Molecular Mechanisms of Muscle Fatigue: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Muscle fatigue (MF) declines the capacity of muscles to complete a task over time at a constant load. MF is usually short-lasting, reversible, and is experienced as a feeling of tiredness or lack of energy. The leading causes of short-lasting fatigue are related to overtraining, undertraining/deconditioning, or physical injury. Conversely, MF can be persistent and more serious when associated with pathological states or following chronic exposure to certain medication or toxic composites. In conjunction with chronic fatigue, the muscle feels floppy, and the force generated by muscles is always low, causing the individual to feel frail constantly. The leading cause underpinning the development of chronic fatigue is related to muscle wasting mediated by aging, immobilization, insulin resistance (through high-fat dietary intake or pharmacologically mediated Peroxisome Proliferator-Activated Receptor (PPAR) agonism), diseases associated with systemic inflammation (arthritis, sepsis, infections, trauma, cardiovascular and respiratory disorders (heart failure, chronic obstructive pulmonary disease (COPD))), chronic kidney failure, muscle dystrophies, muscle myopathies, multiple sclerosis, and, more recently, coronavirus disease 2019 (COVID-19). The primary outcome of displaying chronic muscle fatigue is a poor quality of life. 

  • neuroinflammation
  • skeletal muscle
  • atrophy
  • muscle function
  • fatigue

1. Muscle Architecture

The musculoskeletal system is one of the central organ systems in the body. It consists of muscles, tendons, cartilage, ligaments, connective tissues, and nerves.
There are three main types of muscle tissue: skeletal, cardiac, and smooth [1]. Skeletal muscles are fibrous tissues found in humans or animals mainly attached by tendons to the skeleton’s bones. They can contract/shorten upon neuro-mediated calcium stimulation, thereby moving the whole body while maintaining the position of parts of the body.
A skeletal muscle is made up of multiple fascicles, and each one includes numerous muscle fibers (Figure 1, [2]). The muscle fibers are, in turn, composed of myofibrils. The myofibrils are composed of overlapping, protein-made, thick (myosin) and thin (actin) myofilaments highly organized as sarcomere units, which are de facto the contractile units of the muscle. The sheaths made of connective tissue that encapsulate the bundle of myofibrils, muscle fibers, and the outer side of the muscle are named endomysium, perimysium, and epimysium, respectively.
Figure 1. Muscle organization (A. Bonetta and LF Bonewald, originally adapted from Servier Medical Art—https://smart.servier.com/smart_image/tendon-anatomy/ accessed on 1 October 2021) [2].

This entry is adapted from the peer-reviewed paper 10.3390/ijms222111587

References

  1. 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.
  2. Bonetto, A.; Bonewald, L.F. Bone and muscle. In Basic and Applied Bone Biology; Academic Press: Cambridge, MA, USA, 2019; pp. 317–332.
  3. Wan, J.J.; Qin, Z.; Wang, P.Y.; Sun, Y.; Liu, X. Muscle fatigue: General understanding and treatment. Exp. Mol. Med. 2017, 49, e384.
  4. Gandevia, S.C. Spinal and supraspinal factors in human muscle fatigue. Physiol. Rev. 2001, 81, 1725–1789.
  5. Bigland-Ritchie, B.; Jones, D.A.; Hosking, G.P.; Edwards, R.H. Central and peripheral fatigue in sustained maximum voluntary contractions of human quadriceps muscle. Clin. Sci. Mol. Med. 1978, 54, 609–614.
  6. Alkner, B.A.; Tesch, P.A. Knee extensor and plantar flexor muscle size and function following 90 days of bed rest with or without resistance exercise. Eur. J. Appl. Physiol. 2004, 93, 294–305.
  7. Reid, W.D.; MacGowan, N.A. Respiratory muscle injury in animal models and humans. Mol. Cell. Biochem. 1998, 179, 63–80.
  8. Constantin, D.; Menon, M.K.; Houchen-Wolloff, L.; Morgan, M.D.; Singh, S.J.; Greenhaff, P.; Steiner, M.C. Skeletal muscle molecular responses to resistance training and dietary supplementation in COPD. Thorax 2013, 68, 625–633.
  9. Jagoe, R.T.; Goldberg, A.L. What do we really know about the ubiquitin-proteasome pathway in muscle atrophy? Curr. Opin. Clin. Nutr. Metab. Care 2001, 4, 183–190.
  10. Bodine, S.C.; Latres, E.; Baumhueter, S.; Lai, V.K.; Nunez, L.; Clarke, B.A.; Poueymirou, W.T.; Panaro, F.J.; Na, E.; Dharmarajan, K.; et al. Identification of ubiquitin ligases required for skeletal muscle atrophy. Science 2001, 294, 1704–1708.
  11. Rabuel, C.; Renaud, E.; Brealey, D.; Ratajczak, P.; Damy, T.; Alves, A.; Habib, A.; Singer, M.; Payen, D.; Mebazaa, A. Human septic myopathy: Induction of cyclooxygenase, heme oxygenase and activation of the ubiquitin proteolytic pathway. Anesthesiology 2004, 101, 583–590.
  12. Constantin, D.; McCullough, J.; Mahajan, R.P.; Greenhaff, P.L. Novel events in the molecular regulation of muscle mass in critically ill patients. J. Physiol. 2011, 589, 3883–3895.
  13. Klaude, M.; Mori, M.; Tjader, I.; Gustafsson, T.; Wernerman, J.; Rooyackers, O. Protein metabolism and gene expression in skeletal muscle of critically ill patients with sepsis. Clin. Sci. 2012, 122, 133–142.
  14. McKeran, R.O.; Halliday, D.; Purkiss, P.; Royston, P. 3-Methylhistidine excretion as an index of myofibrillar protein catabolism in neuromuscular disease. J. Neurol. Neurosurg. Psychiatry 1979, 42, 536–541.
  15. Jespersen, J.G.; Nedergaard, A.; Reitelseder, S.; Mikkelsen, U.R.; Dideriksen, K.J.; Agergaard, J.; Kreiner, F.; Pott, F.C.; Schjerling, P.; Kjaer, M. Activated protein synthesis and suppressed protein breakdown signaling in skeletal muscle of critically ill patients. PLoS ONE 2011, 6, e18090.
  16. Winkelman, C. Inactivity and inflammation in the critically ill patient. Crit. Care Clin. 2007, 23, 21–34.
  17. Baracos, V.E.; DeVivo, C.; Hoyle, D.H.; Goldberg, A.L. Activation of the ATP-ubiquitin-proteasome pathway in skeletal muscle of cachectic rats bearing a hepatoma. Am. J. Physiol. 1995, 268 Pt 1, E996–E1006.
  18. Guarnieri, G.; Toigo, G.; Situlin, R.; Del Bianco, M.A.; Crapesi, L. Cathepsin B and D activity in human skeletal muscle in disease states. Adv. Exp. Med. Biol. 1988, 240, 243–256.
  19. Wagenmakers, A.J. Muscle function in critically ill patients. Clin. Nutr. 2001, 20, 451–454.
  20. Costelli, P.; Reffo, P.; Penna, F.; Autelli, R.; Bonelli, G.; Baccino, F.M. Ca(2+)-dependent proteolysis in muscle wasting. Int. J. Biochem. Cell. Biol. 2005, 37, 2134–2146.
  21. Lee, S.J. Regulation of muscle mass by myostatin. Annu. Rev. Cell. Dev. Biol. 2004, 20, 61–86.
  22. Morissette, M.R.; Cook, S.A.; Buranasombati, C.; Rosenberg, M.A.; Rosenzweig, A. Myostatin inhibits IGF-I-induced myotube hypertrophy through Akt. Am. J. Physiol. Cell Physiol. 2009, 297, C1124–C1132.
  23. McFarlane, C.; Plummer, E.; Thomas, M.; Hennebry, A.; Ashby, M.; Ling, N.; Smith, H.; Sharma, M.; Kambadur, R. Myostatin induces cachexia by activating the ubiquitin proteolytic system through an NF-kappaB-independent, FoxO1-dependent mechanism. J. Cell Physiol. 2006, 209, 501–514.
  24. Siu, P.M.; Alway, S.E. Mitochondria-associated apoptotic signalling in denervated rat skeletal muscle. J. Physiol. 2005, 565 Pt 1, 309–323.
  25. Schwartz, L.M. Skeletal Muscles Do Not Undergo Apoptosis During Either Atrophy or Programmed Cell Death-Revisiting the Myonuclear Domain Hypothesis. Front. Physiol. 2018, 9, 1887.
  26. Sandri, M.; Sandri, C.; Gilbert, A.; Skurk, C.; Calabria, E.; Picard, A.; Walsh, K.; Schiaffino, S.; Lecker, S.H.; Goldberg, A.L. Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 2004, 117, 399–412.
  27. Furuyama, T.; Kitayama, K.; Yamashita, H.; Mori, N. Forkhead transcription factor FOXO1 (FKHR)-dependent induction of PDK4 gene expression in skeletal muscle during energy deprivation. Biochem. J. 2003, 375 Pt 2, 365–371.
  28. Constantin-Teodosiu, D. Regulation of muscle pyruvate dehydrogenase complex in insulin resistance: Effects of exercise and dichloroacetate. Diabetes. Metab. J. 2013, 37, 301–314.
  29. Investigators, N.-S.S.; Finfer, S.; Liu, B.; Chittock, D.R.; Norton, R.; Myburgh, J.A.; McArthur, C.; Mitchell, I.; Foster, D.; Dhingra, V.; et al. Hypoglycemia and risk of death in critically ill patients. N. Engl. J. Med. 2012, 367, 1108–1118.
  30. Powers, S.K.; Kavazis, A.N.; McClung, J.M. Oxidative stress and disuse muscle atrophy. J. Appl. Physiol. (1985) 2007, 102, 2389–2397.
  31. Powers, S.K.; Smuder, A.J.; Criswell, D.S. Mechanistic links between oxidative stress and disuse muscle atrophy. Antioxid. Redox Signal 2011, 15, 2519–2528.
  32. Abrigo, J.; Elorza, A.A.; Riedel, C.A.; Vilos, C.; Simon, F.; Cabrera, D.; Estrada, L.; Cabello-Verrugio, C. Role of Oxidative Stress as Key Regulator of Muscle Wasting during Cachexia. Oxid. Med. Cell Longev. 2018, 2018, 2063179.
  33. Bedard, K.; Krause, K.H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313.
  34. Forrester, S.J.; Kikuchi, D.S.; Hernandes, M.S.; Xu, Q.; Griendling, K.K. Reactive Oxygen Species in Metabolic and Inflammatory Signaling. Circ. Res. 2018, 122, 877–902.
  35. Scherz-Shouval, R.; Elazar, Z. Regulation of autophagy by ROS: Physiology and pathology. Trends Biochem. Sci. 2011, 36, 30–38.
  36. Brocca, L.; Pellegrino, M.A.; Desaphy, J.F.; Pierno, S.; Camerino, D.C.; Bottinelli, R. Is oxidative stress a cause or consequence of disuse muscle atrophy in mice? A proteomic approach in hindlimb-unloaded mice. Exp. Physiol. 2010, 95, 331–350.
  37. Lang, T.; Streeper, T.; Cawthon, P.; Baldwin, K.; Taaffe, D.R.; Harris, T.B. Sarcopenia: Etiology, clinical consequences, intervention, and assessment. Osteoporos. Int. 2010, 21, 543–559.
  38. Brooks, S.V.; Faulkner, J.A. Skeletal muscle weakness in old age: Underlying mechanisms. Med. Sci. Sports Exerc. 1994, 26, 432–439.
  39. Chen, W.; Datzkiw, D.; Rudnicki, M.A. Satellite cells in ageing: Use it or lose it. Open. Biol. 2020, 10, 200048.
  40. Verdijk, L.B.; Koopman, R.; Schaart, G.; Meijer, K.; Savelberg, H.H.; van Loon, L.J. Satellite cell content is specifically reduced in type II skeletal muscle fibers in the elderly. Am. J. Physiol. Endocrinol. Metab. 2007, 292, E151–E157.
  41. Ryall, J.G.; Schertzer, J.D.; Lynch, G.S. Cellular and molecular mechanisms underlying age-related skeletal muscle wasting and weakness. Biogerontology 2008, 9, 213–228.
  42. Lexell, J.; Taylor, C.C. Variability in muscle fibre areas in whole human quadriceps muscle: Effects of increasing age. J. Anat. 1991, 174, 239–249.
  43. Frontera, W.R.; Suh, D.; Krivickas, L.S.; Hughes, V.A.; Goldstein, R.; Roubenoff, R. Skeletal muscle fiber quality in older men and women. Am. J. Physiol. Cell Physiol. 2000, 279, C611–C618.
  44. Goodpaster, B.H.; Park, S.W.; Harris, T.B.; Kritchevsky, S.B.; Nevitt, M.; Schwartz, A.V.; Simonsick, E.M.; Tylavsky, F.A.; Visser, M.; Newman, A.B. The loss of skeletal muscle strength, mass, and quality in older adults: The health, aging and body composition study. J. Gerontol. A Biol. Sci. Med. Sci. 2006, 61, 1059–1064.
  45. Mawson, A.R.; Croft, A.M. Gulf War Illness: Unifying Hypothesis for a Continuing Health Problem. Int. J. Environ. Res. Public Health 2019, 16, 111.
  46. Ramirez-Sanchez, I.; Navarrete-Yanez, V.; Garate-Carrillo, A.; Loredo, M.; Lira-Romero, E.; Estrada-Mena, J.; Campeau, A.; Gonzalez, D.; Carrillo-Terrazas, M.; Moreno-Ulloa, A.; et al. Development of muscle atrophy and loss of function in a Gulf-War illness model: Underlying mechanisms. Sci. Rep. 2020, 10, 14526.
  47. Thomson, R.M.; Parry, G.J. Neuropathies associated with excessive exposure to lead. Muscle Nerve 2006, 33, 732–741.
  48. Nyirenda, M.H.; Morandi, E.; Vinkemeier, U.; Constantin-Teodosiu, D.; Drinkwater, S.; Mee, M.; King, L.; Podda, G.; Zhang, G.X.; Ghaemmaghami, A.; et al. TLR2 stimulation regulates the balance between regulatory T cell and Th17 function: A novel mechanism of reduced regulatory T cell function in multiple sclerosis. J. Immunol. 2015, 194, 5761–5774.
  49. Mendelson, M.; Nel, J.; Blumberg, L.; Madhi, S.A.; Dryden, M.; Stevens, W.; Venter, F.W.D. Long-COVID: An evolving problem with an extensive impact. S. Afr. Med. J. 2020, 111, 10–12.
  50. Ma, K.; Mallidis, C.; Bhasin, S.; Mahabadi, V.; Artaza, J.; Gonzalez-Cadavid, N.; Arias, J.; Salehian, B. Glucocorticoid-induced skeletal muscle atrophy is associated with upregulation of myostatin gene expression. Am. J. Physiol. Endocrinol. Metab. 2003, 285, E363–E371.
More
This entry is offline, you can click here to edit this entry!