variance based metric to identify outliers

154

prior to statistical analysis). All chips passed these QC-steps. The probe-set level intensities

155

for the arrays were normalized using the Robust Multi-array Analysis method (RMA)

156

implemented within the R statistical software environment using the Oligo package

157

(Bioconductor project). Raw data and RMA normalized expression values are publicly

158

available through Gene Expression Omnibus (GEO) accession number GSE74194. Prior to

159

differential expression analysis probe-sets without available annotation and probe-sets

160

displaying low-variance across samples (IQR <0.5) were excluded from further analysis.

161

Annotation was carried out in the R environment using hugene21sttranscriptcluster.db, and

162

the annotation and genefilter packages from Bioconductor. Differential expression was

163

estimated through pairwise-statistical analysis of microarrays (SAM) using 1000 permutations

164

on the R-platform. Significantly up- and down-regulated probe-sets were analyzed for gene-

165

ontology enrichment using the web-based Database for Annotation, Visualization and

166

Integrated Discovery (DAVID) version 6.7 (13, 14). Probe-sets with a FDR of 10% and a

167

fold change of 1.5-fold were analyzed for enrichment of biological functions. To correct

168

for possible selection bias of genes, only probe-sets used in the SAM analysis were used as

169

background. Enriched ontologies with a FDR of 10% were categorized as significant. For

170

further pathway analysis, we used the web-based bioinformatics tool Ingenuity Pathway

171

Analysis (IPA, http://www.ingenuity.com, Qiagen). IPA is a knowledge database generated

172

from peer-reviewed scientific publications that enables discovery of biological networks in

173

gene expression data and determines the biological functions most significant to those

174

networks. Affymetrix transcript identifiers were uploaded onto IPA and were re-annotated

175

and queried against the verified IPA knowledge database, and probe-sets analyzed in the

176

SAM-analysis were used as a reference set. Both up- and down-regulated probe-sets were

177

considered, and interactions annoted as predicted with high confidence or experimentially

178

validated were used in the IPA analysis. Enrichment of the focus genes in networks in IPA is

179

assessed via Fishers exact test. Furthermore, the software identifies top functions associated

180

with each network via enrichment scores (z-score), highlighting the predicted biological

181

significance of the results.

182

Validation of microarray

183

Real-time PCR for five selected mRNA targets was carried out on the same RNA extract as

184

the array, using TaqMan probes and primers as previously reported (27). Target gene

185

expression was reported as a ratio to reference genes (GAPDH and 18S) using the 2-CT

186

formula. Real-time PCR data were then scaled and plotted (Fig. 2) together with the

187

corresponding normalized probe-set expression levels of the array (Probe-set ID: 16974830,

188

17009093, 16906471, 16683771 & 17080788).

189
190
191
192
193
194
195
196

197

Results

198

Exercise responses and functional measures

199

During the acute resistance exercise bout, the leg that had completed aerobic exercise 15 min

200

earlier, produced 10% lower peak power than the rested RE-leg. The end-point results of the

201

5-wk training program have been reported previously (27). In brief, after 5 weeks training,

202

gains in peak power (~20%) were similar between AE+RE and RE. Likewise, the increase in

203

peak torque did not differ between AE+RE (10%) and RE (12%). With regard to muscle size,

204

AE+RE increased (p < 0.05) quadriceps muscle volume by 6%, compared with 3% for RE.

205

AE+RE, but not RE, increased endurance performance (22%) and citrate synthase activity

206

(18%).

207

Gene expression analysis

208

After annotation and filtering 11,869 probe-sets were analyzed using pairwise SAM analysis.

209

Of these probe-sets, 49 were down- and 127 up-regulated (1.5-fold) by AE+RE as compared

210

with RE, having a Q-value (corresponding to FDR) of 10% (See Supplemental Digital

211

Content 1, Differential Expression Analysis). Analysis of biological functions significantly

212

enriched amongst the up-regulated genes using DAVID identified seven ontologies, all of

213

which can be defined as variations of carbohydrate metabolism (Table 1). In contrast, 11

214

biological functions were identified as enriched among the down-regulated genes. Most

215

notably were variations of response to hormone stimulus, MAPKKK cascade, regulation

216

of skeletal muscle tissue development and angiogenesis (Table 1). Some key down-

217

regulated genes found in several of these clusters were transcriptional and epigenetic

218

regulators such as HDAC and TXNIP. The 25 most significantly differentially expressed

219

genes (up- and down-regulated by AE+RE compared with RE) are displayed in Table 2.

220

Several of these genes are well-known markers involved in muscle adaptations to exercise

221

(e.g. AMPK, PGC-1, MuRF-1, myostatin). Some cytokines (IL18 and IL-311RA) and
9

222

several miRNAs (mir-133, mir-1, mir-623) appeared among the down-regulated genes. As

223

indicated earlier, we compared five exercise-relevant differentially expressed genes (PGC-1,

224

VEGF, myostatin, MuRF-1, atrogin-1) from the array with q-PCR data previously published

225

from the same RNA (27). In both the microarray and the q-PCR, the five genes were found to

226

be significantly differentially expressed in the corresponding manner (Fig. 2).

227

Upstream analysis

228

Ingenuitys Upstream Regulator Analysis in IPA is a tool that predicts upstream regulators

229

from gene expression data based on the literature and compiled in the Ingenuity Knowledge

230

Base. A Fishers Exact Test p-value was calculated to assess the significance of enrichment of

231

the gene expression data for the genes downstream of an upstream regulator. In addition,

232

directionality and magnitude of the differentially expressed genes was calculated to a z-score

233

of directional consistency. A high positive z-score indicates activation for that pathway

234

whereas a negative z-score is associated with an inhibition by AE+RE as compared with RE.

235

The top-ranking regulatory molecules based solely on overlap (i.e. p-value of Fishers Exact

236

test) were MYC and TNF- (-log10 p-value >10), followed by HGF, IL1 and FOXO3.

237

However, z-score of all of these pathways were in the range of -0.6 0.2 illustrating that

238

while many genes in these pathways are differentially expressed, the directionality of the

239

altered gene expression is inconsistent with an activation or inhibition of the pathway.

240

When considering the regulatory molecules with a gene expression pattern consistent with

241

what is expected (i.e. z-score >2), the #1 pathway predicted to be activated was VEGF (p-

242

value 3e-7, z-score 2.7) and miR-181a which was predicted to be inhibited (p-value 1.7e-7, z-

243

score -2.6), see Figure 3 and 4, and Table 3. The pathways predicted to be activated second to

244

VEGF were CREB (p-value 5.6e-4, z-score 2.2), TET2 (p-value 6.9e-4, z-score 2.0) and

245

mTOR (p-value 5.3e-3, z-score 2.4). The second and third most inhibited regulators were JnK

246

(p-value 4.5e-5, z-score -2.4) and Nf, followed by MAP2K4 and MAP3K8 (p-value 7.8e-5,
10

247

z-score -2.5). See Figure 3 and 4, Table 3, and Supplemental Digital Content 2 (Upstream

248

Analysis).

249
250
251
252
253
254
255
256
257
258
259
260
261
262
263
264
265
266
267
268
269
270
271
11

272

Discussion

273
274

This study provides the first comprehensive gene expression analysis comparing concurrent

275

aerobic and resistance exercise (AE+RE) with resistance exercise alone (RE). The main

276

finding was that an exhaustive aerobic exercise insult, performed prior to resistance exercise,

277

augmented important gene signatures that regulate contrasting functions such as oxidative

278

metabolism on one hand, and myofiber growth and tissue regeneration on the other. Thus, we

279

have characterized a molecular signature of concurrent exercise that could aid in explaining

280

how aerobic exercise may boost, rather than compromise, muscle hypertrophy to chronic

281

resistance training.

282

While we and others have reported on single molecular markers in response to concurrent

283

aerobic and resistance exercise (7, 24, 27), neither study provided an in-depth transcriptional

284

map of this exercise mode. We hypothesized that there must be substantial differences in the

285

transcriptome response of an exercise regime (AE+RE) producing twice as robust

286

hypertrophy as RE alone (27). Indeed, our results showed that 176 genes were differently

287

expressed 1.5-fold when aerobic exercise preceded a bout of resistance exercise. This would

288

suggest that these genes, and their associated targets and regulators, play important roles in

289

eliciting phenotype differentiation depending on exercise stimulus imposed over 5 weeks.

290

In an effort to explore the rather substantial difference in expression across exercise modes,

291

we performed analysis for enrichment of gene ontologies related to biological functions.

292

Perhaps not surprising, the most activated biological functions induced by AE+RE were

293

processes involved in carbohydrate metabolism. Indeed, it has been shown that aerobic

294

exercise promotes gene programs involved in glucose homeostasis and oxidative metabolism

295

(28). Similarly, the gene showing the most significant difference in expression across exercise

296

modes was AMPK, a well-known regulator of carbohydrate metabolism. Furthermore, PGC12

297

1 was highly up-regulated by AE+RE, confirming that this gene and its exercise-responsive

298

splice variants are up-regulated by both aerobic and resistance exercise, and more so by

299

combined exercise (26, 42). The fact that AMPK and PGC-1 were prominent genes in this

300

unbiased expression analysis further underlines their role in promoting an oxidative

301

phenotype (20). It also fits with the impressive increase in endurance performance and CS

302

activity reported by us following AE+RE training (27).

303

Among functions being down-regulated by AE+RE compared with RE, it is worth noting we

304

observed genes involved in regulation of skeletal muscle tissue development. Genes

305

clustered in this group have a negative effect on muscle hypertrophy, and one of the most

306

down-regulated gene was myostatin, an established negative regulator of hypertrophy (36). In

307

addition, we found key genes involved in muscle protein breakdown to be up-regulated, e.g.

308

MuRF-1 and atrogin-1. This adds further credibility to the recent notion that these ubiquitin

309

ligase proteins may be critical in the healthy skeletal muscle remodeling process, in fact

310

facilitating contraction-induced muscle growth (15). Overall, the response of key genes and

311

associated biological functions activated by AE+RE concords with the greater muscle mass

312

accretion following the concurrent exercise regime.

313

To identify transcriptional regulators potentially explaining the observed gene expression

314

differences between AE+RE and RE, we used IPA upstream regulator analysis. The analysis

315

revealed that factors centralized around the key regulators VEGF, CREB, TET2 and mTOR

316

were highly activated by AE+RE, whereas JnK, Nf, MAPK and several miRNAs were

317

inhibited regulators. While mTOR is a growth-inducer, its role in regulating muscle mass is

318

not entirely unequivocal. Thus, recent research suggest high responders to resistance exercise

319

display an inhibited mTOR-activation signature (33), and that the muscle anabolic response to

320

exercise is not solely dependent on mTOR activation (34). Nevertheless, mTOR aligns with

321

muscle hypertrophy (29) and is deemed crucial for the hypertrophic response and for
13

322

protection against atrophy (3). As our first hand physiological data would be hard to challenge

323

(27), the demonstration of greater activation of mTOR following AE+RE than RE would be

324

indicative of a greater protein synthetic response. Ultimately, this aids in explaining the more

325

robust hypertrophic response to concurrent execise.

326

MAPK- and Nf-associated signaling can be both adaptive and maladaptive in skeletal

327

muscle depending on the conditions (18). However, given that exercise seems potent to

328

increase MAPK and Nf in the non-diseased state (18, 41), it strikes that MAPK/JnK and

329

Nf were identified as inhibited regulators in the current study. In support of this notion,

330

among the top genes being down-regulated by AE+RE, we found genes involved in myokine

331

(e.g. IL-18, IL-31RA) and MAPK/ERK signaling (ATF3). Thus, while activation of these

332

genes in response to acute exercise appears to benefit muscle adaptations, chronic activation

333

could very well exacerbate protein breakdown and hence muscle wasting (18). In light of this,

334

the AE+RE-induced down-regulation of these genes was perhaps due to a volume-related

335

attenuation of MAPK-signaling (22).

336

A less studied factor in human skeletal muscle remodeling is the transcriptional regulator

337

CREB, which was significantly activated as an upstream regulator in the current analysis. It

338

has been reported that high-intensity exercise activates CREB in mice, and the activation of

339

this transcriptional complex induces anabolic changes driving muscle hypertrophy during

340

subsequent recovery (4). Furthermore, over-expressing cells with CREB-regulated co-

341

regulators induced hypertrophy in transgenic mouse models and cultured myotubes (4). Our

342

findings spur future studies to examine weather CREB and its associated targets play a key

343

role in exercise-induced adaptations of human skeletal muscle.

344

miRNAs are accepted post-transcriptional regulators of protein translation which appear to

345

block translation of transcribed genes in human skeletal muscle (9). Several miRNA routes

14

346

were inhibited by AE+RE in the upstream regulator analysis. In further support of this notion,

347

some of the most established myomirs (miR-133 and miR-206/miR1) were significantly

348

less expressed after AE+RE compared with RE. Hence, it is tempting to speculate that the

349

blunted miRNA expression in AE+RE could reverse inhibitory effects on target gene

350

transcription. This in turn may allow for more efficient protein translation from available

351

mRNA transcripts, resulting in more robust muscle protein accretion following the combined

352

exercise paradigm. Interestingly, miRNAs also seem to regulate TET2 signaling (6), which

353

was heavily activated by AE+RE in the current study. To the authors knowledge, we are the

354

first to depict the TET2 pathway as a potential regulator of adaptations to combined training.

355

TET2 is a master epigenetic regulator of smooth muscle plasticity (21), yet its role in human

356

skeletal muscle is relatively unknown. The TET2 transcript was fairly abundant and

357

detectable in all samples, yet it was discarded from further analysis due to low variability

358

across samples (SD ~0.2). Post-hoc analysis of TET2 did not indicate it being differentially

359

expressed. Therefore, a potential role for TET2 in the context of skeletal muscle response to

360

exercise does not seem to involve transcriptional regulation of TET2 itself. However, given

361

that TET2 may be under heavy miRNA regulation (6), recalling we discovered several

362

suppressed miRNAs in our gene list, future studies should explore the potential interaction

363

between miRNA, TET2 and muscle adaptive responses to exercise in more detail.

364

The ontology analysis surprisingly revealed that AE+RE, while improving endurance

365

capacity, down-regulated the biological function angiogenesis. This was due to the fact that

366

several known angiogenic factors were down-regulated by combined exercise. In contrast, the

367

pro-angiogenic growth factor VEGF (10) showed substantially greater expression after

368

AE+RE than RE, and this was confirmed through q-PCR. This highlights the risk of drawing

369

conclusions about complex biological processes based on measurements of single factors. In

370

further support of a suppressed angiogenic response, we have reported that 5 weeks AE+RE
15

371

(25) did not induce capillarization (23). Thus, albeit the training period may have been too

372

short for vascular growth to occur, it cannot be excluded that our finding indicates an

373

interference effect in vascular remodeling induced by adding resistance exercise to aerobic

374

exercise.

375

It is obvious that in order to assess the effects of adding aerobic exercise prior to resistance

376

exercise, the resistance exercise protocol needs to be matched and hence total work performed

377

is substantially greater in the AE+RE leg. Although this is an essential part of the design for

378

this and other comparable concurrent exercise studies, it does not rule out the possibility that

379

differences in workloads account for the different gene expression profiles across legs. As the

380

primary goal of this study was to comprehensively explore gene expression profiles of the two

381

exercise modes, we did not assess protein-related changes of any specific target, nor did we

382

specifically profile transcripts in the polysome pool (5). However, we appreciate that further

383

validation work could have revealed even more significant information. The specific one-

384

legged model was selected because it allows for a unique inter-individual design with

385

repeated-measure analysis of samples originating from the same individual and hence the

386

same genetic variance, training history and nutrient status. We acknowledge that the acute

387

nature of the imposed exercise challenges and subsequent biopsy sampling time-point could

388

produce large noise and non-specific transcriptional responses (39). This was reflected in our

389

data set where members of several key regulatory pathways were differentially expressed in a

390

discordant manner i.e. both up- and down-regulation. While this highlights the complexity in

391

interpreting acute exercise data, it also emphasizes a major strength of the current study, i.e.

392

first hand data of muscle adaptations to the exercise modes under study (40). In the absence of

393

such knowledge, it becomes virtually impossible to interpret the acute molecular response to

394

exercise.

395
16

396

Perspectives and Significance

397

We provide novel data revealing that the skeletal muscle, subjected to concurrent exercise,

398

shows striking transcriptional responses when initiating gene programs generally thought to

399

be antagonistic with each other. In addition to providing unbiased support for factors with

400

established roles in regulating muscle growth and an oxidative phenotype in response to

401

concurrent exercise (e.g. mTOR, myostatin, AMPK, PGC-1), our in-depth transcriptional

402

map also identified several new candidate genes and putative networks that markedly differed

403

in expression between AE+RE and RE. Thus, central regulators such as CREB, TET2,

404

MAPK, Nf, and several miRNAs appeared as novel factors that could be part in regulating

405

vast differences in types and magnitudes of adaptations across combined exercise paradigms.

406

The current findings also highlight the difficulty in assessing single markers as a proxy of

407

physiological adaptations if no end-point measures of chronic adaptations are at hand.

408

Collectively, our findings should stimulate further exhaustive investigations of the

409

transcriptional and molecular basis of exercise-induced muscle adaptations using different

410

exercise combinations in various populations.

411
412

Acknowledgements

413

The authors would like to thank BEA - the core facility for Bioinformatics and Expression

414

Analysis service facility at Novum, Karolinska Institutet in Huddinge, for their helpful

415

contribution. This study was supported by grants from the Swedish National Centre for

416

Research in Sports (PAT), the European Space Agency (PAT, TG), the Marianne & Marcus

417

Wallenberg Foundation (TG), the Swedish Medical Research Council (TG), and the Swedish

418

Society for Medical Research (SSFM, ER).

419
420
17

421

References

422

1. Bamman MM, Petrella JK, Kim JS, Mayhew DL, and Cross JM. Cluster analysis tests

423

the importance of myogenic gene expression during myofiber hypertrophy in humans. J Appl

424

Physiol 102: 2232-2239, 2007.

425

2. Bergstrm J. Muscle Electrolytes in Man. Scand J Lab Med Invest 14: 1-110, 1962.

426

3. Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R, Zlotchenko E,

427

Scrimgeour A, Lawrence JC, Glass DJ, and Yancopoulos GD. Akt/mTOR pathway is a

428

crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat

429

Cell Biol 3: 1014-1019, 2001.

430

4. Bruno NE, Kelly KA, Hawkins R, Bramah-Lawani M, Amelio AL, Nwachukwu JC,

431

Nettles KW, and Conkright MD. Creb coactivators direct anabolic responses and enhance

432

performance of skeletal muscle. EMBO J 33: 1027-1043, 2014.

433

5. Chen YW, Nader GA, Baar KR, Fedele MJ, Hoffman EP, and Esser KA. Response of

434

rat muscle to acute resistance exercise defined by transcriptional and translational profiling. J

435

Physiol 545: 27-41, 2002.

436

6. Cheng J, Guo S, Chen S, Mastriano SJ, Liu C, D'Alessio AC, Hysolli E, Guo Y, Yao

437

H, Megyola CM, Li D, Liu J, Pan W, Roden CA, Zhou XL, Heydari K, Chen J, Park IH,

438

Ding Y, Zhang Y, and Lu J. An extensive network of TET2-targeting MicroRNAs regulates

439

malignant hematopoiesis. Cell Rep 5: 471-481, 2013.

440

7. Donges CE, Burd NA, Duffield R, Smith GC, West DW, Short MJ, Mackenzie R,

441

Plank LD, Shepherd PR, Phillips SM, and Edge JA. Concurrent resistance and aerobic

442

exercise stimulates both myofibrillar and mitochondrial protein synthesis in sedentary middle-

443

aged men. J Appl Physiol 112: 1992-2001, 2012.

18

444

8. Fernandez-Gonzalo R, Lundberg TR, and Tesch PA. Acute molecular responses in

445

untrained and trained muscle subjected to aerobic and resistance exercise training versus

446

resistance training alone. Acta Physiol 209: 283-294, 2013.

447

9. Gallagher IJ, Scheele C, Keller P, Nielsen AR, Remenyi J, Fischer CP, Roder K,

448

Babraj J, Wahlestedt C, Hutvagner G, Pedersen BK, and Timmons JA. Integration of

449

microRNA changes in vivo identifies novel molecular features of muscle insulin resistance in

450

type 2 diabetes. Genome Med 2: 9, 2010.

451

10. Gustafsson T. Vascular remodelling in human skeletal muscle. Biochem Soc Trans 39:

452

1628-1632, 2011.

453

11. Hawley JA. Molecular responses to strength and endurance training: are they

454

incompatible? Appl Physiol Nutr Metab 34: 355-361, 2009.

455

12. Hickson RC. Interference of strength development by simultaneously training for

456

strength and endurance. Eur J Appl Physiol Occup Physiol 45: 255-263, 1980.

457

13. Huang da W, Sherman BT, and Lempicki RA. Bioinformatics enrichment tools: paths

458

toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res 37: 1-13,

459

2009.

460

14. Huang da W, Sherman BT, and Lempicki RA. Systematic and integrative analysis of

461

large gene lists using DAVID bioinformatics resources. Nat Protoc 4: 44-57, 2009.

462

15. Hwee DT, Baehr LM, Philp A, Baar K, and Bodine SC. Maintenance of muscle mass

463

and load-induced growth in Muscle RING Finger 1 null mice with age. Aging Cell 13: 92-

464

101, 2014.

465

16. Kim JS, Petrella JK, Cross JM, and Bamman MM. Load-mediated downregulation of

466

myostatin mRNA is not sufficient to promote myofiber hypertrophy in humans: a cluster

467

analysis. J Appl Physiol 103: 1488-1495, 2007.

19

468

17. Konopka AR and Harber MP. Skeletal muscle hypertrophy after aerobic exercise

469

training. Exercise and sport sciences reviews 42: 53-61, 2014.

470

18. Kramer HF and Goodyear LJ. Exercise, MAPK, and NF-kappaB signaling in skeletal

471

muscle. J Appl Physiol 103: 388-395, 2007.

472

19. Kumar V, Atherton P, Smith K, and Rennie MJ. Human muscle protein synthesis and

473

breakdown during and after exercise. J Appl Physiol 106: 2026-2039, 2009.

474

20. Lantier L, Fentz J, Mounier R, Leclerc J, Treebak JT, Pehmoller C, Sanz N,

475

Sakakibara I, Saint-Amand E, Rimbaud S, Maire P, Marette A, Ventura-Clapier R,

476

Ferry A, Wojtaszewski JF, Foretz M, and Viollet B. AMPK controls exercise endurance,

477

mitochondrial oxidative capacity, and skeletal muscle integrity. FASEB J 28: 3211-3224,

478

2014.

479

21. Liu R, Jin Y, Tang WH, Qin L, Zhang X, Tellides G, Hwa J, Yu J, and Martin KA.

480

Ten-eleven translocation-2 (TET2) is a master regulator of smooth muscle cell plasticity.

481

Circulation 128: 2047-2057, 2013.

482

22. Long YC, Widegren U, and Zierath JR. Exercise-induced mitogen-activated protein

483

kinase signalling in skeletal muscle. Proc Nutr Soc 63: 227-232, 2004.

484

23. Lundberg T. The effects of aerobic exercise on human skeletal muscle adaptations to

485

resistance exercise. Doctoral thesis 181 in Sports Science. Mid Sweden University: 1-73,

486

2014.

487

24. Lundberg TR, Fernandez-Gonzalo R, Gustafsson T, and Tesch PA. Aerobic exercise

488

alters skeletal muscle molecular responses to resistance exercise. Med Sci Sports Exerc 44:

489

1680-1688, 2012.

490

25. Lundberg TR, Fernandez-Gonzalo R, Gustafsson T, and Tesch PA. Aerobic exercise

491

does not compromise muscle hypertrophy response to short-term resistance training. J Appl

492

Physiol 114: 81-89, 2013.

20

493

26. Lundberg TR, Fernandez-Gonzalo R, Norrbom J, Fischer H, Tesch PA, and

494

Gustafsson T. Truncated splice variant PGC-1alpha4 is not associated with exercise-induced

495

human muscle hypertrophy. Acta Physiol 212: 142-151, 2014.

496

27. Lundberg TR, Fernandez-Gonzalo R, and Tesch PA. Exercise-induced AMPK

497

activation does not interfere with muscle hypertrophy in response to resistance training in

498

men. J Appl Physiol 116: 611-620, 2014.

499

28. Mahoney DJ, Parise G, Melov S, Safdar A, and Tarnopolsky MA. Analysis of global

500

mRNA expression in human skeletal muscle during recovery from endurance exercise.

501

FASEB J 19: 1498-1500, 2005.

502

29. Mayhew DL, Kim JS, Cross JM, Ferrando AA, and Bamman MM. Translational

503

signaling responses preceding resistance training-mediated myofiber hypertrophy in young

504

and old humans. J Appl Physiol 107: 1655-1662, 2009.

505

30. Mitchell CJ, Churchward-Venne TA, Parise G, Bellamy L, Baker SK, Smith K,

506

Atherton PJ, and Phillips SM. Acute post-exercise myofibrillar protein synthesis is not

507

correlated with resistance training-induced muscle hypertrophy in young men. PloS one 9:

508

e89431, 2014.

509

31. Mitchell CJ, Churchward-Venne TA, West DW, Burd NA, Breen L, Baker SK, and

510

Phillips SM. Resistance exercise load does not determine training-mediated hypertrophic

511

gains in young men. J Appl Physiol 113: 71-77, 2012.

512

32. Ozaki H, Loenneke JP, Thiebaud RS, and Abe T. Cycle training induces muscle

513

hypertrophy and strength gain: strategies and mechanisms. Acta Physiol Hung 102: 1-22,

514

2015.

515

33. Phillips BE, Williams JP, Gustafsson T, Bouchard C, Rankinen T, Knudsen S, Smith

516

K, Timmons JA, and Atherton PJ. Molecular networks of human muscle adaptation to

517

exercise and age. PLoS genetics 9: e1003389, 2013.

21

518

34. Philp A, Schenk S, Perez-Schindler J, Hamilton DL, Breen L, Laverone E, Jeromson

519

S, Phillips SM, and Baar K. Rapamycin does not prevent increases in myofibrillar or

520

mitochondrial protein synthesis following endurance exercise. J Physiol 593: 4275-4284,

521

2015.

522

35. Raue U, Trappe TA, Estrem ST, Qian HR, Helvering LM, Smith RC, and Trappe S.

523

Transcriptome signature of resistance exercise adaptations: mixed muscle and fiber type

524

specific profiles in young and old adults. J Appl Physiol 112: 1625-1636, 2012.

525

36. Schuelke M, Wagner KR, Stolz LE, Hubner C, Riebel T, Komen W, Braun T, Tobin

526

JF, and Lee SJ. Myostatin mutation associated with gross muscle hypertrophy in a child.

527

New Eng J Med 350: 2682-2688, 2004.

528

37. Tesch PA, Ekberg A, Lindquist DM, and Trieschmann JT. Muscle hypertrophy

529

following 5-week resistance training using a non-gravity-dependent exercise system. Acta

530

Physiol Scand 180: 89-98, 2004.

531

38. Thalacker-Mercer A, Stec M, Cui X, Cross J, Windham S, and Bamman M. Cluster

532

analysis reveals differential transcript profiles associated with resistance training-induced

533

human skeletal muscle hypertrophy. Physiol Genomics 45: 499-507, 2013.

534

39. Timmons JA. Variability in training-induced skeletal muscle adaptation. J Appl Physiol

535

110: 846-853, 2011.

536

40. Timmons JA and Sundberg CJ. Oligonucleotide microarray expression profiling:

537

human skeletal muscle phenotype and aerobic exercise training. IUBMB Life 58: 15-24, 2006.

538

41. Vella L, Caldow MK, Larsen AE, Tassoni D, Della Gatta PA, Gran P, Russell AP,

539

and Cameron-Smith D. Resistance exercise increases NF-kappaB activity in human skeletal

540

muscle. Am J Physiol Regul Integr Comp Physiol 302: R667-673, 2012.

22

541

42. Ydfors M, Fischer H, Mascher H, Blomstrand E, Norrbom J, and Gustafsson T. The

542

truncated splice variants, NT-PGC-1alpha and PGC-1alpha4, increase with both endurance

543

and resistance exercise in human skeletal muscle. Physiol Rep 1: e00140, 2013.

544
545
546
547

Supplemental Digital Content

548

Supplemental Digital Content 1.xls: Differential Expression analysis

549

Supplemental Digital Content 2.xls: Upstream Analysis

550
551
552
553
554
555
556
557
558
559
560
561
562
563

23

564

Figure legends

565
566

Figure 1. Schematics of the acute exercise experiment. Muscle biopsies from each leg (B)

567

were obtained 3 h after an acute bout of resistance exercise with (AE+RE) or without (RE)

568

preceding aerobic exercise.

569
570

Figure 2. Validation of the microarray and previously reported real-time PCR data. Lines

571

represent individual real-time PCR observations whereas the microarray data are depicted as

572

boxplots. These are plotted as median, first and fourth quartile, and range.

573
574

Figure 3. Upstream regulator analysis by Ingenuity pathway analysis (IPA). The figures

575

illustrate the top-ranking pathways predicted as activated in Upstream regulator analysis.

576

Genes in red were greater expressed in AE+RE compared with RE whereas genes in green

577

were lower expressed. An orange line indicates predicted up-regulation (by AE+RE) whereas

578

a blue line indicates predicted down-regulation. A yellow line indicates expression being

579

contradictory to the prediction.

580
581

Figure 4. Upstream regulator analysis by Ingenuity pathway analysis (IPA). The figures

582

illustrate the top-ranking pathways predicted as inhibited in Upstream regulator analysis.

583

Genes in red were greater expressed in AE+RE compared with RE whereas genes in green

584

were lower expressed. An orange line indicates predicted up-regulation (by AE+RE) whereas

585

a blue line indicates predicted down-regulation. A yellow line indicates expression being

586

contradictory to the prediction.


24

Myostatin

MuRF-1

Atrogin-1

VEGF

PGC-1

Table 1. Gene ontology (GO) categories related to biological processes induced by AE+RE
compared with RE as generated by DAVID.
GO term
Up-regulated processes
GO:0010906~regulation of glucose metabolic process
GO:0010675~regulation of cellular carbohydrate metabolic process
GO:0006109~regulation of carbohydrate metabolic process
GO:0019318~hexose metabolic process
GO:0015980~energy derivation by oxidation of organic compounds
GO:0006006~glucose metabolic process
GO:0005996~monosaccharide metabolic process
Down-regulated processes
GO:0007167~enzyme linked receptor protein signaling pathway
GO:0010033~response to organic substance
GO:0009725~response to hormone stimulus
GO:0042127~regulation of cell proliferation
GO:0009719~response to endogenous stimulus
GO:0007169~transmembrane receptor protein tyrosine kinase signaling pathway
GO:0001525~angiogenesis
GO:0009266~response to temperature stimulus
GO:0051789~response to protein stimulus
GO:0000165~MAPKKK cascade
GO:0048641~regulation of skeletal muscle tissue development

Fold
Enrichment

FDR (%)

15.71
14.89
14.89
4.67
7.69
5.30
4.12

2.81
3.30
3.30
5.27
5.59
7.58
9.47

6.66
4.08
5.43
3.62
4.94
6.77
7.61
12.18
11.63
6.48
23.10

0.06
0.32
0.76
0.85
1.32
2.40
5.43
5.72
6.48
9.44
9.21

Table 2. The 25 top up- and down-regulated genes by AE+RE compared with RE out of 176
differentially expressed genes (FDR 10%).
Top genes up-regulated by AE+RE compared with RE
Fold change Gene symbol
Entrez Gene Name
10.85
PRKAG2
protein kinase, AMP-activated, gamma 2 non-catalytic subunit
2.96
TRIM63
tripartite motif containing 63, E3 ubiquitin protein ligase
2.87
KCNQ4
potassium channel, voltage gated KQT-like subfamily Q, member 4
2.85
RPS6KA2-IT1 RPS6KA2 intronic transcript 1
2.75
PPARGC1A
peroxisome proliferator-activated receptor gamma, coactivator 1 alpha
2.61
CHST15
carbohydrate (N-acetylgalactosamine 4-sulfate 6-O) sulfotransferase 15
2.61
FAM102A
family with sequence similarity 102, member A
2.60
SNAI3
snail family zinc finger 3
2.48
MAFF
v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog F
2.39
ESRRG
estrogen-related receptor gamma
2.37
CES3
carboxylesterase 3
2.37
SLC20A1
solute carrier family 20 (phosphate transporter), member 1
2.36
NR4A3
nuclear receptor subfamily 4, group A, member 3
2.36
PFKFB2
6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 2
2.32
GPR157
G protein-coupled receptor 157
2.32
SLC25A33
solute carrier family 25 (pyrimidine nucleotide carrier), member 33
2.26
SLC38A1
solute carrier family 38, member 1
2.26
SYNJ2
synaptojanin 2
2.25
SLC16A6
solute carrier family 16, member 6
2.18
PXDC1
PX domain containing 1
2.16
LOC101928327 uncharacterized LOC101928327
2.14
CIART
circadian associated repressor of transcription
2.11
ANGPTL2
angiopoietin-like 2
2.08
MIR3147
microRNA 3147
2.05
BHLHE40
basic helix-loop-helix family, member e40
Top genes down-regulated by AE+RE compared with RE
Fold change Gene symbol
Entrez Gene Name
0.36
OTUD1
OTU deubiquitinase 1
0.41
ATF3
activating transcription factor 3
0.42
SERPINE1
serpin peptidase inhibitor, clade E, member 1
0.44
IL18
interleukin 18
0.45
IL31RA
interleukin 31 receptor A
0.47
KCNQ5-IT1
KCNQ5 intronic transcript 1
0.47
GADD45A
growth arrest and DNA-damage-inducible, alpha
0.47
ARRDC4
arrestin domain containing 4
0.48
NEXN-AS1
NEXN antisense RNA 1
0.48
CYR61
cysteine-rich, angiogenic inducer, 61
0.51
BTG2
BTG family, member 2
0.51
IER5
immediate early response 5
0.52
RUNX1-IT1
RUNX1 intronic transcript 1
0.53
mir-133
microRNA 133b
0.54
CTGF
connective tissue growth factor
0.54
ENAH
enabled homolog (Drosophila)
0.54
CA4
carbonic anhydrase IV
0.55
FOS
FBJ murine osteosarcoma viral oncogene homolog
0.56
PLAU
plasminogen activator, urokinase
0.56
SPNS2
spinster homolog 2 (Drosophila)
0.57
PFKFB3
6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3
0.57
SLC8A3
solute carrier family 8 (sodium/calcium exchanger), member 3
0.57
TIAM2
T-cell lymphoma invasion and metastasis 2
0.57
ARID5A
AT rich interactive domain 5A (MRF1-like)
0.58
MSTN
myostatin

Table 3. The top ranked activated/inhibited central regulators in the IPA analysis.
Predicted
activation
state

Zscore

P-value of
overlap

miR-181a-5p (and other miRNAs w/seed ACAUUCA)

Inhibited

-2.57

0.000000167

Vegf

Activated

2.67

0.000000303

Jnk

Inhibited

-2.35

0.0000447

NFkB (complex)

Inhibited

-2.52

0.0000775

MAP2K4

Inhibited

-2.19

0.000101

MAP3K8

Inhibited

-2.00

0.000107

miR-141-3p (and other miRNAs w/seed AACACUG)

Inhibited

-2.81

0.000341

miR-291a-3p (and other miRNAs w/seed AAGUGCU)

Inhibited

-3.25

0.000544

Creb

Activated

2.18

0.000561

miR-17-5p (and other miRNAs w/seed AAAGUGC)

Inhibited

-2.82

0.000633

TET2

Activated

2.00

0.000685

MTOR

Activated

2.41

0.00525

Upstream regulator

Vous aimerez peut-être aussi