CC BY-NC-ND 4.0 · Int J Sports Med 2022; 43(03): 195-205
DOI: 10.1055/a-1551-9294
Review

The Molecular Signature of High-intensity Training in the Human Body

Patrick Wahl
1   Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Cologne, Germany
2   The German Research Center of Elite Sport Cologne, German Sport University Cologne, Koln, Germany
3   MSH Medical School Hamburg, Institute of Interdisciplinary Exercise Science and Sports Medicine, Hamburg, Germany
,
Wilhelm Bloch
2   The German Research Center of Elite Sport Cologne, German Sport University Cologne, Koln, Germany
4   Molecular and Cellular Sport Medicine, German Sport university, Cologne, Germany
,
Sebastian Proschinger
5   Department for Molecular and Cellular Sports Medicine, Institute for Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
› Author Affiliations

Abstract

High-intensity training is becoming increasingly popular outside of elite sport for health prevention and rehabilitation. This expanded application of high-intensity training in different populations requires a deeper understanding of its molecular signature in the human body. Therefore, in this integrative review, cellular and systemic molecular responses to high-intensity training are described for skeletal muscle, cardiovascular system, and the immune system as major effectors and targets of health and performance. Different kinds of stimuli and resulting homeostatic perturbations (i. e., metabolic, mechanical, neuronal, and hormonal) are reflected, taking into account their role in the local and systemic deflection of molecular sensors and mediators, and their role in tissue and organ adaptations. In skeletal muscle, a high metabolic perturbation induced by high-intensity training is the major stimulus for skeletal muscle adaptation. In the cardio-vascular system, high-intensity training induces haemodynamic stress and deflection of the Ca 2+ handling as major stimuli for functional and structural adaptation of the heart and vessels. For the immune system haemodynamic stress, hormones, exosomes, and O2 availability are proposed stimuli that mediate their effects by alteration of different signalling processes leading to local and systemic (anti)inflammatory responses. Overall, high-intensity training shows specific molecular signatures that demonstrate its high potential to improve health and physical performance.



Publication History

Received: 07 April 2021

Accepted: 13 July 2021

Accepted Manuscript online:
15 July 2021

Article published online:
12 October 2021

© 2021. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial-License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/).

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Ross LM, Porter RR, Durstine JL. High-intensity interval training (HIIT) for patients with chronic diseases. J Sport Health Sci 2016; 5: 139-144
  • 2 Hoppeler H, Baum O, Lurman G. et al. Molecular mechanisms of muscle plasticity with exercise. Compr Physiol 2011; 1: 1383-1412
  • 3 Ben-Zeev T, Okun E. High-intensity functional training: Molecular mechanisms and benefits. Neuromolecular Med 2021; 3: 335-338
  • 4 Jiménez-Maldonado A, García-Suárez PC, Rentería I. et al. Impact of high-intensity interval training and sprint interval training on peripheral markers of glycemic control in metabolic syndrome and type 2 diabetes. Biochim Biophys Acta Mol Basis Dis 2020; 1866: 165820
  • 5 Gibala MJ, Little JP, van Essen M. et al. Short-term sprint interval versus traditional endurance training: Similar initial adaptations in human skeletal muscle and exercise performance. J Physiol 2006; 575: 901-911
  • 6 Seiler S, Tønnessen E. Intervals, thresholds, and long slow distance: The role of intensity and duration in endurance training. Sportscience 2009; 13: 32-53
  • 7 Buchheit M, Laursen PB. High-intensity interval training, solutions to the programming puzzle: Part I: Cardiopulmonary emphasis. Sports Med 2013; 43: 313-338
  • 8 Yu H, Chen X, Zhu W. et al. A quasi-experimental study of Chinese top-level speed skaters’ training load: Threshold versus polarized model. Int J Sports Physiol Perform 2012; 7: 103-112
  • 9 Stöggl T, Sperlich B. Polarized training has greater impact on key endurance variables than threshold, high intensity, or high volume training. Front Physiol 2014; 5: 33
  • 10 Wackerhage H. Molecules ageing and exercise. In: Wackerhage H. (ed) Muscular Exercise Physiology: An Introduction. London: Routledge; 2014
  • 11 Laursen PB. Training for intense exercise performance: High-intensity or high-volume training?. Scand J Med Sci Sports 2010; 20: 1-10
  • 12 Egan B, Zierath JR. Exercise metabolism and the molecular regulation of skeletal muscle adaptation. Cell Metab 2013; 17: 162-184
  • 13 O'Neill HM. AMPK and exercise: Glucose uptake and insulin sensitivity. Diabetes Metab J 2013; 37: 1-21
  • 14 Bae HR, Kim DH, Park MH. et al. β-Hydroxybutyrate suppresses inflammasome formation by ameliorating endoplasmic reticulum stress via AMPK activation. Oncotarget 2016; 7: 66444-66454
  • 15 Liu JQ, Zhang L, Yao J. et al. AMPK alleviates endoplasmic reticulum stress by inducing the ER-chaperone ORP150 via FOXO1 to protect human bronchial cells from apoptosis. Biochem Biophys Res Commun 2018; 497: 564-570
  • 16 Dasgupta B, Chhipa RR. Evolving lessons on the complex role of ampk in normal physiology and cancer. Trends Pharmacol Sci 2016; 37: 192-206
  • 17 Arany Z, Foo S-Y, Ma Y. et al. HIF-independent regulation of VEGF and angiogenesis by the transcriptional coactivator PGC-1alpha. Nature 2008; 451: 1008-1012
  • 18 Gavin TP, Ruster RS, Carrithers JA. et al. No difference in the skeletal muscle angiogenic response to aerobic exercise training between young and aged men. J Physiol 2007; 585: 231-239
  • 19 Hoier B, Nordsborg N, Andersen S. et al. Pro-and anti-angiogenic factors in human skeletal muscle in response to acute exercise and training. J Physiol 2012; 590: 595-606
  • 20 Jensen L, Bangsbo J, Hellsten Y. Effect of high intensity training on capillarization and presence of angiogenic factors in human skeletal muscle. J Physiol 2004; 557: 571-582
  • 21 Klausen K, Andersen LB, Pelle I. Adaptive changes in work capacity, skeletal muscle capillarization and enzyme levels during training and detraining. Acta Physiol Scand 1981; 113: 9-16
  • 22 Wackerhage H. Molecular Exercise Physiology: An Introduction. London: Routledge; 2014
  • 23 Popov DV. Adaptation of skeletal muscles to contractile activity of varying duration and intensity: The role of PGC-1α. Biochemistry (Mosc) 2018; 83: 613-628
  • 24 Gibala M. Molecular responses to high-intensity interval exercise. Appl Physiol Nutr Metab 2009; 34: 428-432
  • 25 Kupr B, Handschin C. Complex coordination of cell plasticity by a PGC-1α-controlled transcriptional network in skeletal muscle. Front Physiol 2015; 6: 325
  • 26 Lira VA, Benton CR, Yan Z. et al. PGC-1alpha regulation by exercise training and its influences on muscle function and insulin sensitivity. Am J Physiol Endocrinol Metab 2010; 299: E145-E161
  • 27 Little JP, Safdar A, Cermak N. et al. Acute endurance exercise increases the nuclear abundance of PGC-1alpha in trained human skeletal muscle. Am J Physiol Regul Integr Comp Physiol 2010; 298: R912-R917
  • 28 Wright DC, Han D-H, Garcia-Roves PM. et al. Exercise-induced mitochondrial biogenesis begins before the increase in muscle PGC-1alpha expression. J Biol Chem 2007; 282: 194-199
  • 29 Smith BK, Mukai K, Lally JS. et al. AMP-activated protein kinase is required for exercise-induced peroxisome proliferator-activated receptor co-activator 1 translocation to subsarcolemmal mitochondria in skeletal muscle. J Physiol 2013; 591: 1551-1561
  • 30 Coccimiglio IF, Clarke DC. ADP is the dominant controller of AMP-activated protein kinase activity dynamics in skeletal muscle during exercise. PLoS Comput Biol 2020; 16: e1008079
  • 31 Egan B, Carson BP, Garcia-Roves PM. et al. Exercise intensity-dependent regulation of peroxisome proliferator-activated receptor coactivator-1 mRNA abundance is associated with differential activation of upstream signalling kinases in human skeletal muscle. J Physiol 2010; 588: 1779-1790
  • 32 Di Donato DM, West DWD, Churchward-Venne TA. et al. Influence of aerobic exercise intensity on myofibrillar and mitochondrial protein synthesis in young men during early and late postexercise recovery. Am J Physiol Endocrinol Metab 2014; 306: E1025-E1032
  • 33 Combes A, Dekerle J, Webborn N. et al. Exercise-induced metabolic fluctuations influence AMPK, p38-MAPK and CaMKII phosphorylation in human skeletal muscle. Physiol Rep 2015; 3: e12462
  • 34 Bartlett JD, Hwa Joo C, Jeong T-S. et al. Matched work high-intensity interval and continuous running induce similar increases in PGC-1α mRNA, AMPK, p38, and p53 phosphorylation in human skeletal muscle. J Appl Physiol 1985 2012: 1135-1143
  • 35 Cochran AJR, Percival ME, Tricarico S. et al. Intermittent and continuous high-intensity exercise training induce similar acute but different chronic muscle adaptations. Exp Physiol 2014; 99: 782-791
  • 36 Gibala MJ, McGee SL, Garnham AP. et al. Brief intense interval exercise activates AMPK and p38 MAPK signaling and increases the expression of PGC-1alpha in human skeletal muscle. J Appl Physiol 1985 2009: 929-934
  • 37 Coffey VG, Zhong Z, Shield A. et al. Early signaling responses to divergent exercise stimuli in skeletal muscle from well-trained humans. FASEB J 2006; 20: 190-192
  • 38 Watt MJ, Southgate RJ, Holmes AG. et al. Suppression of plasma free fatty acids upregulates peroxisome proliferator-activated receptor (PPAR) alpha and delta and PPAR coactivator 1alpha in human skeletal muscle, but not lipid regulatory genes. J Mol Endocrinol 2004; 33: 533-544
  • 39 Burgomaster KA, Howarth KR, Phillips SM. et al. Similar metabolic adaptations during exercise after low volume sprint interval and traditional endurance training in humans. J Physiol 2008; 586: 151-160
  • 40 Little JP, Safdar A, Wilkin GP. et al. A practical model of low-volume high-intensity interval training induces mitochondrial biogenesis in human skeletal muscle: Potential mechanisms. J Physiol 2010; 588: 1011-1022
  • 41 Hennig R, Lømo T. Firing patterns of motor units in normal rats. Nature 1985; 314: 164-166
  • 42 Lee-Young RS, Canny BJ, Myers DE. et al. AMPK activation is fiber type specific in human skeletal muscle: Effects of exercise and short-term exercise training. J Appl Physiol (1985) 2009; 107: 283-289
  • 43 Kristensen DE, Albers PH, Prats C. et al. Human muscle fibre type-specific regulation of AMPK and downstream targets by exercise. J Physiol 2015; 593: 2053-2069
  • 44 Russell AP, Feilchenfeldt J, Schreiber S. et al. Endurance training in humans leads to fiber type-specific increases in levels of peroxisome proliferator-activated receptor-gamma coactivator-1 and peroxisome proliferator-activated receptor-alpha in skeletal muscle. Diabetes 2003; 52: 2874-2881
  • 45 Kemi OJ, Ellingsen Ø, Ceci M. et al. Aerobic interval training enhances cardiomyocyte contractility and Ca2+cycling by phosphorylation of CaMKII and Thr-17 of phospholamban. J Mol Cell Cardiol 2007; 43: 354-361
  • 46 Wisløff U, Loennechen JP, Falck G. et al. Increased contractility and calcium sensitivity in cardiac myocytes isolated from endurance trained rats. Cardiovasc Res 2001; 50: 495-508
  • 47 Stølen TO, Høydal MA, Kemi OJ. et al. Interval training normalizes cardiomyocyte function, diastolic Ca 2+control, and SR Ca2+release synchronicity In a mouse model of diabetic cardiomyopathy. Circ Res 2009; 105: 527-536
  • 48 Cassidy S, Thoma C, Houghton D. et al. High-intensity interval training: A review of its impact on glucose control and cardiometabolic health. Diabetologia 2017; 60: 7-23
  • 49 Gunadi JW, Tarawan VM, Setiawan I. et al. Cardiac hypertrophy is stimulated by altered training intensity and correlates with autophagy modulation in male Wistar rats. BMC Sports Sci Med Rehabil 2019; 11: 9
  • 50 Ghanimati R, Rajabi H, Ramezani F. et al. The effect of preconditioning with high-intensity training on tissue levels of G-CSF, its receptor and C-kit after an acute myocardial infarction in male rats. BMC Cardiovasc Disord 2020; 20: 75
  • 51 Naderi N, Hemmatinafar M, Gaeini AA. et al. High-intensity interval training increase GATA4, CITED4 and c-Kit and decreases C/EBPβ in rats after myocardial infarction. Life Sci 2019; 221: 319-326
  • 52 Fallahi A, Gaeini A, Shekarfroush S. et al. Cardioprotective effect of high intensity interval training and nitric oxide metabolites (NO2  , NO3  ). Iran J Public Health 2015; 44: 1270-1276
  • 53 Novoa U, Arauna D, Moran M. et al. High-intensity exercise reduces cardiac fibrosis and hypertrophy but does not restore the nitroso-redox imbalance in diabetic cardiomyopathy. Oxid Med Cell Longev 2017; 2017: 7921363
  • 54 Schmitz B, Breulmann FL, Jubran B. et al. A three-step approach identifies novel shear stress-sensitive endothelial microRNAs involved in vasculoprotective effects of high-intensity interval training (HIIT). Oncotarget 2019; 10: 3625-3640
  • 55 Wahl P, Wehmeier UF, Jansen FJ. et al. Acute effects of different exercise protocols on the circulating vascular microRNAs -16, -21, and -126 in trained subjects. Front Physiol 2016; 7: 643
  • 56 Wahl P, Jansen F, Achtzehn S. et al. Effects of high intensity training and high volume training on endothelial microparticles and angiogenic growth factors. PLoS One 2014; 9: e96024
  • 57 Adams V, Reich B, Uhlemann M. et al. Molecular effects of exercise training in patients with cardiovascular disease: Focus on skeletal muscle, endothelium, and myocardium. Am J Physiol Heart Circ Physiol 2017; 313: H72-H88
  • 58 Schmitz B, Niehues H, Lenders M. et al. Effects of high-intensity interval training on microvascular glycocalyx and associated microRNAs. Am J Physiol Heart Circ Physiol 2019; 316: H1538-H1551
  • 59 Adams V, Alves M, Fischer T. et al. High-intensity interval training attenuates endothelial dysfunction in a Dahl salt-sensitive rat model of heart failure with preserved ejection fraction. J Appl Physiol (1985) 2015; 119: 745-752
  • 60 Tsai HH, Lin CP, Lin YH. et al. High-intensity Interval training enhances mobilization/functionality of endothelial progenitor cells and depressed shedding of vascular endothelial cells undergoing hypoxia. Eur J Appl Physiol 2016; 116: 2375-2388
  • 61 Neufer PD, Bamman MM, Muoio DM. et al. understanding the cellular and molecular mechanisms of physical activity-induced health benefits. Cell Metab 2015; 22: 4-11
  • 62 Walsh NP, Gleeson MM, Shephard RJ. et al. Institutional Repository Position statement part one : Immune function and exercise. Exerc Immunol Rev 2011; 17: 6-63
  • 63 Krüger K, Alack K, Ringseis R. et al. Apoptosis of T-Cell subsets after acute high-intensity interval exercise. Med Sci Sports Exerc 2016; 48: 2021-2029
  • 64 Wadley AJ, Cullen T, Vautrinot J. et al. High intensity interval exercise increases the frequency of peripheral PD-1 + CD8 + central memory T-cells and soluble PD-L1 in humans. Brain Behav Immun Health 2020; 3: 100049
  • 65 Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J Clin Oncol 2015; 33: 1974-1982
  • 66 Schenk A, Joisten N, Walzik D. et al. Acute exercise impacts AhR and PD-1 levels of CD8 + T-cells—Exploratory results from a randomized cross-over trial comparing endurance versus resistance exercise. Eur J Appl Physiol 2020; 121: 637-644
  • 67 Radak Z, Ishihara K, Tekus E. et al. Exercise, oxidants, and antioxidants change the shape of the bell-shaped hormesis curve. Redox Biol 2017; 12: 285-290
  • 68 Krüger K, Mooren FC. Exercise-induced leukocyte apoptosis. Exerc Immunol Rev 2014; 20: 117-134
  • 69 Muri J, Kopf M. Redox regulation of immunometabolism. Nat Rev Immunol 2020; 21: 363-381
  • 70 Jamurtas AZ, Fatouros IG, Deli CK. et al. The effects of acute low-volume HIIT and aerobic exercise on leukocyte count and redox status. J Sports Sci Med 2018; 17: 501-508
  • 71 Fisher G, Schwartz DD, Quindry J. et al. Lymphocyte enzymatic antioxidant responses to oxidative stress following high-intensity interval exercise. J Appl Physiol (1985) 2011; 110: 730-737
  • 72 De Oliveira Ottone V, Costa KB, Tossige-Gomes R. et al. Late neutrophil priming following a single session of high-intensity interval exercise. Int J Sports Med 2019; 40: 171-179
  • 73 Bartlett DB, Shepherd SO, Wilson OJ. et al. Neutrophil and monocyte bactericidal responses to 10 weeks of low-volume high-intensity interval or moderate-intensity continuous training in sedentary adults. Oxid Med Cell Longev 2017; 2017: 8148742
  • 74 Durrer C, Francois M, Neudorf H. et al. Acute high-intensity interval exercise reduces human monocyte toll-like receptor 2 expression in type 2 diabetes. Am J Physiol Regul Integr Comp Physiol 2017; 312: R529-R538
  • 75 Robinson E, Durrer C, Simtchouk S. et al. Short-term high-intensity interval and moderate-intensity continuous training reduce leukocyte TLR4 in inactive adults at elevated risk of type 2 diabetes. J Appl Physiol (1985) 2015; 119: 508-516
  • 76 Joisten N, Proschinger S, Rademacher A. et al. High-intensity interval training reduces neutrophil-to-lymphocyte ratio in persons with multiple sclerosis during inpatient rehabilitation. Mult Scler 2021; 27: 1136–1139
  • 77 Wahl P, Mathes S, Bloch W. et al. Acute impact of recovery on the restoration of cellular immunological homeostasis. Int J Sports Med 2020; 41: 12-20
  • 78 Furman D, Campisi J, Verdin E. et al. Chronic inflammation in the etiology of disease across the life span. Nat Med 2019; 25: 1822-1832
  • 79 Horn A, Van der Meulen JH, Defour A. et al. Mitochondrial redox signaling enables repair of injured skeletal muscle cells. Sci Signal 2017; 10: eaaj1978
  • 80 Schieber M, Chandel NS. ROS function in redox signaling and oxidative stress. Curr Biol 2014; 24: 453-462
  • 81 Louzada RA, Bouviere J, Matta LP. et al. Redox signaling in widespread health benefits of exercise. Antioxid Redox Signal 2020. Online ahead of print. DOI: 10.1089/ars.2019.7949
  • 82 Parker L, Stepto NK, Shaw CS. et al. Acute high-intensity interval exercise-induced redox signaling is associated with enhanced insulin sensitivity in obese middle-aged men. Front Physiol 2016; 7: 411
  • 83 Bartlett DB, Willis LH, Slentz CA. et al. Ten weeks of high-intensity interval walk training is associated with reduced disease activity and improved innate immune function in older adults with rheumatoid arthritis: A pilot study. Arthritis Res Ther 2018; 20: 127