Next Article in Journal
Li3V2(PO4)3 Cathode Material: Synthesis Method, High Lithium Diffusion Coefficient and Magnetic Inhomogeneity
Previous Article in Journal
Applying Proteomics and Computational Approaches to Identify Novel Targets in Blast-Associated Post-Traumatic Epilepsy
Previous Article in Special Issue
Skeletal Muscle-Specific Bis Depletion Leads to Muscle Dysfunction and Early Death Accompanied by Impairment in Protein Quality Control
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Acute, Short-, and Long-Term Effects of Endurance Exercise on Skeletal Muscle Transcriptome Profiles

1
Department of Sports Medicine, Medical Clinic, Eberhard-Karls-University of Tübingen, 72076 Tübingen, Germany
2
Department of Sport and Rehabilitation Medicine, University of Ulm, 89075 Ulm, Germany
3
Department of Exercise Physiology and Sports Therapy, University of Gießen, 35394 Gießen, Germany
4
Department of Medicine, Faculty of Health, University of Witten/Herdecke, 58455 Witten, Germany
*
Author to whom correspondence should be addressed.
Author affiliation when research was conducted.
Int. J. Mol. Sci. 2024, 25(5), 2881; https://doi.org/10.3390/ijms25052881
Submission received: 31 January 2024 / Revised: 27 February 2024 / Accepted: 28 February 2024 / Published: 1 March 2024
(This article belongs to the Special Issue Recent Advances in Skeletal Muscle Physiology and Pathophysiology)

Abstract

:
A better understanding of the cellular and molecular mechanisms that are involved in skeletal muscle adaptation to exercise is fundamentally important to take full advantage of the enormous benefits that exercise training offers in disease prevention and therapy. The aim of this study was to elucidate the transcriptional signatures that distinguish the endurance-trained and untrained muscles in young adult males (24 ± 3.5 years). We characterized baseline differences as well as acute exercise-induced transcriptome responses in vastus lateralis biopsy specimens of endurance-trained athletes (ET; n = 8; VO2max, 67.2 ± 8.9 mL/min/kg) and sedentary healthy volunteers (SED; n = 8; VO2max, 40.3 ± 7.6 mL/min/kg) using microarray technology. A second cohort of SED volunteers (SED-T; n = 10) followed an 8-week endurance training program to assess expression changes of selected marker genes in the course of skeletal muscle adaptation. We deciphered differential baseline signatures that reflected major differences in the oxidative and metabolic capacity of the endurance-trained and untrained muscles. SED-T individuals in the training group displayed an up-regulation of nodal regulators of oxidative adaptation after 3 weeks of training and a significant shift toward the ET signature after 8 weeks. Transcriptome changes provoked by 1 h of intense cycling exercise only poorly overlapped with the genes that constituted the differential baseline signature of ETs and SEDs. Overall, acute exercise-induced transcriptional responses were connected to pathways of contractile, oxidative, and inflammatory stress and revealed a complex and highly regulated framework of interwoven signaling cascades to cope with exercise-provoked homeostatic challenges. While temporal transcriptional programs that were activated in SEDs and ETs were quite similar, the quantitative divergence in the acute response transcriptomes implicated divergent kinetics of gene induction and repression following an acute bout of exercise. Together, our results provide an extensive examination of the transcriptional framework that underlies skeletal muscle plasticity.

1. Introduction

Skeletal muscle is the most abundant tissue in the human body. It gives us posture, protects our bones, and allows our bodies to move. But skeletal muscle is also centrally important in maintaining metabolic and physiological homeostasis. It serves as a regulator of interorgan crosstalk for energy and protein metabolism, exhibits immunobiological properties, and has a profound impact on the central and peripheral nervous system [1,2,3]. To adequately cope with environmental and physiological challenges, skeletal muscle possesses the unique ability to change its size, structural composition, as well as metabolic properties. In this way, specific exercise regimes can be exploited to provoke pronounced adjustments in muscular force, endurance capacity, and/or contractile velocity [4,5]. Functional adaptations are brought about by qualitative and quantitative changes in fiber size and composition that become manifest in a specific assemblage of myofibrillar protein isoforms, a rearrangement of metabolic pathways, and altered Ca2+ handling characteristics [6,7]. Such adaptive remodeling of skeletal muscle arises from the cumulative effect of frequently repeated exercise bouts that acutely challenge systemic and local homeostasis at the metabolic, structural, and inflammatory levels. Surprisingly, the mechanistic framework that links these acute exercise-provoked homeostatic perturbations to long-term training adaptations is largely unknown. Despite much effort in the field, there still is no comprehensive understanding of how single-exercise stimuli should become memorized and how subsequent adaptive reprogramming of myofibrillar gene expression and signaling pathways is accomplished [8,9,10].
Strong epidemiological and emerging clinical evidence shows that habitual physical activity is associated with multiple health outcomes [11]. Understanding the molecular mechanisms that govern skeletal muscle plasticity has the potential to broaden fundamental biological knowledge. This may provide a basis for discovering novel strategies and targets for disease prevention and treatment of dysregulated skeletal muscle physiology in various clinical conditions.
High-throughput methods in the fields of transcriptomics, proteomics, and metabolomics provide valuable data compilations to gain insight into the complex signaling pathways that coordinately act to initiate and shape metabolic and structural remodeling in human skeletal muscle in response to acute exercise and long-term training stimulation [12,13,14,15,16,17,18,19,20,21,22,23]. Using a label-free quantitative proteomic approach, our groups could recently decipher various metabolically active proteins with training status-dependent abundances and differential acute response patterns in human vastus lateralis muscle [21]. Likewise, through the use of a multi-analyte profiling strategy, we could provide a list of differentially affected circulating mediators with diverse metabolic and immunological properties in plasma samples of endurance-trained and sedentary individuals in response to intensive cycling exercise [24]. To extend these results to the transcriptional level, we further exploited these study protocols to determine the general impact of strength and endurance exercise on skeletal muscle transcriptome profiles [25].
Here, we now provide an in-depth analysis of training status-dependent gene expression signatures in vastus lateralis muscle from young endurance-trained (ET) and untrained sedentary individuals (SED) under resting conditions as well as in response to acute exercise. To interpret the biological relevance of observed differential gene expression patterns, microarray data were analyzed for functional and regulatory gene signatures. Expression levels of selected target genes were further exploited to assess the adaptational changes of the untrained muscle in the course of a standardized 8-week endurance training intervention.

2. Results

2.1. Study Design

Healthy, young male volunteers were recruited in this study and categorized based on their physical activity history and VO2max levels into untrained sedentary individuals (SED; n = 8) and endurance-trained individuals (ET; n = 8) (see the Section 4 for details). All study participants followed an acute exercise protocol that consisted of 60 min of high-intensity cycling exercise on a cycle ergometer at a main power requiring 80% of the individual VO2max (including 10 min of initial warm-up cycling). Baseline microbiopsy samples of the vastus lateralis muscle were obtained 24 h before the acute exercise protocol was started. Post-exercise biopsies were taken at 30 min and at 3 h after cessation of acute cycling exercise (Figure 1).
The anthropometric and performance data for the participants in the acute exercise setup are shown in Table 1.

2.2. Differential Gene Expression Signatures between the Trained and Untrained Skeletal Muscle at Rest

To compare the transcriptomic landscape of the trained and untrained muscle, baseline microbiopsy samples from eight SED individuals and eight ET individuals were used for microarray gene expression profiling. By applying a fold change cut-off of |≥| 1.5 (p < 0.05), our microarray analysis identified a total number of 214 genes with differential baseline expression levels between the SED group and the ET group (Figure 2, Table S1).
Among them, 127 genes displayed increased expression in the SED group, while 87 genes showed higher relative transcript levels in ET individuals (Figure 2B). Hierarchical clustering of differentially expressed probe sets showed that this gene signature distinguished between SED and ET individuals (Figure 2A). The vastus lateralis muscle is commonly characterized by a mixed fiber type composition containing both type I and type II fibers at almost equal levels in healthy, untrained individuals [26]. GO term enrichment analysis revealed that differentially expressed genes at rest were primarily attributable to cellular components and functions involved in myofibrillar structure and activity (Figure 2C). A literature search for experimentally verified fiber-type specific expression profiles within these enriched gene sets revealed a preferential up-regulation of slow-twitch (type I) muscle candidate genes in the ET group, while SED individuals showed a tendency toward higher expression of genes predominantly attributable to fast-twitch (type II) muscle fibers (Table 2). The same pattern appeared to account for differentially expressed genes involved in key metabolic pathways of substrate breakdown, with relatively higher expression levels for genes encoding glycolytic enzymes in the SED group and up-regulated oxidative metabolism genes in the ET group (Figure 3A,B).
Candidate upstream regulators that are predictively involved in determining the SED muscle phenotype included the beta subunit of hypoxia-inducible factors (HIFs) ARNT2 (aryl hydrocarbon receptor nuclear translocator 2), as well as the transcription factor (TFs) early growth response 2 (EGR2), zinc finger protein GLI1 (glioma-associated oncogene homolog 1), and class E basic helix–loop–helix protein SIM1 (single-minded family BHLH transcription factor 1) (Figure 3C). In ET muscles, we found activation signatures for upstream regulatory influence by pro-angiogenic factors, including vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF), as well as the pro-inflammatory cytokine interferon (IFN)-gamma (Figure 3D). Stimulated TF activity in the ET group was predicted for several well-known regulators of endurance exercise-dependent gene expression, including the peroxisome proliferator-activated receptor γ (PPARG), the tumor suppressor protein p53 (TP53), and the estrogen-related receptor α (ESRRA). Moreover, we also found predicted activation of candidate upstream regulators that are involved in tumor necrosis factor receptor (TNF-R)-associated factor (TRAF) signaling, namely interferon regulatory factor 7 (IRF7) and nuclear factor-kappa B (NF-κB). Significantly up-regulated targets of the NF-κB signature in the ET muscle included caspase 4 (CASP4), the immunoglobulin kappa light chain (IGKC), calcium-transporting ATPase (ATP2A2), the tryptophan catabolizing enzyme indoleamine 2,3-dioxygenase (IDO1), the kinase insert domain receptor (KDR) of VEGF, the Ca2+/calmodulin-dependent protein kinase Pim-3 (PIM3), as well as the tumor necrosis factor superfamily member TRAIL (TNFSF10).

2.3. Gene Expression Responses of the Trained and Untrained Skeletal Muscle Following an Acute Bout of Endurance Exercise

To examine the impact of an acute bout of endurance exercise on skeletal muscle gene expression, all ET and SED individuals underwent 60 min of strenuous cycling exercise at a main intensity corresponding to 80% of the individual VO2max (including 10 min of warm-up). For both groups, this individually adjusted exercise protocol provoked comparable increases in blood lactate levels and heart rate responses (Table 1). Microbiopsy samples of the vastus lateralis muscle were taken at rest, 30 min (+30′) after completion of exercise, and after a recovery period of 3 h (+3 h). Overall, we found a total number of 759 genes that significantly reacted in response to exercise (fold change vs. baseline |≥| 2, p < 0.05), with 614 genes being up-regulated and 145 genes becoming down-regulated (Figure 4A–E; Table S1). Exercise response patterns included 600 genes (478 up, 122 down) at 30 min post exercise and 491 genes (435 up, 56 down) after 3 h of recovery.
IPA pathway analysis revealed prominent gene signatures connected to pathways of acute contractile, metabolic, oxidative, and inflammatory stress responses in both groups at both post-exercise sampling points (Figure 5A,B). Remarkably, at +30′, the untrained muscle of the SED individuals displayed higher activation scores for the nuclear receptors liver X receptor (LXR) and farnesoid X receptor (FXR), which are known to be involved in balancing lipid and glucose metabolism [40].
Potential upstream regulators positively involved in acute exercise-dependent gene regulation included several signal transducers and TFs commonly associated both with inflammatory (signal transducer and activator of transcription 1, STAT1; STAT3; STAT4; NFκB; NFκB Subunit, RELA; progesterone receptor, PGR) and regenerative processes (cAMP responsive element binding protein 1, CREB1; SMAD family member 3, SMAD3; catenin beta 1, CTNNB1), as well as with oxidative stress balance (HIF1A; endothelial PAS domain protein 1, EPAS1 (HIF2A); nuclear factor of activated T-cells, NFAT) (Figure 5C,D). Overall, acute response patterns of the trained and untrained muscles displayed considerable quantitative divergence, with only 308 genes being significantly affected at +30′ both in the ET and the SED group, and 238 genes being included in both response signatures at +3 h (fold change vs. baseline |≥| 2, p < 0.05).
Significant enriched GO terms for molecular functions associated with higher exercise-provoked responsiveness in the SED group (fold difference SED/ET ≥ 1.5, p < 0.05) included ‘transcriptional activator activity’ (p = 5.01 × 10−7) and ‘steroid hormone receptor activity’ (p = 1.78 × 10−4), with genes encoding early growth response 1 (EGR1), HIF1A, nuclear receptor subfamily 4 group A member 1 (NR4A1) and member 3 (NR4A3), AP-1 transcription factor subunit (FOSL1), abhydrolase domain-containing protein 2 (ABHD2), and retinoic acid receptor alpha (RARA). From the lists of exercise-affected stress signaling genes, we found six genes displaying training status-dependent differential response patterns, all of them with increased responsiveness in the untrained skeletal muscle. These genes included the transcriptional repressor BACH1 as well as the transcriptional activator of the small MAF protein family MAFF, the angiotensin precursor AGT, and the heat shock protein family members DNAJB4, DNAJC1, and HSP8. In contrast, functional categories enriched in acute exercise-affected genes with higher responsiveness in the ET group (fold difference ET/SED ≥ 1.5, p < 0.05) were attributable to GO terms ‘phosphatidylinositol-3-kinase activity’ (p = 3.41 × 10−3) and ‘insulin receptor binding’ (p = 1.35 × 10−5), with prominent genes encoding insulin receptor substrate 2 (IRS2), as well as regulatory subunits 1 (PIK3R1) and 3 (PIK3R3) of phosphoinositide-3-kinase.
In total, differential expression levels between the SED group and the ET group could be observed for 188 genes at time point +30′ and for 268 genes at +3 h, with an overlap of 63 genes being included in both differential expression signatures. Considering all differentially expressed genes from both post-exercise sampling points, the most highly enriched GO term in the ET group was ‘mitochondrial matrix’ (p = 6.73 × 10−20), while in the SED group, we identified increased expression of genes primarily enriched in GO terms ‘cell growth’ (p = 2.03 × 10−11) and ‘cell proliferation’ (p = 1.05 × 10−10).
From the list of the 214 genes with differential baseline expression between the SED group and the ET group, 65 genes were also detected being differentially expressed at +30′, and 78 of these genes displayed differential expression levels at +3 h (fold difference |≥| 1.5, p < 0.05). To determine the most stable signature genes of the differential baseline profile, we performed cluster analysis and identified a set of 39 exercise-resilient genes (Figure 6). This stable training status-dependent profile included several well-known candidates that determine skeletal muscle architecture, contraction pattern, and metabolism, like, e.g., genes coding for muscle contractile proteins, lipoprotein lipase (LPL), LDH isoforms, or UCP3. In addition, we found some unexpected targets that are yet poorly defined or have not been addressed so far in connection with phenotypic skeletal muscle adaptation pattern, like the genes encoding shisa family member 2 (SHISA2), dysbindin domain containing 1 (DBNDD1), nanos C2HC-type zinc finger 1 (NANOS1), transmembrane protein 56 (TMEM56), cell growth-inhibiting protein 39 (FAM129A), or signal peptide CUB domain (SCUBE2).
In contrast to this stable gene signature, 35 genes from the differential resting expression pattern became significantly affected by the acute exercise intervention (fold change vs. baseline |≥| 2, p < 0.05). A list of these genes, including a detailed illustration of how they react to the exercise intervention, is given in the Supplementary Materials (Table S2). These genes obviously become epigenetically poised in the course of endurance training and represent flexible key signaling nodes of skeletal muscle plasticity that contribute both to the maintenance of long-term adaptation patterns as well as to the short-term adaptational response to exercise.

2.4. Expression Pattern of Selected Target Genes in the Untrained Skeletal Muscle in the Course of a Standardized Eight-Week Endurance Training Intervention

To provide independent validation of the microarray data, we included an additional cohort of 10 untrained individuals (SED-T) to absolve the 60 min cycling exercise protocol. Microbiopsy samples of the vastus lateralis muscle were taken at rest and at 3 h (+3 h) after completion of exercise. For confirmation of exercise-affected expression pattern by qPCR, we selected a subset of 15 candidate genes comprising targets with pronounced and moderate responsiveness, as well as targets that were considered unresponsive (fold change |≤| 2) to acute exercise. Selected candidate genes included NR4A3, interleukin 6 (IL6) and interleukin 6 receptor (IL6R), ZFP36 ring finger protein (ZFP36), TNF receptor superfamily member 12A (TNFRSF12A), tripartite motif-containing protein 63 (TRIM63), F-box protein 32 (FBXO32), actin-binding Rho-activating protein (ABRA), peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PPARGC1A), vascular endothelial growth factor A (VEGFA), myostatin (MSTN), actinin alpha 3 (ACTN3), lactate dehydrogenase isoforms A (LDHA) and B (LDHB), and lipoprotein lipase (LPL). As shown in the Supplementary Materials (Figure S1), for 14 genes, comparisons between microarray data and qPCR analysis in the independent data set were in close agreement. In contrast to the microarray data set, the acute exercise effect on ABRA expression was not consistent in the independent SED-T cohort. However, it should be noted that indicated increases in the microarray cohort of SED individuals were below the fold change threshold of |≥| 2 that was applied to filter differential expression levels.
Following the acute exercise testing, all SED-T volunteers followed a standardized 8-week endurance training program, as described in detail in the Section 4. This training routine led to significant improvements in cardiorespiratory fitness. The anthropometric and performance data for the participants in the training intervention program are shown in Table 3.
Basal expression profiles of vastus lateralis muscle for 11 target genes were analyzed by qPCR after 3 weeks (3 wks) of training as well as at the end of the training session (8 wks) and compared to pre-training baseline levels (rest) (Figure 7A,B). Selected genes included long-term-adaptation markers (ABRA, ACTN3, LDHA, LDHB, LPL, MSTN) as well as master regulators of oxidative energy metabolism (PARGC1A and VEGFA) and known markers of skeletal muscle atrophy (FBXO32, TNFRSF12A, TRIM63). To assess to what extent the untrained muscle became shifted toward an endurance-trained phenotype by the 8-week training intervention, we compared training-provoked changes in baseline expression levels with the differential baseline expression profile of ET and SED individuals from our microarray data (Figure 7C). This way, we aimed to decipher the short-term and long-term effects of endurance training on skeletal muscle expression patterns. As shown in Figure 7A, after 3 weeks of training, for most target genes, we observed only minor or inconsistent changes in basal transcriptional expression. However, we found a significant transcriptional up-regulation of the key metabolic regulator PPARGC1A gene as well as of the angiogenic marker VEGFA, indicating the initiation of training-induced metabolic and vascular adaptation programs. After 8 weeks of endurance exercise training, PPARGC1A transcript levels had ceased to baseline expression, whereas the differential expression markers ABRA, ACTN3, LDHB, and MSTN had approached the endurance-trained expression profile (Figure 7B). We further observed a significant reduction in transcript levels for the atrophy-related marker genes FBXO32 and TRIM63. In contrast, robust alteration of baseline transcriptional activity for the metabolic markers LDHA and LPL was not yet manifested by 8 weeks of training.

3. Discussion

In the present study, we provide a comprehensive list of differentially expressed genes that distinguish the basal transcriptome pattern of human vastus lateralis muscle from healthy young ET and SED individuals. Most of the differentially expressed transcripts encode proteins that mediate muscle contraction and force production, ion homeostasis, as well as metabolic substrate utilization and oxidation, which mirror the major functional differences between slow- and fast-twitch muscle fibers. In line with our recent in-depth analysis of training-induced proteomic patterns in vastus lateralis muscle [21], we identified transcriptional signatures that reflect an improved oxidative capacity of the endurance-trained skeletal muscle. Approximately 55% of the genes that we found differentially expressed in the basal skeletal muscle transcriptome of our young ETs have also been reported by Lindholm et al. to become significantly affected in young sedentary individuals following 3 months of endurance training intervention [41], and about 30% were also found differentially expressed in middle-aged men and women as a result of decades-long training in a recent cross-sectional study by Chapman et al. [42]. Adaptations in metabolic substrate flux can exemplarily be illustrated by a shift in the LDH isoenzymes expression pattern that was consistently observed in all three studies. Basal down-regulation of LDHA and up-regulation of LDHB in the course of endurance training indicate an increased lactate threshold, augmented mitochondrial pyruvate fueling, and enhanced mitochondrial respiration [43]. Remodeling of the tetrameric LDH complex has previously been shown to be highly correlated with exercise performance and skeletal muscle oxidative capacity [44,45]. Intriguingly, we could show that 8 weeks of regular endurance exercise did already suffice to modulate skeletal muscle LDH isoform baseline expression, with LDHB becoming significantly up-regulated at the end of the training intervention.
It has been demonstrated that the transcriptional coactivator PGC-1α (PPARGC1A), a nodal regulator of mitochondrial biogenesis [46], is required for elevated LDHB transcription [44,47], whereas the LDHA promoter region contains the consensus sequences for, and is known to become regulated by, the TF HIF1α [48]. Consistently, in the skeletal muscle of SED individuals, we identified a signature of enhanced baseline upstream regulator activity for the nuclear translocator ARNT2 and the basic helix–loop–helix TF SIM1. ARNT2 forms functional heterodimers with HIF1α and with SIM1 to drive, i.e., enhanced transcription of glycolytic enzymes, like PGK1 and LDHA [49], as well as expression of epidermal growth factor receptor (EGFR) and its ligand EGF [50,51], as observed in the untrained muscle of SED individuals.
EGF-EGFR signaling has recently emerged as an important and highly conserved regulatory pathway of fast-twitch skeletal muscle fiber type determination, and reciprocally, EGFR blockade both in vitro and in vivo appears as a suitable tool to promote a slow-twitch muscle phenotype [52]. Transient EGFR signaling, however, is also crucially important for skeletal muscle regeneration, and redox regulation of EGFR offers protection against oxidative stress and provides increased resistance to hypoxia [53,54]. EGF may also contribute to the expression of UCP3 [55], a mitochondrial inner membrane transport protein that has been subject to much controversial debate in connection with skeletal muscle physiology. UCP3 belongs to the UCP subfamily of mitochondrial anion carrier proteins and is commonly considered to play important roles in oxidative stress resistance and fatty acid metabolism; however, its mechanisms of action as well as its physiological role in the exercising muscle are still enigmatic [56,57]. In line with our data, a suppressive effect of endurance training on skeletal muscle UCP3 expression has been described by Russell et al., with a more pronounced impact on type I fibers [58], and UCP3 content of vastus lateralis muscle has been proposed to reversely correlate with cycling efficiency in trained as well as untrained individuals [59]. On the other hand, overexpression of UCP3 in muscles of mice offers protection against high-fat diet-induced insulin resistance [60], increases fatty acid oxidation, reduces oxidative stress levels, and augments the effects of endurance training on whole-body energy expenditure and fiber type characteristics [61]. Mouse studies that are based on large genetic or environmental perturbations, such as knockout mice, overexpression models, or extremely high cholesterol diets, are invaluable to understanding the basic properties of isolated key signaling modules but do not necessarily allow an adequate understanding of how these factors are integrated into distinct networks under physiological conditions. Moreover, it can lead to mistaken conclusions if singular factors with pleiotropic functions are attributed with beneficial or detrimental characteristics.
One prominent representative is the TF NF-κB, which is commonly considered a “bad guy” in muscle physiology and is known to mediate severe muscle wasting in age and disease [62]. However, activation of NF-κB signaling via the alternative, or non-canonical, pathway has also been demonstrated to trigger mitochondrial biogenesis and to improve the metabolic capacity of skeletal muscle [63,64]. In the baseline differential expression pattern from ET individuals, we not only detected activation signatures for commonly approved transcriptional mediators of skeletal muscle endurance phenotype, like PPARG, TP53, and ESRRA [13,65,66,67,68], but also found indications for enhanced NF-κB activity. Although we cannot distinguish whether this reflects canonical or non-canonical signaling, increased expression of the anti-inflammatory enzyme IDO may hint at the latter [69]. Moreover, the non-canonical NF-κB pathway is known to become activated through particular TNF receptor family members [70], like the sheddable type II transmembrane protein TRAIL (TNFSF10) that we found being transcriptionally elevated in the ET muscle at rest. Again, TRAIL also seemingly shows a Janus-faced nature as, on the one side, it has been linked to muscle fiber damage in conditions of inflammatory myopathies [71] but also has been shown capable of reducing inflammation, improving insulin responsiveness, and attenuating metabolic abnormalities in diabetes [72,73].
While the precise contribution of NF-κB signaling to the baseline gene expression pattern remains elusive, in the exercising muscle, we clearly could depict NF-κB together with the TF HIF1α as being among the major drivers of the acute exercise-provoked transcriptional stress response, both in SED as well as in ET individuals, as illustrated by a significant enrichment of related pathways in the exercise-provoked gene signatures. Both TFs have previously been shown to become activated in skeletal muscle during acute exercise, accumulate in the nucleus, and associate with chromatin at their target sites [51,74,75]. Moreover, there is increasing evidence that these two signaling pathways are intimately linked and coordinately act together in shaping exercise-provoked myofibrillar stress signaling to cope with acute shortages in energy and oxygen supplies, accumulation of reduced oxygen intermediates, perturbations in intracellular calcium levels, enforced mechanical strain at the contractile apparatus, and increased release of stress hormones [75,76,77]. In the exercising muscle, NF-κB and HIF1α cooperate with multiple signaling molecules and pathways [75,78]. Nodes of crosstalk are mediated by other transcriptional regulators for which we found enriched downstream target signatures within the acute exercise-provoked expression patterns, such as members of the STAT family of TFs, the cAMP-response element binding protein (CREB), and the β-catenin signaling cascade (CTNNB1). These transcriptional regulators either directly interact with HIF and NF-κB subunits or affect the expression of common stress-responsive target genes [79,80,81].
The most prominent class of molecules that act as initiators of the acute exercise-provoked stress response are reactive oxygen species (ROS). Physiological triggering of ROS by exercise has been shown essential for normal skeletal muscle contractility and subsequent adaptation processes, while, on the other hand, excess or chronic exposure to ROS is detrimentally implicated in conditions of skeletal muscle wasting induced by disuse, aging, or disease [82,83]. Oxidative stress initiates cellular defense mechanisms that centrally involve the antioxidant-response-element signaling pathway via activation of nuclear factor erythroid 2-related factor 2 (NRF2), a TF that controls genes encoding protective antioxidants and phase II detoxification enzymes [84]. Consistently, we detected increased exercise-induced expression of NRF2 target genes, including mitochondrial superoxide dismutase (SOD2), which converts highly cytotoxic superoxide radicals to hydrogen peroxide, and heme oxygenase-1 (HMOX1), which provides bilirubin to scavenge and degrade hydrogen peroxide. Moreover, we observed acute transcriptional induction of inhibitor of differentiation (ID) proteins (ID1, ID3), a class of transcriptional regulators that have recently been found to promote antioxidant gene expression through the regulation of small MAF protein expression [85]. While increased expression of IDs was restricted to the acute exercise-induced transcriptional response, with similar profiles in ET and SED individuals at 30 min post exercise, up-regulated expression of the small MAF transcript MAFF was prolonged until 3 h post exercise, with significantly higher levels in the SED group. Small MAF proteins are capable of forming repressive homodimers as well as transcriptionally active MAF-NRF2 heterodimers that are required for efficient NRF2 binding to its target promoter sites [86]. Reciprocally, small MAF proteins can also heterodimerize with the TF BACH1, a negative regulator of antioxidant-response-element-mediated gene expression, thereby promoting active resolution of the oxidative stress response. Also of interest is that the transcriptional expression of BACH1 in the vastus lateralis muscle became acutely triggered by our exercise protocol and remained elevated after 3 h of recovery. As a general theme in acute stress responses, simultaneous co-triggering of self-regulatory gene expression networks is a necessary means for rapid and flexible adjustment of cellular homeostasis to ensure proper modulation as well as timely resolution of, in the long-term, potentially harmful stress-provoked signaling cascades [87].
A primary limitation associated with the current study is that our post-exercise samples constitute a compilation of individual “snapshots” from very dynamic gene expression processes with response kinetics that may individually vary not only as a result of the training history but also due to numerous genetic and environmental factors. Despite significant quantitative differences in transcript numbers and expression intensities, the transcriptome programs activated by the intense exercise bout appeared broadly similar among ETs and SEDs, and we could not detect a clear training status-dependent theme in the acute exercise response patterns. Largely similar acute transcriptional responses have also been reported by Rubenstein et al. when muscle samples from active/endurance-trained and sedentary older adults were compared after 40 min of submaximal cycling [88]. In contrast, a substantial quantitative as well as qualitative effect of the training status on the acute exercise-provoked transcriptional response was demonstrated by Furrer et al. when quadriceps muscles of mice that had undergone 4 weeks of endurance training were compared with sedentary controls [46]. The authors noted that training status-dependent shifts in peak expression have a major impact on post-exercise transcriptomes at a given timepoint, indicating that it is rather the kinetic pattern of gene induction and repression than differences in specific gene expression signatures that distinguish the trained from the untrained muscle in its acute transcriptional response to exercise. Necessarily, endurance exercise training studies in mice cannot be directly translated to humans due to profound species-specific differences in evolutionary background, morphometry, physiology, life history, and skeletal muscle fiber composition and adaptation potential. However, in congruence with Furrer et al. [46], we observed that acute exercise-responsive genes only poorly overlapped with the persistent transcriptomic changes in the trained muscle, mainly affecting genes that were up-regulated in the untrained baseline profile. Apparently, the basal transcriptomic output that characterizes the trained muscle is only a final echo of the epigenetic alterations that become imprinted in the course of prolonged endurance training [13,41,46,89], where probably each bout of exercise is remembered by distinct chromatin marks, altered DNA methylation patterns, changes in spatial chromatin structure, and/or diverse non-coding RNA species [8]. In this way, gene loci that shape and determine skeletal muscle plasticity can reversibly be altered to reside either in an active, poised, bivalent, or suppressed state. Not surprisingly, most stably up-regulated or down-regulated genes distinctly distinguished the trained from the untrained muscle, irrespective of the muscular activation status, encoded structural proteins of the contractile apparatus, membrane transport proteins, and metabolic enzymes with known fiber-type preferences. In addition, we also found some unexpected marker genes that are yet poorly defined. Here, SHISA2, an endoplasmic reticulum (ER)-localized protein with up-regulated expression in the untrained muscle, is of particular interest as it has recently been shown to regulate myogenesis and satellite cell fusion [90].
From the many factors that have been implied in exercise adaptation, the transcriptional coactivator PGC-1α (encoded by the gene PPARGC1A) has been shown indispensable for normal transcriptional muscle plasticity, both in controlling basic pathways of the acute stress response as well as in shaping the physiological long-term training response [7,10,46]. In our training study, after 3 weeks of regular endurance exercise, we observed a significant up-regulation of PPARGC1A, alongside increased expression of the angiogenic factor VEGFA, that preceded the remodeling of the basal transcriptome landscape toward the ET phenotype, which became apparent after 8 weeks. This supports the model that muscular adaptation to endurance training is not altogether a gradual process but rather relies on an adaptive threshold system that allows constant assessment of whether the benefits to blunt the homeostatic threats imposed by frequently repeated exercise bouts outweigh the costs of structural and biochemical remodeling of skeletal muscle architecture and metabolism. Most importantly, it cannot be emphasized enough that three training sessions per week over only 2 months suffice not only to significantly improve the overall cardiorespiratory fitness of untrained individuals but also to substantially shift the skeletal muscle transcriptome toward an endurance phenotype and to significantly diminish the expression of atrophy-related marker genes.
Findings from the present study should be interpreted with the following considerations in mind. The skeletal muscle biopsy samples analyzed in this study only represent a small part of a mixed-fiber type muscle and also include varying amounts of other cell types. We also recognize that expression levels between transcript and protein are not necessarily correlated. This study lacked a parallel proteome analysis and a quantitative characterization of fiber type composition. Finally, as the current study involved only small cohorts of young, healthy males, our findings should be verified in other populations using larger sample sizes.
In summary, we provide an extensive examination of the transcriptional patterns that distinguish the endurance-trained muscle from the untrained muscle in young adult males, both at baseline as well as in response to an acute exercise challenge. We aimed to predict genes and pathways that are important to determine and maintain skeletal muscle phenotype and metabolism. Our data offer a glimpse at the fascinating complexity of the multifaceted mechanistic framework that allows our muscles to cope with the homeostatic challenges of acute exercise and to remodel their transcriptional landscape in the course of prolonged exercise training. A better understanding of the cellular and molecular mechanisms involved in exercise adaptation is fundamentally important to take full advantage of the enormous benefits that exercise training offers in disease prevention and therapy.

4. Materials and Methods

4.1. Study Participants and Phenotyping

Healthy young males were recruited and classified based on their self-reported physical activity as described in detail previously [21]. To meet the inclusion criteria for the endurance-trained (ET) group, participants had to engage in regular endurance exercise training of >5 h per week for at least 5 years. The sedentary group (SED) comprised subjects that habitually spent less than 2 h per week with moderate physical activity. Potential volunteers underwent preliminary baseline screening, including lactate diagnostics and maximal bicycle spiroergometry. Participants undertook a graded exercise test to volitional exhaustion on a cycle ergometer (Ergoselect 200, Ergoline GmbH, Bitz, Germany) for the determination of maximal oxygen uptake (VO2max), peak power output, and the lactate thresholds [lactate turning point 1 (LTP1) and lactate turning point 2 (LTP2)]. Before starting the test, resting blood pressure and capillary blood lactate concentrations [La−] were measured. The test began with a 2 min resting period on the bike, followed by 25 W step increments every 3 min, starting at 50 W until task failure. [La−] was analyzed (Biosen S-Line, EKF diagnostics, Barleben, Germany) by collecting capillary blood samples from the right earlobe during the last 20 s of each stage and right after volitional exhaustion. Blood pressure was taken again at 100 W and immediately after volitional exhaustion. Heart rate and electrocardiogram were constantly recorded throughout the test (12-channel PC ECG, Custo med GmbH, Ottobrunn, Germany). Breath-by-breath pulmonary gas exchange and ventilation were measured using a metabolic cart (MetaLyzer, CORTEX Biophysics GmbH, Leipzig, Germany). Calibration was performed before each test, following the manufacturer’s instructions. The course of the VO2 was interpolated using a spline procedure and smoothed. The plateau phase, which occurs when the O2 kinetics level off, was used as a criterion for determining the VO2max. Breath-by-breath data were averaged over 20 s. The power (in Watt) performed at the time of 100%, 80%, and 60% VO2max was recorded. To be included in this study, ET subjects had to exhibit an aerobic capacity of >57 mL/min/kg BW, while the VO2max upper limit for the SED group was set at 49 mL/min/kg BW. The study protocol was approved by the local ethics committees of the University of Tübingen, the Justus-Liebig University Giessen, and the University of Ulm. Each participant underwent a medical screening and had to give informed written consent.

4.2. Acute Exercise Setup and Training Intervention

Within 4 weeks from the initial performance diagnostic, the study participants followed the acute exercise protocol, which consisted of 60 min of high-intensity cycling exercise on a cycle ergometer (Ergometrics 800 S, Ergoline) at a main power requiring 80% of the individual VO2max (including 10 min of initial warm-up cycling at 60% VO2max) (Figure 1). All exercise sessions were conducted in the morning (9:00 a.m.) after an overnight fast and after receiving a standardized breakfast (8:00 a.m.).
In a follow-up study, additional SED volunteers (SED-T) were recruited to absolve an 8-week training program that consisted of 3 supervised endurance exercise sessions per week on a cycle ergometer. Before and after the training period, all participants underwent lactate diagnostics and maximal spiroergometry to determine individual anaerobic threshold (IAT) and VO2max. Exercise training intensity was monitored by heart rate (HR) and manually adjusted by the cycle ergometer load. The training protocol consisted of 15 min warm-up followed by 65 min of continuous cycling in the target HR range. Training intensity was progressively increased every 2 weeks, starting with a power output corresponding to 60% of the individual VO2max, followed by 65%, 70%, and finally 75% in weeks 7 and 8.
The anthropometric and performance data for the participants in the acute exercise setup and in the training intervention program are shown in Table 1 and Table 3, respectively.

4.3. Muscle Biopsies and Microarray Analysis

All muscle biopsies were taken under sterile conditions from the lateral portion of the m. vastus lateralis of the right leg using a fine-needle punch biopsy technique (Plus Speed; Peter Pflugbeil, Zorneding, Germany) under local anesthesia (1% Meaverin). Baseline samples of the resting muscle were obtained 24 h before the acute exercise protocol was started. Post-exercise biopsies were taken at 30 min and at 3 h after cessation of acute cycling exercise (Figure 1). Snap-frozen muscle biopsy samples were homogenized with a TissueRuptor homogenizer (Qiagen, Hilden, Germany), and total RNA was extracted and purified using Rneasy Fibrous Tissue Mini kit and Rneasy Micro Kit (Qiagen), according to the manufacturer’s recommendations. RNA integrity was assessed with Agilent 2100 Bioanalyzer (Agilent Technologies, Waldbronn, Germany), and only high-quality RNA (RNA integrity number > 7) was submitted to RNA profiling using Human Genome U219 (HG-U219) microarray platform (Affymetrix, Santa Clara, CA, USA). Preparation of biotin-labeled complementary RNA probes, hybridization, and scanning of arrays was carried out according to the manufacturer’s protocols by the Microarray Facility Tübingen (MFT Services, Tübingen, Germany). Scanned images (DAT files) were transformed into intensities (CEL files) by GCOS (Affymetrix). The raw intensity values were background corrected, log2 transformed, quartile normalized, and summarized using the RMA algorithm in Genomics Suite 6.6 (Partek). The data are deposited in the NCBI Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/) and are accessible through GEO Series accession number GSE250122. Differentially expressed genes were determined using the package “limma” (version 3.24.14), which analyzes the expression data by fitting linear models and determines statistical significance with moderated t-statistics [91]. The significance threshold for differential expression at baseline and after exercise intervention between and within groups was set to a minimum log2 fold change of 0.6 (fold change |≥| 1.5) for unpaired samples and a minimum absolute log2 fold change of 1 (fold change |≥| 2) for paired samples together with p < 0.05. Principal component analysis (PCA) scatter plots were used to visualize patterns in the gene expression data (Figure S2). Full lists of differentially expressed probe sets are provided in the Supplementary Materials (Table S1). Enriched gene ontology (GO) terms in differentially expressed genes were identified using InCroMap [92]. Differentially expressed genes were further analyzed for significant enrichment in affected targets of known upstream regulators (Fisher’s exact test) by use of IPA® Upstream Regulator Analysis (IPA; Ingenuity Systems, Redwood City, CA, USA). Heatmaps and dendrograms were created with JMP statistical software (Version 11; SAS Institute, Cary, NC, USA) by hierarchical cluster analysis of normalized log2 expression intensities using Ward’s method. Volcano plots were applied to visualize fold changes and t-test values of the comparisons between exercise groups and time points.

4.4. Quantitative RT-PCR Analysis

Microarray data were further validated in the independent cohort of sedentary individuals (SED-T) that followed the acute cycling protocol prior to participating in the 8-week training program. Skeletal muscle biopsies were analyzed by quantitative RT-PCR (qPCR) at baseline, 3 h after acute exercise, during (3 wks) the training program, and 2 days after the last training session (8 wks). Frozen biopsy samples were homogenized (4000 rpm, 2× 30 s) with Precellys beads (VWR, Darmstadt, Germany) in TRIzol reagent (ThermoFisher Scientific, Waltham, MA, USA) using a BeadBug™ microtube homogenizer (Benchmark Scientific, Inc., Edison, NJ, USA), and total RNA was isolated according to the manufacturer’s protocol. RNA was further purified using the RNeasy Micro Kit (Qiagen) as per manufacturer’s instructions, including an on-column DNase treatment step. First-strand cDNA was synthesized from 0.3 µg of total RNA, using random primers and M-MLV reverse transcriptase RNase H Minus, Point Mutant (Promega, Mannheim, Germany). Quantitative RT-PCR was performed from diluted cDNA (1:10) on an iCycler single-color detection system (BioRad, München, Germany) with SsoFast EvaGreen Supermix (BioRad) in a total reaction volume of 15 μL. Detailed information for each target-specific primer pair is provided in the Supplementary Materials (Table S3). Primers were used at a final concentration of 0.3 µM. All samples were run in duplicate. PCR was performed for 40 cycles at 95 °C for 5 s and at 59 °C for 15 s after initial incubation at 95 °C for 30 s. PCR product specificity was confirmed by melting curve analysis. Five candidate reference genes (TATA-box binding protein, TBP; ribosomal protein L13a, RPL13A; large ribosomal protein P0, RPLP0; beta-2-microglobulin, B2M; glyceraldehyde-3-phosphate dehydrogenase, GAPDH) were analyzed for most stable expression pattern using RefFinder [93]. The geometric mean of the three most stably expressed reference genes (TBP, RPL13A, RPLPO) was applied to transform threshold cycles of target genes into normalized expression values using the delta Ct method. Fold differences were calculated using the delta-delta Ct method [94].

4.5. Statistical Analysis

Physiological parameters, qPCR data, and subsets of microarray data were analyzed using JMP statistical software (Version 11; SAS Institute, Cary, NC, USA). Differences in subject characteristics and physical performance outcomes between and within study groups were determined via paired and unpaired t-tests. Relative gene expression levels and fold changes were log2 transformed. Repeated-measurement analysis of variance was used to compare differences regarding changes in expression levels at different timepoints. p-values were corrected for multiple testing using the Benjamini–Hochberg method. Data were considered significant with p-values of less than 0.05.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms25052881/s1.

Author Contributions

Study conception and design: J.M.S., F.C.M., K.K. and A.M.N.; data collection: T.B., M.Z., J.H., M.S., A.F. and C.B.; analysis and interpretation of results: T.B., M.Z., M.S., J.H. and A.F.; draft manuscript preparation: T.B. and A.M.N. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant from the German Federal Institute of Sports Science (Bundesinstitut für Sportwissenschaft; BISp), Germany (FKZ 2509BI1902, AZ 081902/09-13). We further acknowledge support from the Open Access Publishing Fund of the University of Tübingen.

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki and approved by the local ethics committees of the University of Tübingen (039/12; approval 23 February 2012), the Justus-Liebig University Giessen (204/11; approval 9 November 2011), and the University of Ulm (267/11; approval 7 November 2011).

Informed Consent Statement

Informed consent was obtained from all subjects involved in this study.

Data Availability Statement

Microarray data are deposited in the NCBI Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/) and are accessible through GEO Series accession number GSE250122.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

ACSS1, acyl-CoA synthetase short-chain family member 1; ARNT2, aryl hydrocarbon receptor nuclear translocator 2; ASL, argininosuccinate lyase; CD36, fatty acid translocase; CKB, creatine kinase brain; EGR2, early growth response 2; ESRRA, estrogen-related receptor alpha; FGF2, fibroblast growth factor 2; GLI1, glioma-associated oncogene family zinc finger 1; GLUT4, glucose transporter type 4; GPD1, glycerol-3-phosphate dehydrogenase 1 (soluble); GPD2, glycerol-3-phosphate dehydrogenase 2 (mitochondrial); GPT2, glutamic pyruvate transaminase; IFNG, interferon gamma; IRF7, interferon regulatory factor 7; LDHA, lactate dehydrogenase A; LDHB, lactate dehydrogenase B; LPL, lipoprotein lipase; MCT2 (SLC16A7), monocarboxylate monocarboxylic acid transporter 2; MCT4 (SLC16A4), monocarboxylate monocarboxylic acid transporter 4; MPC1 (BRP44L), mitochondrial pyruvate carrier 1; NFκB, nuclear factor kappa B (complex); PDHB, pyruvate dehydrogenase, beta; PFKFP2, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 2; PFKM, phosphofructokinase, muscle; PGK1, phosphoglycerate kinase 1; PPARG, peroxisome proliferator activated receptor gamma; SIM1, single-minded family BHLH transcription factor 1; TBC1D1, TBC1 domain family member 1; UCP3, uncoupling protein 3; TP53, tumor protein P53; VEGF, vascular endothelial growth factor.

References

  1. Argiles, J.M.; Campos, N.; Lopez-Pedrosa, J.M.; Rueda, R.; Rodriguez-Manas, L. Skeletal Muscle Regulates Metabolism via Interorgan Crosstalk: Roles in Health and Disease. J. Am. Med. Dir. Assoc. 2016, 17, 789–796. [Google Scholar] [CrossRef]
  2. Chow, L.S.; Gerszten, R.E.; Taylor, J.M.; Pedersen, B.K.; van Praag, H.; Trappe, S.; Febbraio, M.A.; Galis, Z.S.; Gao, Y.; Haus, J.M.; et al. Exerkines in health, resilience and disease. Nat. Rev. Endocrinol. 2022, 18, 273–289. [Google Scholar] [CrossRef] [PubMed]
  3. Pillon, N.J.; Bilan, P.J.; Fink, L.N.; Klip, A. Cross-talk between skeletal muscle and immune cells: Muscle-derived mediators and metabolic implications. Am. J. Physiol. Endocrinol. Metab. 2013, 304, E453–E465. [Google Scholar] [CrossRef] [PubMed]
  4. Fluck, M. Functional, structural and molecular plasticity of mammalian skeletal muscle in response to exercise stimuli. J. Exp. Biol. 2006, 209, 2239–2248. [Google Scholar] [CrossRef] [PubMed]
  5. Hughes, D.C.; Ellefsen, S.; Baar, K. Adaptations to Endurance and Strength Training. Cold Spring Harb. Perspect. Med. 2018, 8, a029769. [Google Scholar] [CrossRef] [PubMed]
  6. Schiaffino, S.; Reggiani, C. Fiber types in mammalian skeletal muscles. Physiol. Rev. 2011, 91, 1447–1531. [Google Scholar] [CrossRef] [PubMed]
  7. Smith, J.A.B.; Murach, K.A.; Dyar, K.A.; Zierath, J.R. Exercise metabolism and adaptation in skeletal muscle. Nat. Rev. Mol. Cell Biol. 2023, 24, 607–632. [Google Scholar] [CrossRef] [PubMed]
  8. Beiter, T.; Niess, A.M.; Moser, D. Transcriptional memory in skeletal muscle. Don’t forget (to) exercise. J. Cell Physiol. 2020, 235, 5476–5489. [Google Scholar] [CrossRef]
  9. Egan, B.; Sharples, A.P. Molecular responses to acute exercise and their relevance for adaptations in skeletal muscle to exercise training. Physiol. Rev. 2023, 103, 2057–2170. [Google Scholar] [CrossRef]
  10. Furrer, R.; Hawley, J.A.; Handschin, C. The molecular athlete: Exercise physiology from mechanisms to medals. Physiol. Rev. 2023, 103, 1693–1787. [Google Scholar] [CrossRef]
  11. Kujala, U.M. Evidence on the effects of exercise therapy in the treatment of chronic disease. Br. J. Sports Med. 2009, 43, 550–555. [Google Scholar] [CrossRef] [PubMed]
  12. Catoire, M.; Mensink, M.; Boekschoten, M.V.; Hangelbroek, R.; Muller, M.; Schrauwen, P.; Kersten, S. Pronounced effects of acute endurance exercise on gene expression in resting and exercising human skeletal muscle. PLoS ONE 2012, 7, e51066. [Google Scholar] [CrossRef] [PubMed]
  13. Lindholm, M.E.; Giacomello, S.; Werne, S.B.; Fischer, H.; Huss, M.; Kjellqvist, S.; Sundberg, C.J. The Impact of Endurance Training on Human Skeletal Muscle Memory, Global Isoform Expression and Novel Transcripts. PLoS Genet. 2016, 12, e1006294. [Google Scholar] [CrossRef]
  14. Liu, D.; Sartor, M.A.; Nader, G.A.; Gutmann, L.; Treutelaar, M.K.; Pistilli, E.E.; Iglayreger, H.B.; Burant, C.F.; Hoffman, E.P.; Gordon, P.M. Skeletal muscle gene expression in response to resistance exercise: Sex specific regulation. BMC Genom. 2010, 11, 659. [Google Scholar] [CrossRef] [PubMed]
  15. Lundberg, T.R.; Fernandez-Gonzalo, R.; Tesch, P.A.; Rullman, E.; Gustafsson, T. Aerobic exercise augments muscle transcriptome profile of resistance exercise. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2016, 310, R1279–R1287. [Google Scholar] [CrossRef] [PubMed]
  16. Mahoney, D.J.; Parise, G.; Melov, S.; Safdar, A.; Tarnopolsky, M.A. Analysis of global mRNA expression in human skeletal muscle during recovery from endurance exercise. FASEB J. 2005, 19, 1498–1500. [Google Scholar] [CrossRef]
  17. Nader, G.A.; von Walden, F.; Liu, C.; Lindvall, J.; Gutmann, L.; Pistilli, E.E.; Gordon, P.M. Resistance exercise training modulates acute gene expression during human skeletal muscle hypertrophy. J. Appl. Physiol. 2014, 116, 693–702. [Google Scholar] [CrossRef]
  18. Neubauer, O.; Sabapathy, S.; Ashton, K.J.; Desbrow, B.; Peake, J.M.; Lazarus, R.; Wessner, B.; Cameron-Smith, D.; Wagner, K.H.; Haseler, L.J.; et al. Time course-dependent changes in the transcriptome of human skeletal muscle during recovery from endurance exercise: From inflammation to adaptive remodeling. J. Appl. Physiol. 2014, 116, 274–287. [Google Scholar] [CrossRef]
  19. Padrao, A.I.; Ferreira, R.; Amado, F.; Vitorino, R.; Duarte, J.A. Uncovering the exercise-related proteome signature in skeletal muscle. Proteomics 2016, 16, 816–830. [Google Scholar] [CrossRef] [PubMed]
  20. Raue, U.; Trappe, T.A.; Estrem, S.T.; Qian, H.R.; Helvering, L.M.; Smith, R.C.; Trappe, S. Transcriptome signature of resistance exercise adaptations: Mixed muscle and fiber type specific profiles in young and old adults. J. Appl. Physiol. 2012, 112, 1625–1636. [Google Scholar] [CrossRef] [PubMed]
  21. Schild, M.; Ruhs, A.; Beiter, T.; Zugel, M.; Hudemann, J.; Reimer, A.; Krumholz-Wagner, I.; Wagner, C.; Keller, J.; Eder, K.; et al. Basal and exercise induced label-free quantitative protein profiling of m. vastus lateralis in trained and untrained individuals. J. Proteom. 2015, 122, 119–132. [Google Scholar] [CrossRef] [PubMed]
  22. Vissing, K.; Schjerling, P. Simplified data access on human skeletal muscle transcriptome responses to differentiated exercise. Sci. Data 2014, 1, 140041. [Google Scholar] [CrossRef] [PubMed]
  23. Weigert, C.; Lehmann, R.; Hartwig, S.; Lehr, S. The secretome of the working human skeletal muscle—A promising opportunity to combat the metabolic disaster? Proteom. Clin. Appl. 2014, 8, 5–18. [Google Scholar] [CrossRef] [PubMed]
  24. Schild, M.; Eichner, G.; Beiter, T.; Zugel, M.; Krumholz-Wagner, I.; Hudemann, J.; Pilat, C.; Kruger, K.; Niess, A.M.; Steinacker, J.M.; et al. Effects of Acute Endurance Exercise on Plasma Protein Profiles of Endurance-Trained and Untrained Individuals over Time. Mediat. Inflamm. 2016, 2016, 4851935. [Google Scholar] [CrossRef] [PubMed]
  25. Bizjak, D.A.; Zugel, M.; Treff, G.; Winkert, K.; Jerg, A.; Hudemann, J.; Mooren, F.C.; Kruger, K.; Niess, A.; Steinacker, J.M. Effects of Training Status and Exercise Mode on Global Gene Expression in Skeletal Muscle. Int. J. Mol. Sci. 2021, 22, 12578. [Google Scholar] [CrossRef]
  26. Staron, R.S.; Hagerman, F.C.; Hikida, R.S.; Murray, T.F.; Hostler, D.P.; Crill, M.T.; Ragg, K.E.; Toma, K. Fiber type composition of the vastus lateralis muscle of young men and women. J. Histochem. Cytochem. 2000, 48, 623–629. [Google Scholar] [CrossRef]
  27. Stuart, C.A.; Stone, W.L.; Howell, M.E.; Brannon, M.F.; Hall, H.K.; Gibson, A.L.; Stone, M.H. Myosin content of individual human muscle fibers isolated by laser capture microdissection. Am. J. Physiol. Cell Physiol. 2016, 310, C381–C389. [Google Scholar] [CrossRef]
  28. Delbono, O.; Meissner, G. Sarcoplasmic reticulum Ca2+ release in rat slow- and fast-twitch muscles. J. Membr. Biol. 1996, 151, 123–130. [Google Scholar] [CrossRef]
  29. Dhoot, G.K.; Perry, S.V. Distribution of polymorphic forms of troponin components and tropomyosin in skeletal muscle. Nature 1979, 278, 714–718. [Google Scholar] [CrossRef]
  30. Murgia, M.; Nogara, L.; Baraldo, M.; Reggiani, C.; Mann, M.; Schiaffino, S. Protein profile of fiber types in human skeletal muscle: A single-fiber proteomics study. Skelet. Muscle 2021, 11, 24. [Google Scholar] [CrossRef] [PubMed]
  31. Bicer, S.; Reiser, P.J. Myosin light chain isoform expression among single mammalian skeletal muscle fibers: Species variations. J. Muscle Res. Cell Motil. 2004, 25, 623–633. [Google Scholar] [CrossRef] [PubMed]
  32. Petchey, L.K.; Risebro, C.A.; Vieira, J.M.; Roberts, T.; Bryson, J.B.; Greensmith, L.; Lythgoe, M.F.; Riley, P.R. Loss of Prox1 in striated muscle causes slow to fast skeletal muscle fiber conversion and dilated cardiomyopathy. Proc. Natl. Acad. Sci. USA 2014, 111, 9515–9520. [Google Scholar] [CrossRef] [PubMed]
  33. Drexler, H.C.; Ruhs, A.; Konzer, A.; Mendler, L.; Bruckskotten, M.; Looso, M.; Gunther, S.; Boettger, T.; Kruger, M.; Braun, T. On marathons and Sprints: An integrated quantitative proteomics and transcriptomics analysis of differences between slow and fast muscle fibers. Mol. Cell Proteom. 2012, 11, M111. [Google Scholar] [CrossRef] [PubMed]
  34. Swoap, S.J.; Hunter, R.B.; Stevenson, E.J.; Felton, H.M.; Kansagra, N.V.; Lang, J.M.; Esser, K.A.; Kandarian, S.C. The calcineurin-NFAT pathway and muscle fiber-type gene expression. Am. J. Physiol. Cell Physiol. 2000, 279, C915–C924. [Google Scholar] [CrossRef] [PubMed]
  35. Vistisen, B.; Roepstorff, K.; Roepstorff, C.; Bonen, A.; van Deurs, B.; Kiens, B. Sarcolemmal FAT/CD36 in human skeletal muscle colocalizes with caveolin-3 and is more abundant in type 1 than in type 2 fibers. J. Lipid Res. 2004, 45, 603–609. [Google Scholar] [CrossRef] [PubMed]
  36. Deshmukh, A.S.; Steenberg, D.E.; Hostrup, M.; Birk, J.B.; Larsen, J.K.; Santos, A.; Kjobsted, R.; Hingst, J.R.; Scheele, C.C.; Murgia, M.; et al. Deep muscle-proteomic analysis of freeze-dried human muscle biopsies reveals fiber type-specific adaptations to exercise training. Nat. Commun. 2021, 12, 304. [Google Scholar] [CrossRef]
  37. Gautel, M.; Furst, D.O.; Cocco, A.; Schiaffino, S. Isoform transitions of the myosin binding protein C family in developing human and mouse muscles: Lack of isoform transcomplementation in cardiac muscle. Circ. Res. 1998, 82, 124–129. [Google Scholar] [CrossRef] [PubMed]
  38. Chemello, F.; Bean, C.; Cancellara, P.; Laveder, P.; Reggiani, C.; Lanfranchi, G. Microgenomic analysis in skeletal muscle: Expression signatures of individual fast and slow myofibers. PLoS ONE 2011, 6, e16807. [Google Scholar] [CrossRef]
  39. Mills, M.; Yang, N.; Weinberger, R.; Vander Woude, D.L.; Beggs, A.H.; Easteal, S.; North, K. Differential expression of the actin-binding proteins, alpha-actinin-2 and -3, in different species: Implications for the evolution of functional redundancy. Hum. Mol. Genet. 2001, 10, 1335–1346. [Google Scholar] [CrossRef]
  40. Cariou, B.; van Harmelen, K.; Duran-Sandoval, D.; van Dijk, T.H.; Grefhorst, A.; Abdelkarim, M.; Caron, S.; Torpier, G.; Fruchart, J.C.; Gonzalez, F.J.; et al. The farnesoid X receptor modulates adiposity and peripheral insulin sensitivity in mice. J. Biol. Chem. 2006, 281, 11039–11049. [Google Scholar] [CrossRef]
  41. Lindholm, M.E.; Marabita, F.; Gomez-Cabrero, D.; Rundqvist, H.; Ekstrom, T.J.; Tegner, J.; Sundberg, C.J. An integrative analysis reveals coordinated reprogramming of the epigenome and the transcriptome in human skeletal muscle after training. Epigenetics 2014, 9, 1557–1569. [Google Scholar] [CrossRef]
  42. Chapman, M.A.; Arif, M.; Emanuelsson, E.B.; Reitzner, S.M.; Lindholm, M.E.; Mardinoglu, A.; Sundberg, C.J. Skeletal Muscle Transcriptomic Comparison between Long-Term Trained and Untrained Men and Women. Cell Rep. 2020, 31, 107808. [Google Scholar] [CrossRef]
  43. Porporato, P.E.; Dhup, S.; Dadhich, R.K.; Copetti, T.; Sonveaux, P. Anticancer targets in the glycolytic metabolism of tumors: A comprehensive review. Front. Pharmacol. 2011, 2, 49. [Google Scholar] [CrossRef] [PubMed]
  44. Liang, X.; Liu, L.; Fu, T.; Zhou, Q.; Zhou, D.; Xiao, L.; Liu, J.; Kong, Y.; Xie, H.; Yi, F.; et al. Exercise Inducible Lactate Dehydrogenase B Regulates Mitochondrial Function in Skeletal Muscle. J. Biol. Chem. 2016, 291, 25306–25318. [Google Scholar] [CrossRef] [PubMed]
  45. Gan, Z.; Burkart-Hartman, E.M.; Han, D.H.; Finck, B.; Leone, T.C.; Smith, E.Y.; Ayala, J.E.; Holloszy, J.; Kelly, D.P. The nuclear receptor PPARbeta/delta programs muscle glucose metabolism in cooperation with AMPK and MEF2. Genes Dev. 2011, 25, 2619–2630. [Google Scholar] [CrossRef] [PubMed]
  46. Furrer, R.; Heim, B.; Schmid, S.; Dilbaz, S.; Adak, V.; Nordstrom, K.J.V.; Ritz, D.; Steurer, S.A.; Walter, J.; Handschin, C. Molecular control of endurance training adaptation in male mouse skeletal muscle. Nat. Metab. 2023, 5, 2020–2035. [Google Scholar] [CrossRef] [PubMed]
  47. Summermatter, S.; Santos, G.; Perez-Schindler, J.; Handschin, C. Skeletal muscle PGC-1alpha controls whole-body lactate homeostasis through estrogen-related receptor alpha-dependent activation of LDH B and repression of LDH A. Proc. Natl. Acad. Sci. USA 2013, 110, 8738–8743. [Google Scholar] [CrossRef] [PubMed]
  48. Valvona, C.J.; Fillmore, H.L.; Nunn, P.B.; Pilkington, G.J. The Regulation and Function of Lactate Dehydrogenase A: Therapeutic Potential in Brain Tumor. Brain Pathol. 2016, 26, 3–17. [Google Scholar] [CrossRef] [PubMed]
  49. Kim, J.W.; Tchernyshyov, I.; Semenza, G.L.; Dang, C.V. HIF-1-mediated expression of pyruvate dehydrogenase kinase: A metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 2006, 3, 177–185. [Google Scholar] [CrossRef]
  50. Yarden, Y.; Sliwkowski, M.X. Untangling the ErbB signalling network. Nat. Rev. Mol. Cell Biol. 2001, 2, 127–137. [Google Scholar] [CrossRef]
  51. Mason, S.D.; Howlett, R.A.; Kim, M.J.; Olfert, I.M.; Hogan, M.C.; McNulty, W.; Hickey, R.P.; Wagner, P.D.; Kahn, C.R.; Giordano, F.J.; et al. Loss of skeletal muscle HIF-1alpha results in altered exercise endurance. PLoS Biol. 2004, 2, e288. [Google Scholar] [CrossRef]
  52. Ciano, M.; Mantellato, G.; Connolly, M.; Paul-Clark, M.; Willis-Owen, S.; Moffatt, M.F.; Cookson, W.; Mitchell, J.A.; Polkey, M.I.; Hughes, S.M.; et al. EGF receptor (EGFR) inhibition promotes a slow-twitch oxidative, over a fast-twitch, muscle phenotype. Sci. Rep. 2019, 9, 9218. [Google Scholar] [CrossRef]
  53. Wang, Y.X.; Feige, P.; Brun, C.E.; Hekmatnejad, B.; Dumont, N.A.; Renaud, J.M.; Faulkes, S.; Guindon, D.E.; Rudnicki, M.A. EGFR-Aurka Signaling Rescues Polarity and Regeneration Defects in Dystrophin-Deficient Muscle Stem Cells by Increasing Asymmetric Divisions. Cell Stem Cell 2019, 24, 419–432.e416. [Google Scholar] [CrossRef]
  54. Truong, T.H.; Ung, P.M.; Palde, P.B.; Paulsen, C.E.; Schlessinger, A.; Carroll, K.S. Molecular Basis for Redox Activation of Epidermal Growth Factor Receptor Kinase. Cell Chem. Biol. 2016, 23, 837–848. [Google Scholar] [CrossRef]
  55. Pedersen, S.B.; Kristensen, K.; Bruun, J.M.; Flyvbjerg, A.; Vinter-Jensen, L.; Richelsen, B. Systemic administration of epidermal growth factor increases UCP3 mRNA levels in skeletal muscle and adipose tissue in rats. Biochem. Biophys. Res. Commun. 2000, 279, 914–919. [Google Scholar] [CrossRef] [PubMed]
  56. Busiello, R.A.; Savarese, S.; Lombardi, A. Mitochondrial uncoupling proteins and energy metabolism. Front. Physiol. 2015, 6, 36. [Google Scholar] [CrossRef] [PubMed]
  57. Pohl, E.E.; Rupprecht, A.; Macher, G.; Hilse, K.E. Important Trends in UCP3 Investigation. Front. Physiol. 2019, 10, 470. [Google Scholar] [CrossRef] [PubMed]
  58. Russell, A.P.; Somm, E.; Praz, M.; Crettenand, A.; Hartley, O.; Melotti, A.; Giacobino, J.P.; Muzzin, P.; Gobelet, C.; Deriaz, O. UCP3 protein regulation in human skeletal muscle fibre types I, IIa and IIx is dependent on exercise intensity. J. Physiol. 2003, 550, 855–861. [Google Scholar] [CrossRef] [PubMed]
  59. Mogensen, M.; Bagger, M.; Pedersen, P.K.; Fernstrom, M.; Sahlin, K. Cycling efficiency in humans is related to low UCP3 content and to type I fibres but not to mitochondrial efficiency. J. Physiol. 2006, 571, 669–681. [Google Scholar] [CrossRef] [PubMed]
  60. Choi, C.S.; Fillmore, J.J.; Kim, J.K.; Liu, Z.X.; Kim, S.; Collier, E.F.; Kulkarni, A.; Distefano, A.; Hwang, Y.J.; Kahn, M.; et al. Overexpression of uncoupling protein 3 in skeletal muscle protects against fat-induced insulin resistance. J. Clin. Investig. 2007, 117, 1995–2003. [Google Scholar] [CrossRef] [PubMed]
  61. Aguer, C.; Fiehn, O.; Seifert, E.L.; Bezaire, V.; Meissen, J.K.; Daniels, A.; Scott, K.; Renaud, J.M.; Padilla, M.; Bickel, D.R.; et al. Muscle uncoupling protein 3 overexpression mimics endurance training and reduces circulating biomarkers of incomplete beta-oxidation. FASEB J. 2013, 27, 4213–4225. [Google Scholar] [CrossRef] [PubMed]
  62. Mourkioti, F.; Rosenthal, N. NF-kappaB signaling in skeletal muscle: Prospects for intervention in muscle diseases. J. Mol. Med. 2008, 86, 747–759. [Google Scholar] [CrossRef] [PubMed]
  63. Bakkar, N.; Ladner, K.; Canan, B.D.; Liyanarachchi, S.; Bal, N.C.; Pant, M.; Periasamy, M.; Li, Q.; Janssen, P.M.; Guttridge, D.C. IKKalpha and alternative NF-kappaB regulate PGC-1beta to promote oxidative muscle metabolism. J. Cell Biol. 2012, 196, 497–511. [Google Scholar] [CrossRef]
  64. Shintaku, J.; Peterson, J.M.; Talbert, E.E.; Gu, J.M.; Ladner, K.J.; Williams, D.R.; Mousavi, K.; Wang, R.; Sartorelli, V.; Guttridge, D.C. MyoD Regulates Skeletal Muscle Oxidative Metabolism Cooperatively with Alternative NF-kappaB. Cell Rep. 2016, 17, 514–526. [Google Scholar] [CrossRef]
  65. Bartlett, J.D.; Close, G.L.; Drust, B.; Morton, J.P. The emerging role of p53 in exercise metabolism. Sports Med. 2014, 44, 303–309. [Google Scholar] [CrossRef] [PubMed]
  66. Drake, J.C.; Wilson, R.J.; Yan, Z. Molecular mechanisms for mitochondrial adaptation to exercise training in skeletal muscle. FASEB J. 2016, 30, 13–22. [Google Scholar] [CrossRef]
  67. Saleem, A.; Hood, D.A. Acute exercise induces tumour suppressor protein p53 translocation to the mitochondria and promotes a p53-Tfam-mitochondrial DNA complex in skeletal muscle. J. Physiol. 2013, 591, 3625–3636. [Google Scholar] [CrossRef]
  68. Wattez, J.S.; Eury, E.; Hazen, B.C.; Wade, A.; Chau, S.; Ou, S.C.; Russell, A.P.; Cho, Y.; Kralli, A. Loss of skeletal muscle estrogen-related receptors leads to severe exercise intolerance. Mol. Metab. 2023, 68, 101670. [Google Scholar] [CrossRef]
  69. Agudelo, L.Z.; Femenia, T.; Orhan, F.; Porsmyr-Palmertz, M.; Goiny, M.; Martinez-Redondo, V.; Correia, J.C.; Izadi, M.; Bhat, M.; Schuppe-Koistinen, I.; et al. Skeletal muscle PGC-1alpha1 modulates kynurenine metabolism and mediates resilience to stress-induced depression. Cell 2014, 159, 33–45. [Google Scholar] [CrossRef]
  70. Hayden, M.S.; Ghosh, S. Regulation of NF-kappaB by TNF family cytokines. Semin. Immunol. 2014, 26, 253–266. [Google Scholar] [CrossRef]
  71. Alger, H.M.; Raben, N.; Pistilli, E.; Francia, D.L.; Rawat, R.; Getnet, D.; Ghimbovschi, S.; Chen, Y.W.; Lundberg, I.E.; Nagaraju, K. The role of TRAIL in mediating autophagy in myositis skeletal muscle: A potential nonimmune mechanism of muscle damage. Arthritis Rheum. 2011, 63, 3448–3457. [Google Scholar] [CrossRef]
  72. Bernardi, S.; Toffoli, B.; Tisato, V.; Bossi, F.; Biffi, S.; Lorenzon, A.; Zauli, G.; Secchiero, P.; Fabris, B. TRAIL reduces impaired glucose tolerance and NAFLD in the high-fat diet fed mouse. Clin. Sci. 2018, 132, 69–83. [Google Scholar] [CrossRef]
  73. Koliaki, C.; Katsilambros, N. Repositioning the Role of Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) on the TRAIL to the Development of Diabetes Mellitus: An Update of Experimental and Clinical Evidence. Int. J. Mol. Sci. 2022, 23, 3225. [Google Scholar] [CrossRef]
  74. Ameln, H.; Gustafsson, T.; Sundberg, C.J.; Okamoto, K.; Jansson, E.; Poellinger, L.; Makino, Y. Physiological activation of hypoxia inducible factor-1 in human skeletal muscle. FASEB J. 2005, 19, 1009–1011. [Google Scholar] [CrossRef]
  75. Gallego-Selles, A.; Galvan-Alvarez, V.; Martinez-Canton, M.; Garcia-Gonzalez, E.; Morales-Alamo, D.; Santana, A.; Gonzalez-Henriquez, J.J.; Dorado, C.; Calbet, J.A.L.; Martin-Rincon, M. Fast regulation of the NF-kappaB signalling pathway in human skeletal muscle revealed by high-intensity exercise and ischaemia at exhaustion: Role of oxygenation and metabolite accumulation. Redox Biol. 2022, 55, 102398. [Google Scholar] [CrossRef] [PubMed]
  76. Lindholm, M.E.; Rundqvist, H. Skeletal muscle hypoxia-inducible factor-1 and exercise. Exp. Physiol. 2016, 101, 28–32. [Google Scholar] [CrossRef] [PubMed]
  77. Li, J.; Li, Y.; Atakan, M.M.; Kuang, J.; Hu, Y.; Bishop, D.J.; Yan, X. The Molecular Adaptive Responses of Skeletal Muscle to High-Intensity Exercise/Training and Hypoxia. Antioxidants 2020, 9, 656. [Google Scholar] [CrossRef] [PubMed]
  78. Favier, F.B.; Britto, F.A.; Freyssenet, D.G.; Bigard, X.A.; Benoit, H. HIF-1-driven skeletal muscle adaptations to chronic hypoxia: Molecular insights into muscle physiology. Cell Mol. Life Sci. 2015, 72, 4681–4696. [Google Scholar] [CrossRef] [PubMed]
  79. Ma, B.; Hottiger, M.O. Crosstalk between Wnt/beta-Catenin and NF-kappaB Signaling Pathway during Inflammation. Front. Immunol. 2016, 7, 378. [Google Scholar] [CrossRef] [PubMed]
  80. D’Ignazio, L.; Bandarra, D.; Rocha, S. NF-kappaB and HIF crosstalk in immune responses. FEBS J. 2016, 283, 413–424. [Google Scholar] [CrossRef] [PubMed]
  81. Prescott, J.A.; Mitchell, J.P.; Cook, S.J. Inhibitory feedback control of NF-kappaB signalling in health and disease. Biochem. J. 2021, 478, 2619–2664. [Google Scholar] [CrossRef] [PubMed]
  82. Powers, S.K.; Deminice, R.; Ozdemir, M.; Yoshihara, T.; Bomkamp, M.P.; Hyatt, H. Exercise-induced oxidative stress: Friend or foe? J. Sport. Health Sci. 2020, 9, 415–425. [Google Scholar] [CrossRef] [PubMed]
  83. Merry, T.L.; Ristow, M. Mitohormesis in exercise training. Free Radic. Biol. Med. 2016, 98, 123–130. [Google Scholar] [CrossRef] [PubMed]
  84. Ngo, V.; Duennwald, M.L. Nrf2 and Oxidative Stress: A General Overview of Mechanisms and Implications in Human Disease. Antioxidants 2022, 11, 2345. [Google Scholar] [CrossRef]
  85. Bensellam, M.; Montgomery, M.K.; Luzuriaga, J.; Chan, J.Y.; Laybutt, D.R. Inhibitor of differentiation proteins protect against oxidative stress by regulating the antioxidant-mitochondrial response in mouse beta cells. Diabetologia 2015, 58, 758–770. [Google Scholar] [CrossRef]
  86. Consoli, V.; Sorrenti, V.; Grosso, S.; Vanella, L. Heme Oxygenase-1 Signaling and Redox Homeostasis in Physiopathological Conditions. Biomolecules 2021, 11, 589. [Google Scholar] [CrossRef]
  87. Beiter, T.; Hoene, M.; Prenzler, F.; Mooren, F.C.; Steinacker, J.M.; Weigert, C.; Niess, A.M.; Munz, B. Exercise, skeletal muscle and inflammation: ARE-binding proteins as key regulators in inflammatory and adaptive networks. Exerc. Immunol. Rev. 2015, 21, 42–57. [Google Scholar]
  88. Rubenstein, A.B.; Hinkley, J.M.; Nair, V.D.; Nudelman, G.; Standley, R.A.; Yi, F.; Yu, G.; Trappe, T.A.; Bamman, M.M.; Trappe, S.W.; et al. Skeletal muscle transcriptome response to a bout of endurance exercise in physically active and sedentary older adults. Am. J. Physiol. Endocrinol. Metab. 2022, 322, E260–E277. [Google Scholar] [CrossRef]
  89. Williams, K.; Carrasquilla, G.D.; Ingerslev, L.R.; Hochreuter, M.Y.; Hansson, S.; Pillon, N.J.; Donkin, I.; Versteyhe, S.; Zierath, J.R.; Kilpelainen, T.O.; et al. Epigenetic rewiring of skeletal muscle enhancers after exercise training supports a role in whole-body function and human health. Mol. Metab. 2021, 53, 101290. [Google Scholar] [CrossRef]
  90. Liu, Z.; Wang, C.; Liu, X.; Kuang, S. Shisa2 regulates the fusion of muscle progenitors. Stem Cell Res. 2018, 31, 31–41. [Google Scholar] [CrossRef] [PubMed]
  91. Smyth, G.K. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 2004, 3, 3. [Google Scholar] [CrossRef] [PubMed]
  92. Wrzodek, C.; Eichner, J.; Buchel, F.; Zell, A. InCroMAP: Integrated analysis of cross-platform microarray and pathway data. Bioinformatics 2013, 29, 506–508. [Google Scholar] [CrossRef]
  93. Chen, D.; Pan, X.; Xiao, P.; Farwell, M.A.; Zhang, B. Evaluation and identification of reliable reference genes for pharmacogenomics, toxicogenomics, and small RNA expression analysis. J. Cell Physiol. 2011, 226, 2469–2477. [Google Scholar] [CrossRef] [PubMed]
  94. Livak, K.J.; Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001, 25, 402–408. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic illustration of the acute exercise setup. Biopsies were obtained from the vastus lateralis muscle of the right leg. Baseline expression (rest) was determined from samples taken 24 h before the acute exercise experiment. Exercise consisted of 10 min of moderate warm-up cycling and 50 min of strenuous cycling exercise adjusted in its intensity to 80% of the individual oxygen uptake capacity (VO2max). Acute exercise-provoked expression profiles were evaluated from biopsy samples taken at 30 min (+30′) and at 3 h (+3 h) after the cycling exercise.
Figure 1. Schematic illustration of the acute exercise setup. Biopsies were obtained from the vastus lateralis muscle of the right leg. Baseline expression (rest) was determined from samples taken 24 h before the acute exercise experiment. Exercise consisted of 10 min of moderate warm-up cycling and 50 min of strenuous cycling exercise adjusted in its intensity to 80% of the individual oxygen uptake capacity (VO2max). Acute exercise-provoked expression profiles were evaluated from biopsy samples taken at 30 min (+30′) and at 3 h (+3 h) after the cycling exercise.
Ijms 25 02881 g001
Figure 2. Baseline transcriptome signatures. (A) Heat map of differentially expressed probe sets (fold difference |≥| 1.5, p < 0.05) at rest in vastus lateralis muscle between endurance-trained (ET, n = 8) and sedentary (SED, n = 8) individuals. Probe sets (rows) and individuals (columns) have been ordered based on hierarchical clustering. Relative expression values are represented as a color scale according to the deviation from the average level of expression across all individuals, ranging from green for low expression to red for high expression. (B) Volcano plot of differentially expressed genes between ET and SED. The logarithms of the fold changes are plotted against the negative logarithm of the p-value to base 10. Positive Log2 fold change values represent genes that display significantly higher expression levels in the ET group (red circles), and negative values indicate increased expression in SED individuals (blue circles). The top 10 genes with highest fold change differences are highlighted for both groups. (C) Significantly enriched GO terms in genes with significantly higher baseline levels in ET (red) and SED (blue) individuals, respectively. GO terms were sorted based on corrected p values (−Log10 (Q-value) > 5).
Figure 2. Baseline transcriptome signatures. (A) Heat map of differentially expressed probe sets (fold difference |≥| 1.5, p < 0.05) at rest in vastus lateralis muscle between endurance-trained (ET, n = 8) and sedentary (SED, n = 8) individuals. Probe sets (rows) and individuals (columns) have been ordered based on hierarchical clustering. Relative expression values are represented as a color scale according to the deviation from the average level of expression across all individuals, ranging from green for low expression to red for high expression. (B) Volcano plot of differentially expressed genes between ET and SED. The logarithms of the fold changes are plotted against the negative logarithm of the p-value to base 10. Positive Log2 fold change values represent genes that display significantly higher expression levels in the ET group (red circles), and negative values indicate increased expression in SED individuals (blue circles). The top 10 genes with highest fold change differences are highlighted for both groups. (C) Significantly enriched GO terms in genes with significantly higher baseline levels in ET (red) and SED (blue) individuals, respectively. GO terms were sorted based on corrected p values (−Log10 (Q-value) > 5).
Ijms 25 02881 g002
Figure 3. Differentially expressed metabolic genes and upstream regulators that define the basal transcriptome signature of the endurance-trained and untrained muscle. (A,B) Schematic representation of glycolytic and oxidative substrate flux with localization of key enzymes and transporters that display differential baseline gene expression levels between sedentary (SED) and endurance-trained (ET) individuals in vastus lateralis muscle at rest. Colors represent significant fold differences (p < 0.05) in the comparison of SED/ET (A, blue) and ET/SED (B, red), respectively. (C,D) Ingenuity upstream regulator analysis based on genes with significantly higher baseline expression levels in SED (C, blue) and ET (D, red) individuals, respectively. The top activated upstream regulators are shown with predicted activation Z-score ≥ 2 and a p-value of overlap < 0.05.
Figure 3. Differentially expressed metabolic genes and upstream regulators that define the basal transcriptome signature of the endurance-trained and untrained muscle. (A,B) Schematic representation of glycolytic and oxidative substrate flux with localization of key enzymes and transporters that display differential baseline gene expression levels between sedentary (SED) and endurance-trained (ET) individuals in vastus lateralis muscle at rest. Colors represent significant fold differences (p < 0.05) in the comparison of SED/ET (A, blue) and ET/SED (B, red), respectively. (C,D) Ingenuity upstream regulator analysis based on genes with significantly higher baseline expression levels in SED (C, blue) and ET (D, red) individuals, respectively. The top activated upstream regulators are shown with predicted activation Z-score ≥ 2 and a p-value of overlap < 0.05.
Ijms 25 02881 g003
Figure 4. Acute exercise-provoked transcriptional responses in endurance-trained and untrained muscle. (A) Venn diagram showing the overlap of acute exercise-affected genes (fold change |≥| 2 versus baseline, p < 0.05) between sedentary (SED, blue) and endurance-trained (ET, red) individuals in vastus lateralis muscle at 30 min (+30′) and at 3 h (+3 h) after intense cycling exercise for 1 h. (BE) Volcano plots for acute exercise-affected genes in SED (B,C, blue) and ET (D,E, red) individuals at +30′ (B,D) and at +3 h (C,E) after the exercise protocol. The logarithms of the fold changes are plotted against the negative logarithm of the p-value to base 10. Positive log2 fold change values represent genes that are significantly up-regulated by acute exercise, and negative values indicate genes that become repressed in response to exercise. The top 10 genes with highest fold change differences compared to baseline values at rest are highlighted.
Figure 4. Acute exercise-provoked transcriptional responses in endurance-trained and untrained muscle. (A) Venn diagram showing the overlap of acute exercise-affected genes (fold change |≥| 2 versus baseline, p < 0.05) between sedentary (SED, blue) and endurance-trained (ET, red) individuals in vastus lateralis muscle at 30 min (+30′) and at 3 h (+3 h) after intense cycling exercise for 1 h. (BE) Volcano plots for acute exercise-affected genes in SED (B,C, blue) and ET (D,E, red) individuals at +30′ (B,D) and at +3 h (C,E) after the exercise protocol. The logarithms of the fold changes are plotted against the negative logarithm of the p-value to base 10. Positive log2 fold change values represent genes that are significantly up-regulated by acute exercise, and negative values indicate genes that become repressed in response to exercise. The top 10 genes with highest fold change differences compared to baseline values at rest are highlighted.
Ijms 25 02881 g004
Figure 5. Identification of canonical pathways and upstream regulators that shape skeletal muscle response to acute exercise. (A,B) Comparison of top canonical pathways (ingenuity pathway analysis, IPA) between SED (blue bars) and ET (red bars) individuals that are enriched in exercised-affected gene sets at +30′ (A) and at +3 h (B) after exercise. Bars indicate −log p value (ratio of molecules present in the dataset out of all the function-related molecules). (C,D) Predicted upstream transcriptional regulators of genes that are significantly affected by exercise at +30′ (C) and at +3 h (D) in SED (blue bars) and ET (red bars) individuals. The top activated upstream regulators are shown with predicted activation Z-score |≥| 2 and a p-value of overlap <0.05. Positive Z-scores indicate activated factors, while negative scores indicate that a factor is inhibited.
Figure 5. Identification of canonical pathways and upstream regulators that shape skeletal muscle response to acute exercise. (A,B) Comparison of top canonical pathways (ingenuity pathway analysis, IPA) between SED (blue bars) and ET (red bars) individuals that are enriched in exercised-affected gene sets at +30′ (A) and at +3 h (B) after exercise. Bars indicate −log p value (ratio of molecules present in the dataset out of all the function-related molecules). (C,D) Predicted upstream transcriptional regulators of genes that are significantly affected by exercise at +30′ (C) and at +3 h (D) in SED (blue bars) and ET (red bars) individuals. The top activated upstream regulators are shown with predicted activation Z-score |≥| 2 and a p-value of overlap <0.05. Positive Z-scores indicate activated factors, while negative scores indicate that a factor is inhibited.
Ijms 25 02881 g005
Figure 6. Most stable transcriptional signature of exercise-unresponsive genes that delineates the endurance-trained muscle from the untrained muscle. Heat maps depicting a stable panel of 39 genes in vastus lateralis muscle that retain differential expression levels (fold difference I ≥ I 1.5, p < 0.05) between endurance-trained (ET, red) and sedentary (SED, blue) individuals at baseline (rest) as well as in response to acute exercise (+30′, +3 h). Probe sets (rows) and individuals (columns) have been ordered based on hierarchical clustering. Relative expression values are represented as a color scale according to the deviation from the average level of expression across all individuals, ranging from green for low expression to red for high expression.
Figure 6. Most stable transcriptional signature of exercise-unresponsive genes that delineates the endurance-trained muscle from the untrained muscle. Heat maps depicting a stable panel of 39 genes in vastus lateralis muscle that retain differential expression levels (fold difference I ≥ I 1.5, p < 0.05) between endurance-trained (ET, red) and sedentary (SED, blue) individuals at baseline (rest) as well as in response to acute exercise (+30′, +3 h). Probe sets (rows) and individuals (columns) have been ordered based on hierarchical clustering. Relative expression values are represented as a color scale according to the deviation from the average level of expression across all individuals, ranging from green for low expression to red for high expression.
Ijms 25 02881 g006
Figure 7. Muscular changes in basal transcript levels after 3 and 8 weeks of endurance training. Box plots represent Log2-transformed fold changes (FCs) in relative mRNA levels of selected target genes in vastus lateralis muscle. Short-term ((A), 3 wks) and medium-term ((B), 8 wks) changes in basal gene expression in the course of a standardized endurance exercise training program were assessed by qPCR from sedentary individuals (SED-T; n = 10) and compared to individual pre-training levels (baseline). Long-term adaptational changes in skeletal muscle transcriptional activity are reflected by the differential baseline expression pattern between endurance-trained (ET; n = 8) and sedentary (SED; n = 8) individuals that were derived from microarray data (C). * p < 0.05, ** p < 0.01, *** p < 0.001.
Figure 7. Muscular changes in basal transcript levels after 3 and 8 weeks of endurance training. Box plots represent Log2-transformed fold changes (FCs) in relative mRNA levels of selected target genes in vastus lateralis muscle. Short-term ((A), 3 wks) and medium-term ((B), 8 wks) changes in basal gene expression in the course of a standardized endurance exercise training program were assessed by qPCR from sedentary individuals (SED-T; n = 10) and compared to individual pre-training levels (baseline). Long-term adaptational changes in skeletal muscle transcriptional activity are reflected by the differential baseline expression pattern between endurance-trained (ET; n = 8) and sedentary (SED; n = 8) individuals that were derived from microarray data (C). * p < 0.05, ** p < 0.01, *** p < 0.001.
Ijms 25 02881 g007
Table 1. Physical characteristics and performance data of sedentary (SED) and endurance-trained (ET) subjects that participated in the acute exercise trial. Values represent means ± SD. BMI, body mass index; VO2max, maximal aerobic capacity; ***, significantly different compared with SED group (p < 0.001).
Table 1. Physical characteristics and performance data of sedentary (SED) and endurance-trained (ET) subjects that participated in the acute exercise trial. Values represent means ± SD. BMI, body mass index; VO2max, maximal aerobic capacity; ***, significantly different compared with SED group (p < 0.001).
Physical CharacteristicsAcute Exercise Performance
Age
(Years)
BMI
(kg/m2)
VO2max
(mL/min/kg)
Power Output
at 80% VO2max
(Watt)
Heart Rate
at End
(min−1)
Blood Lactate
at End
(mmol/L)
SED
(n = 8)
22.6 ± 2.823.7 ± 3.740.3 ± 7.6140 ± 40180 ± 76.8 ± 3.2
ET
(n = 8)
25.4 ± 3.822.9 ± 2.067.2 ± 8.9 ***260 ± 60 ***179 ± 97.4 ± 3.8
Table 2. List of genes significantly enriched in GO terms related to muscle structure and activity (GO:0030018, GO:0008307, GO:0005859, GO:0030049, GO:0006936, GO:0030017, GO:0042383) among differentially expressed genes between endurance-trained (ET) and sedentary (SED) individuals in vastus lateralis muscle at rest. Fold differences are given in the comparison of ET/SED with genes that showed relatively higher expression levels in the ET group (ET↑) and in the comparison of SED/ET when baseline levels were higher in the SED group (SED↑). Candidates with known patterns of fiber-type-preferential expression are indicated by ‘slow’ for type I fibers and ‘fast’ for type II fibers. N/D, no data available.
Table 2. List of genes significantly enriched in GO terms related to muscle structure and activity (GO:0030018, GO:0008307, GO:0005859, GO:0030049, GO:0006936, GO:0030017, GO:0042383) among differentially expressed genes between endurance-trained (ET) and sedentary (SED) individuals in vastus lateralis muscle at rest. Fold differences are given in the comparison of ET/SED with genes that showed relatively higher expression levels in the ET group (ET↑) and in the comparison of SED/ET when baseline levels were higher in the SED group (SED↑). Candidates with known patterns of fiber-type-preferential expression are indicated by ‘slow’ for type I fibers and ‘fast’ for type II fibers. N/D, no data available.
Gene
Symbol
Gene TitleProtein FunctionUp-Regulated
in
Fold
Difference
p-ValuePreferential
Fiber Type
Ref.
MYH7BMyosin, heavy chain 7B, cardiac muscle, betaMotor protein of myofilamentsET↑1.92.7 × 10−2slow[27]
ATP2A2ATPase, Ca++ transporting, cardiac muscle, slow twitch 2Sarcoplasmic reticulum calcium pumpET↑1.82.4 × 10−3slow[28]
TNNI3Troponin I type 3 (cardiac)Regulator of actin–myosin interactionET↑1.84.7 × 10−3slow[29]
KCNMA1Potassium large conductance calcium-activated channel, subfamily M, alpha member 1Regulator of muscle membrane potentialET↑1.82.6 × 10−3N/D
CASQ2Calsequestrin 2 (cardiac muscle)Calcium buffer in the sarcoplasmic reticulumET↑1.89.2 × 10−4slow[30]
MYL6BMyosin, light chain 6B, alkali, smooth muscle, and non-muscleConstituent of myofibrillar apparatusET↑1.75.1 × 10−4slow[31]
MYH6Myosin, heavy chain 6, cardiac muscle, alphaMotor protein of myofilamentsET↑1.61.8 × 10−2slow[27]
PROX1Prospero homeobox 1Regulator of contractile protein gene expressionET↑1.68.8 × 10−3slow[32]
MYL3Myosin, light chain 3, alkali; ventricular, skeletal, slowConstituent of myofibrillar apparatusET↑1.52.8 × 10−3slow[27]
MYOM3Myomesin family, member 3Structural constituent of the sarcomereET↑1.56.9 × 10−4slow[33]
SLMAPSarcolemma associated proteinRegulator of excitation–contraction couplingSED↑1.56.7 × 10−3slow[33]
SORBS2Sorbin and SH3 domain containing 2Assembly and maintenance of myofibrilsSED↑1.63.5 × 10−2N/D
PPP3CAProtein phosphatase 3, catalytic subunit, alpha isozymeCalcineurin; regulator of muscle massSED↑1.69.6 × 10−6fast[34]
CAV3Caveolin 3Integral membrane component of caveolaeSED↑1.64.7 × 10−2fast[35]
NEDD4Neural precursor cell expressed, developmentally down-regulated 4Mediator of skeletal muscle atrophySED↑1.64.1 × 10−3N/D
MYL5Myosin, light chain 5, regulatoryConstituent of myofibrillar apparatusSED↑1.63.6 × 10−3fast[31]
KCNJ12Potassium inwardly rectifying channel, subfamily J, member 12Stabilization of the resting membrane potentialSED↑1.68.0 × 10−3N/D
VCLVinculinStructural component of costameresSED↑1.61.3 × 10−2slow/fast[33]
DTNADystrobrevin, alphaStructural component of costameresSED↑1.61.3 × 10−2fast[33]
KIAA1161KIAA1161Putative regulator of myogenesisSED↑1.73.1 × 10−2fast[36]
MYBPC2Myosin binding protein C, fast typeRegulator of myofilament activation and relaxationSED↑1.72.3 × 10−5fast[37]
FHL2Four and a half LIM domains 2Putative regulator of sarcomere assemblySED↑1.72.7 × 10−3slow[38]
HOMER1Homer homolog 1 (Drosophila)Regulator of transient receptor potential channel activitySED↑1.78.7 × 10−4N/D
LMOD1Leiomodin 1 (smooth muscle)Regulator of actin filament lengthSED↑1.92.1 × 10−2N/D
ABRAActin-binding Rho activating proteinSarcomere-binding transcriptional regulatorSED↑2.08.3 × 10−5N/D
ATP2B2ATPase, Ca++ transporting, plasma membrane 2Plasma membrane calcium pumpSED↑2.22.1 × 10−2N/D
MYH1Myosin, heavy chain 1, skeletal muscle, adultMotor protein of myofilamentsSED↑3.51.9 × 10−2fast[36]
ACTN3Actinin, alpha 3Structural component of sarcomeric Z lineSED↑5.34.3 × 10−2fast[39]
Table 3. Baseline physical characteristics of sedentary subjects (SED-T; m; age, 27 ± 2.5) before (pre) and after (post) the 8-week exercise routine. Values represent means ± SD. BMI, body mass index; IAT, individual anaerobic threshold; VO2max, maximal aerobic capacity; ***, significantly different compared with individual pre-training level (p < 0.001).
Table 3. Baseline physical characteristics of sedentary subjects (SED-T; m; age, 27 ± 2.5) before (pre) and after (post) the 8-week exercise routine. Values represent means ± SD. BMI, body mass index; IAT, individual anaerobic threshold; VO2max, maximal aerobic capacity; ***, significantly different compared with individual pre-training level (p < 0.001).
Weight
(kg)
BMI
(kg/m2)
IAT
(Watt/kg)
VO2max
(mL/min/kg)
pre
(n = 10)
79.5 ± 13.925.3 ± 4.41.6 ± 0.536.1 ± 6.3
post
(n = 10)
79.2 ± 14.025.3 ± 4.32.0 ± 0.3 ***43.7 ± 6.4 ***
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Beiter, T.; Zügel, M.; Hudemann, J.; Schild, M.; Fragasso, A.; Burgstahler, C.; Krüger, K.; Mooren, F.C.; Steinacker, J.M.; Nieß, A.M. The Acute, Short-, and Long-Term Effects of Endurance Exercise on Skeletal Muscle Transcriptome Profiles. Int. J. Mol. Sci. 2024, 25, 2881. https://doi.org/10.3390/ijms25052881

AMA Style

Beiter T, Zügel M, Hudemann J, Schild M, Fragasso A, Burgstahler C, Krüger K, Mooren FC, Steinacker JM, Nieß AM. The Acute, Short-, and Long-Term Effects of Endurance Exercise on Skeletal Muscle Transcriptome Profiles. International Journal of Molecular Sciences. 2024; 25(5):2881. https://doi.org/10.3390/ijms25052881

Chicago/Turabian Style

Beiter, Thomas, Martina Zügel, Jens Hudemann, Marius Schild, Annunziata Fragasso, Christof Burgstahler, Karsten Krüger, Frank C. Mooren, Jürgen M. Steinacker, and Andreas M. Nieß. 2024. "The Acute, Short-, and Long-Term Effects of Endurance Exercise on Skeletal Muscle Transcriptome Profiles" International Journal of Molecular Sciences 25, no. 5: 2881. https://doi.org/10.3390/ijms25052881

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop