Int J Sports Med 2024; 45(03): 171-182
DOI: 10.1055/a-2153-9258
Review

Autophagy and Exercise: Current Insights and Future Research Directions

Javier Botella
1   Metabolic Research Unit, School of Medicine and Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Waurn Ponds, Victoria, Australia
,
Christopher S Shaw
2   Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, 3216, VIC, Australia
,
David J Bishop
3   Institute for Health and Sport (iHeS), Victoria University, Melbourne, Australia
› Author Affiliations

Abstract

Autophagy is a cellular process by which proteins and organelles are degraded inside the lysosome. Exercise is known to influence the regulation of autophagy in skeletal muscle. However, as gold standard techniques to assess autophagy flux in vivo are restricted to animal research, important gaps remain in our understanding of how exercise influences autophagy activity in humans. Using available datasets, we show how the gene expression profile of autophagy receptors and ATG8 family members differ between human and mouse skeletal muscle, providing a potential explanation for their differing exercise-induced autophagy responses. Furthermore, we provide a comprehensive view of autophagy regulation following exercise in humans by summarizing human transcriptomic and phosphoproteomic datasets that provide novel targets of potential relevance. These newly identified phosphorylation sites may provide an explanation as to why both endurance and resistance exercise lead to an exercise-induced reduction in LC3B-II, while possibly divergently regulating autophagy receptors, and, potentially, autophagy flux. We also provide recommendations to use ex vivo autophagy flux assays to better understand the influence of exercise, and other stimuli, on autophagy regulation in humans. This review provides a critical overview of the field and directs researchers towards novel research areas that will improve our understanding of autophagy regulation following exercise in humans.



Publication History

Received: 22 November 2022

Accepted: 14 August 2023

Accepted Manuscript online:
15 August 2023

Article published online:
22 September 2023

© 2023. Thieme. All rights reserved.

Georg Thieme Verlag
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Takeshige K, Baba M, Tsuboi S. et al. Autophagy in yeast demonstrated with proteinase-deficient mutants and conditions for its induction. J Cell Biol 1992; 119: 301-311
  • 2 Li WW, Li J, Bao JK. Microautophagy: Lesser-known self-eating. Cell Mol Life Sci 2012; 69: 1125-1136
  • 3 Dice JF. Chaperone-mediated autophagy. Autophagy 2007; 3: 295-299
  • 4 Feng Y, He D, Yao Z. et al. The machinery of macroautophagy. Cell Res 2014; 24: 24-41
  • 5 Ohsumi Y. Historical landmarks of autophagy research. Cell Res 2014; 24: 9-23
  • 6 Bordi M, De Cegli R, Testa B. et al. A gene toolbox for monitoring autophagy transcription. Cell Death Dis 2021; 12: 1-7
  • 7 Varusai TM, Jupe S, Sevilla C. et al. Using Reactome to build an autophagy mechanism knowledgebase. Autophagy 2021; 17: 1543-1554
  • 8 Melia TJ, Lystad AH, Simonsen A. Autophagosome biogenesis: From membrane growth to closure. J Cell Biol 2020; 219
  • 9 Kim J, Kundu M, Viollet B. et al. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol 2011; 13: 132-141
  • 10 Mailler E, Guardia CM, Bai X. et al. The autophagy protein ATG9A enables lipid mobilization from lipid droplets. Nat Commun 2021; 12: 6750
  • 11 Nguyen TN, Lazarou M. A unifying model for the role of the ATG8 system in autophagy. J Cell Sci. 2022 135.
  • 12 Klionsky DJ, Abdel-Aziz AK, Abdelfatah S. et al. Guidelines for the use and interpretation of assays for monitoring autophagy (4th edition)(1). Autophagy 2021; 17: 1-382
  • 13 Mercer TJ, Gubas A, Tooze SA. A molecular perspective of mammalian autophagosome biogenesis. J Biol Chem 2018; 293: 5386-5395
  • 14 Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med 2013; 368: 651-662
  • 15 Aman Y, Schmauck-Medina T, Hansen M. et al. Autophagy in healthy aging and disease. Nat Aging 2021; 1: 634-650
  • 16 Martin-Rincon M, Morales-Alamo D, Calbet J. Exercise-mediated modulation of autophagy in skeletal muscle. Scand J Med Sci Sports 2018; 28: 772-781
  • 17 Yoshii SR, Mizushima N. Monitoring and Measuring Autophagy. Int J Mol Sci 2017; 18: 1865
  • 18 Bonam SR, Bayry J, Tschan MP. et al. Progress and Challenges in the Use of MAP1LC3 as a Legitimate Marker for Measuring Dynamic Autophagy In Vivo. Cells 2020; 9: 1321
  • 19 De Palma C, Morisi F, Cheli S. et al. Autophagy as a new therapeutic target in Duchenne muscular dystrophy. Cell Death Dis 2012; 3: e418
  • 20 Grumati P, Coletto L, Sabatelli P. et al. Autophagy is defective in collagen VI muscular dystrophies, and its reactivation rescues myofiber degeneration. Nat Med 2010; 16: 1313-1320
  • 21 Hargreaves M, Spriet LL. Skeletal muscle energy metabolism during exercise. Nat Metab 2020; 2: 817-828
  • 22 He C, Bassik MC, Moresi V. et al. Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature 2012; 481: 511-515
  • 23 Lo Verso F, Carnio S, Vainshtein A. et al. Autophagy is not required to sustain exercise and PRKAA1/AMPK activity but is important to prevent mitochondrial damage during physical activity. Autophagy 2014; 10: 1883-1894
  • 24 Call JA, Wilson RJ, Laker RC. et al. Ulk1-mediated autophagy plays an essential role in mitochondrial remodeling and functional regeneration of skeletal muscle. Am J Physiol, Cell Physiol 2017; 312: C724-C732
  • 25 Lira VA, Okutsu M, Zhang M. et al. Autophagy is required for exercise training-induced skeletal muscle adaptation and improvement of physical performance. FASEB J 2013; 27: 4184-4193
  • 26 Masiero E, Agatea L, Mammucari C. et al. Autophagy Is Required to Maintain Muscle Mass. Cell Metab 2009; 10: 507-515
  • 27 Schiaffino S, Reggiani C. Fiber types in mammalian skeletal muscles. Physiol Rev 2011; 91: 1447-1531
  • 28 Mizushima N, Yamamoto A, Matsui M. et al. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol Biol Cell 2004; 15: 1101-1111
  • 29 Ogata T, Oishi Y, Higuchi M. et al. Fasting-related autophagic response in slow- and fast-twitch skeletal muscle. Biochem Biophys Res Commun 2010; 394: 136-140
  • 30 Mofarrahi M, Guo Y, Haspel JA. et al. Autophagic flux and oxidative capacity of skeletal muscles during acute starvation. Autophagy 2013; 9: 1604-1620
  • 31 Paré MF, Baechler BL, Fajardo VA. et al. Effect of acute and chronic autophagy deficiency on skeletal muscle apoptotic signaling, morphology, and function. Biochim Biophys Acta Mol Cell Res 2017; 1864: 708-718
  • 32 Raben N, Hill V, Shea L. et al. Suppression of autophagy in skeletal muscle uncovers the accumulation of ubiquitinated proteins and their potential role in muscle damage in Pompe disease. Hum Mol Genet 2008; 17: 3897-3908
  • 33 Wang Y, Pessin JE. Mechanisms for fiber-type specificity of skeletal muscle atrophy. Curr Opin Clin Nutr Metab Care 2013; 16: 243-250
  • 34 Mofarrahi M, Guo Y, Haspel JA. et al. Autophagic flux and oxidative capacity of skeletal muscles during acute starvation. Autophagy 2013; 9: 1604-1620
  • 35 Murgia M, Nogara L, Baraldo M. et al. Protein profile of fiber types in human skeletal muscle: A single-fiber proteomics study. Skeletal Muscle 2021; 11: 24
  • 36 Murgia M, Nogara L, Baraldo M. et al. Protein profile of fiber types in human skeletal muscle: A single-fiber proteomics study. Skelet Muscle 2021; 11: 1-19
  • 37 Murgia M, Nagaraj N, Deshmukh AS. et al. Single muscle fiber proteomics reveals unexpected mitochondrial specialization. EMBO reports 2015; 16: 387-395
  • 38 Deshmukh AS, Steenberg DE, Hostrup 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
  • 39 Morales-Scholz MG, Wette SG, Stokie JR. et al. Muscle fibre-type specific autophagy responses following an overnight fast and mixed meal ingestion in human skeletal muscle. Am J Physiol Endocrinol Metab 2022; 323: E242-E253
  • 40 Reggiori F, Klionsky DJ. Autophagy in the eukaryotic cell. Eukaryotic cell 2002; 1: 11-21
  • 41 Ham DJ, Börsch A, Lin S. et al. The neuromuscular junction is a focal point of mTORC1 signaling in sarcopenia. Nat Commun 2020; 11: 4510
  • 42 Lonsdale J, Thomas J, Salvatore M. et al. The Genotype-Tissue Expression (GTEx) project. Nat Genet 2013; 45: 580-585
  • 43 Laker RC, Drake JC, Wilson RJ. et al. Ampk phosphorylation of Ulk1 is required for targeting of mitochondria to lysosomes in exercise-induced mitophagy. Nat Commun 2017; 8: 1-13
  • 44 Brandt N, Dethlefsen MM, Bangsbo J. et al. PGC-1α and exercise intensity dependent adaptations in mouse skeletal muscle. PloS one 2017; 12: e0185993
  • 45 Wang P, Li CG, Zhou X. et al. A single bout of exhaustive treadmill exercise increased AMPK activation associated with enhanced autophagy in mice skeletal muscle. Clin Exp Parmacol Physiol 2022; 49: 536-543
  • 46 Vainshtein A, Tryon LD, Pauly M. et al. Role of PGC-1α during acute exercise-induced autophagy and mitophagy in skeletal muscle. Am J Physiol, Cell Physiol 2015; 308: C710-C719
  • 47 Botella J, Jamnick NA, Granata C. et al. Exercise and Training Regulation of Autophagy Markers in Human and Rat Skeletal Muscle. Int J Mol Sci 2022; 23: 2619
  • 48 Fritzen AM, Madsen AB, Kleinert M. et al. Regulation of autophagy in human skeletal muscle: Effects of exercise, exercise training and insulin stimulation. J Physiol 2016; 594: 745-761
  • 49 Grumati P, Coletto L, Schiavinato A. et al. Physical exercise stimulates autophagy in normal skeletal muscles but is detrimental for collagen VI-deficient muscles. Autophagy 2011; 7: 1415-1423
  • 50 Zhang D, Lee JH, Kwak SE. et al. Effect of a Single Bout of Exercise on Autophagy Regulation in Skeletal Muscle of High-Fat High-Sucrose Diet-Fed Mice. J Obes Metab Syndr 2019; 28: 175-185
  • 51 Jamart C, Naslain D, Gilson H. et al. Higher activation of autophagy in skeletal muscle of mice during endurance exercise in the fasted state. Am J Physiol Endocrinol Metab 2013; 305: E964-E974
  • 52 Kruse R, Pedersen AJ, Kristensen JM. et al. Intact initiation of autophagy and mitochondrial fission by acute exercise in skeletal muscle of patients with Type 2 diabetes. Clin Sci (Lond) 2017; 131: 37-47
  • 53 Schwalm C, Jamart C, Benoit N. et al. Activation of autophagy in human skeletal muscle is dependent on exercise intensity and AMPK activation. FASEB J 2015; 29: 3515-3526
  • 54 Møller AB, Vendelbo MH, Christensen B. et al. Physical exercise increases autophagic signaling through ULK1 in human skeletal muscle. J Appl Physiol (1985) 2015; 118: 971-979
  • 55 Moberg M, Hendo G, Jakobsson M. et al. Increased autophagy signaling but not proteasome activity in human skeletal muscle after prolonged low-intensity exercise with negative energy balance. Physiol Rep 2017; 5: e13518
  • 56 Arribat Y, Broskey NT, Greggio C. et al. Distinct patterns of skeletal muscle mitochondria fusion, fission and mitophagy upon duration of exercise training. Acta Physiol (Oxf) 2019; 225: e13179
  • 57 Brandt N, Gunnarsson TP, Bangsbo J. et al. Exercise and exercise training-induced increase in autophagy markers in human skeletal muscle. Physiol Rep 2018; 6: e13651
  • 58 Fritzen AM, Madsen AB, Kleinert M. et al. Regulation of autophagy in human skeletal muscle: Effects of exercise, exercise training and insulin stimulation. J Physiol 2016; 594: 745-761
  • 59 Billat VL, Sirvent P, Py G. et al. The concept of maximal lactate steady state: A bridge between biochemistry, physiology and sport science. Sports Med 2003; 33: 407-426
  • 60 Pillon NJ, Gabriel BM, Dollet L. et al. Transcriptomic profiling of skeletal muscle adaptations to exercise and inactivity. Nat Commun 2020; 11: 470
  • 61 Blazev R, Carl CS, Ng Y-K. et al. Phosphoproteomics of three exercise modalities identifies canonical signaling and C18ORF25 as an AMPK substrate regulating skeletal muscle function. Cell Metab 2022; 34: 1561-1577
  • 62 Hoffman NJ, Parker BL, Chaudhuri R. et al. Global Phosphoproteomic Analysis of Human Skeletal Muscle Reveals a Network of Exercise-Regulated Kinases and AMPK Substrates. Cell Metab 2015; 22: 922-935
  • 63 Papinski D, Schuschnig M, Reiter W. et al. Early steps in autophagy depend on direct phosphorylation of Atg9 by the Atg1 kinase. Mol Cell 2014; 53: 471-483
  • 64 Linares Juan F, Duran A, Reina-Campos M. et al. Amino Acid Activation of mTORC1 by a PB1-Domain-Driven Kinase Complex Cascade. Cell Reports 2015; 12: 1339-1352
  • 65 Richter B, Sliter DA, Herhaus L. et al. Phosphorylation of OPTN by TBK1 enhances its binding to Ub chains and promotes selective autophagy of damaged mitochondria. Proc Natl Acad Sci USA 2016; 113: 4039-4044
  • 66 Wild P, Farhan H, McEwan DG. et al. Phosphorylation of the autophagy receptor optineurin restricts Salmonella growth. Science 2011; 333: 228-233
  • 67 Zeng Z, Liang J, Wu L. et al. Exercise-induced autophagy suppresses sarcopenia through Akt/mTOR and Akt/FoxO3a signal pathways and AMPK-mediated mitochondrial quality control. Front Physiol 2020; 11: 583478
  • 68 Ju JS, Jeon SI, Park JY. Autophagy plays a role in skeletal muscle mitochondrial biogenesis in an endurance exercise-trained condition. J Physiol Sci 2016; 66: 417-430
  • 69 Cardinale DA, Gejl KD, Petersen KG. et al. Short-term intensified training temporarily impairs mitochondrial respiratory capacity in elite endurance athletes. J Appl Physiol (1985) 2021; 131: 388-400
  • 70 Hulmi JJ, Tannerstedt J, Selänne H. et al. Resistance exercise with whey protein ingestion affects mTOR signaling pathway and myostatin in men. J Appl Physiol 2009; 106: 1720-1729
  • 71 Alers S, Löffler AS, Wesselborg S. et al. Role of AMPK-mTOR-Ulk1/2 in the Regulation of Autophagy: Cross Talk, Shortcuts, and Feedbacks. Mol Cell Biol 2012; 32: 2-11
  • 72 Pinto AP, da Rocha AL, Marafon BB. et al. Impact of different physical exercises on the expression of autophagy markers in mice. Int J Mol Sci 2021; 22: 2635
  • 73 Hentilä J, Ahtiainen JP, Paulsen G. et al. Autophagy is induced by resistance exercise in young men, but unfolded protein response is induced regardless of age. Acta Physiol (Oxf) 2018; 224: e13069
  • 74 Fry CS, Drummond MJ, Glynn EL. et al. Skeletal muscle autophagy and protein breakdown following resistance exercise are similar in younger and older adults. J Gerontol A Bio Sci Med Sci 2013; 68: 599-607
  • 75 Mazo CE, D’Lugos AC, Sweeney KR. et al. The effects of acute aerobic and resistance exercise on mTOR signaling and autophagy markers in untrained human skeletal muscle. Eur J Appl Physiol 2021; 121: 2913-2924
  • 76 Aas SN, Tømmerbakke D, Godager S. et al. Effects of acute and chronic strength training on skeletal muscle autophagy in frail elderly men and women. Exp Gerontol 2020; 142: 111122
  • 77 Ogborn DI, McKay BR, Crane JD. et al. Effects of age and unaccustomed resistance exercise on mitochondrial transcript and protein abundance in skeletal muscle of men. Am J Physiol Regul Integr Comp Physiol 2015; 308: R734-R741
  • 78 Kwon I, Jang Y, Cho J-Y. et al. Long-term resistance exercise-induced muscular hypertrophy is associated with autophagy modulation in rats. J Physiol Sci 2018; 68: 269-280
  • 79 Cui D, Drake JC, Wilson RJ. et al. A novel voluntary weightlifting model in mice promotes muscle adaptation and insulin sensitivity with simultaneous enhancement of autophagy and mTOR pathway. FASEB J 2020; 34: 7330-7344
  • 80 Loos B, Klionsky DJ, Du Toit A. et al. On the relevance of precision autophagy flux control in vivo – Points of departure for clinical translation. Autophagy 2020; 16: 750-762
  • 81 Tian W, Alsaadi R, Guo Z. et al. An antibody for analysis of autophagy induction. Nat Methods 2020; 17: 232-239
  • 82 Kovsan J, Blüher M, Tarnovscki T. et al. Altered Autophagy in Human Adipose Tissues in Obesity. J Clin Endocrinol Metab 2011; 96: E268-E277
  • 83 Xu C, Markova M, Seebeck N. et al. High-protein diet more effectively reduces hepatic fat than low-protein diet despite lower autophagy and FGF21 levels. Liver Int 2020; 40: 2982-2997
  • 84 Martinez-Lopez N, Tarabra E, Toledo M. et al. System-wide benefits of intermeal fasting by autophagy. Cell Metab 2017; 26: 856-871. e855
  • 85 Bensalem J, Hattersley KJ, Hein LK. et al. Measurement of autophagic flux in humans: An optimized method for blood samples. Autophagy 2021; 17: 3238-3255
  • 86 Pietrocola F, Demont Y, Castoldi F. et al. Metabolic effects of fasting on human and mouse blood in vivo. Autophagy 2017; 13: 567-578
  • 87 Sargeant TJ, Bensalem J. Human autophagy measurement: An underappreciated barrier to translation. Trends Mol Med 2021; 27: 1091-1094
  • 88 Lee Y-K, Lee J-A. Role of the mammalian ATG8/LC3 family in autophagy: Differential and compensatory roles in the spatiotemporal regulation of autophagy. BMB Rep 2016; 49: 424
  • 89 Nguyen TN, Padman BS, Usher J. et al. Atg8 family LC3/GABARAP proteins are crucial for autophagosome-lysosome fusion but not autophagosome formation during PINK1/Parkin mitophagy and starvation. J Cell Biol 2016; 215: 857-874
  • 90 He C, Sumpter JR, Levine B. Exercise induces autophagy in peripheral tissues and in the brain. Autophagy 2012; 8: 1548-1551
  • 91 Bayod S, Del Valle J, Pelegri C. et al. Macroautophagic process was differentially modulated by long-term moderate exercise in rat brain and peripheral tissues. J Physiol Pharmacol 2014; 65: 229-239