Journal List > Ann Pediatr Endocrinol Metab > v.30(2) > 1516090877

Ha, Sung, and Kim: Myokines and interorgan crosstalk: bridging exercise to health promotion and disease prevention

Abstract

Exercise is known to promote physical health and reduce the risk of various diseases. During exercise, skeletal muscle actively contracts to perform movements and secretes hormone-like molecules termed myokines. The beneficial effects of exercise have been assessed with respect to myokine production, and those of irisin on bone, adipose tissue, and the brain have been well documented. Irisin, through its interactions with the integrin αV family, plays a crucial role in bone maintenance, metabolic regulation, and cognitive function. Building on the established understanding of irisin, this discussion will examine the functions and effects of other myokines as key secretory factors in exercise, emphasizing their broader roles in health promotion and the potential for new therapeutic strategies in disease prevention and treatment.

Highlights

· Skeletal muscle and bone act as a secretory organ to interact with other organs.
· Irisin mediates exercise effects on fat, bone and brain via integrin αV family.
· Various myokines have been identified upon different types of exercise.

Introduction

1. Types of exercise

Exercise has positive effects on various organs in the human body. According to the National Institutes of Health, there are four broad categories of exercise: endurance, resistance, balance, and flexibility. Endurance exercise is known to enhance cardiovascular and circulatory system functions; resistance exercise helps to increase muscle size and strength; balance exercise improves the ability to maintain proper posture; and flexibility exercise increases the range of motion in joints, reducing the risk of injury and improving athletic performance [1]. Among these four types of exercise, endurance and resistance exercise have been extensively studied in animal models and in numerous clinical trials. These studies have focused on the cellular effects of exercise in specific organs.

2. Systemic effects of exercise

1) Skeletal muscle

Endurance and resistance exercise induce distinct changes in skeletal muscle, including significantly increasing the ratio of type I to type II muscle fibers [2]. Type I muscle fibers, also known as slow-twitch muscle fibers, have a lower contraction force than type IIB and type IIX fibers but are resistant to fatigue. They primarily utilize lipids as an energy source and produce Adenosine triphosphate (ATP) through oxidative mitochondrial metabolism. In humans, mitochondria in slow-twitch muscle fibers occupy about 6% of the muscle volume and are characterized by a high-density cristae structure. Additionally, the enzymes involved in the tricarboxylic acid (TCA) cycle in type I slow-twitch fibers are twice as active as fast-twitch fibers, as is the coenzyme oxidation capacity of electron transport [3]. These characteristics of slow-twitch fibers enable the skeletal muscle to produce energy for prolonged periods, supporting endurance exercise [2,3].
Conversely, resistance exercise leads to muscle hypertrophy and an increase in type IIB and type IIX muscle fibers [3]. These fibers, also known as fast-twitch muscle fibers, can generate strong contractions but fatigue quickly. Fast-twitch fibers utilize glucose as their primary energy source and rapidly produce ATP through glycolysis, providing energy for high-intensity, short-duration activities. The activity of phosphofructokinase, the rate-limiting enzyme in glycolysis, is higher in fast-twitch fibers than in slow-twitch fibers. Glycogen, the stored form of glucose, is more abundant in fast-twitch fibers, and the rate of glycogen breakdown is faster in these fibers when skeletal muscle is electrically stimulated to induce contraction. These features of fast-twitch fibers facilitate the performance of resistance exercises [3,4].

2) Bone

Bone is the organ that responds to both mechanical stress and hormonal changes during exercise, and numerous studies have been conducted to assess variations in bone upon exercise. For instance, increased bone density in the femur has been demonstrated after chronic regular resistance training in adults, with several biochemical mechanisms elucidated [5-9]. However, the effects of endurance exercise on bone present a more complex picture: some studies have reported positive effects of endurance exercise on bone density [10-13], while others have found no significant changes or even reduced bone density, with an increase in sclerostin, a protein that inhibits bone formation, detected in the plasma following endurance exercise [14]. Therefore, it is crucial to elucidate the effects of different types of exercise on bones, particularly by understanding the biochemical processes involved.
Research on bone cells has primarily focused on osteoblasts and osteoclasts. Osteoblasts are responsible for bone formation, while osteoclasts are involved in bone resorption. When subjected to external physical stimuli, osteoblasts are activated, leading to bone formation. Conversely, during lactation or in cases of calcium deficiency, osteoclasts resorb bone to maintain the body's calcium levels. Bone formation and resorption occur cyclically rather than unidirectionally. In healthy individuals, this cyclical process maintains bone homeostasis by ensuring that old minerals and micro-damaged areas are removed, allowing continuous renewal of bone. However, in conditions where bone resorption exceeds bone formation, as occurs in certain diseases, aging, or menopause, conditions like osteoporosis can develop [15].
The differentiation and activity of osteoblasts and osteoclasts are regulated by external stimuli and internal physiological changes, coordinated by osteocytes. Osteocytes, making up about 90% of bone cells, sense physical stimuli and hormonal changes during exercise to maintain bone homeostasis [15]. Osteocytes are derived from osteoblasts and located in bone canaliculi, where they regulate osteoblasts and osteoclasts to oversee bone remodeling [15,16]. Osteocytes secrete Wingless-related integration site and osteoprotegerin to promote bone formation by activating osteoblasts and inhibiting osteoclasts. Conversely, they secrete sclerostin and receptor activator of nuclear factor κB ligand to promote bone resorption by inhibiting osteoblasts and activating osteoclasts [15]. While the role of osteocytes in maintaining bone homeostasis through specific secretions is well known, research is needed to understand their responses and secretory functions during different types of exercise.

3) Other organs

The positive effects of exercise on health extend beyond the muscles and bones directly involved in physical activity. Exercise can reduce adipose tissue volume and inflammation, mitigate metabolic diseases such as nonalcoholic fatty liver disease, and enhance brain functions like memory. These findings demonstrate that the benefits of exercise are not confined to the muscles and bones but have comprehensive impacts across various body systems. Moreover, the effects on other organs are considered to be the result of complex mechanisms beyond the simple increase in energy expenditure [17].
However, the molecular-level understanding of exercise effects on different organs remains insufficient and requires further research. In particular, to leverage the positive effects of exercise for the development of therapeutic drugs, it is essential to accurately understand the roles and mechanisms of substances secreted during exercise. Therefore, it is essential to conduct biochemical studies on the effects of exercise.

3. Crosstalk between skeletal muscle and other organs upon exercise

As the primary organ recruited during exercise, skeletal muscle not only directly provides the contractile force for exercise, but also mediates the beneficial effects to other organs. For instance, studies have shown that muscle-specific knockout of peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), a coactivator protein known to increase mitochon-drial biogenesis and oxidative metabolism in skeletal muscle [18-24], leads to disturbance of glucose homeostasis due to hypoinsulinemia and hyperglycemia. The activity of PGC-1α is thought to be limited to skeletal muscle during the transformation to slow-twitch muscle fiber and to promotion of mitogenesis and vascularization. A study by Lin et al. [25] showed that skeletal muscle-specific PGC-1α overexpression leads to the formation of more 'reddish' muscle than the wild-type, in which slow-twitch muscle fibers predominate. An additional role of skeletal muscle as a mediator of interorgan crosstalk via secretory factors was suggested by Handschin et al. [26] after investigating the reduction of insulin secretion by pancreatic β cells in skeletal-muscle-specific PGC-1α-knockout mice. The effects of muscle-specific PGC-1α knockout on systemic variations were investigated, and the morphology of pancreatic β cells, especially the size, was significantly reduced in the transgenic mice [19-21]. It was subsequently reported that skeletal-muscle-specific PGC-1α overexpression produced effects in various organs, including increasing thermogenic capacity in subcutaneous fat [27], preventing bone loss, and enhancing cognitive function [28] (Fig. 1). Together, these findings suggest that muscle-specific changes are not confined to the muscle itself but have effects on other organs such as the pancreas. These findings have evoked interest in the interactions between muscle and other organs, particularly focusing on the role of myokines—proteins secreted by skeletal muscle [26]. The crosstalk between skeletal muscle and other organs is thought to be mediated by various molecules, including circulating lipids and peptides and even central nervous system signals. In addition, there is strong evidence that cytokines, including interleukin-6 (IL-6), can act as myokines [29].

4. Secretory factors released during endurance exercise

In response to endurance exercise, muscle tissues release a range of secretory factors known as myokines that mediate diverse physiological effects such as metabolism, inflammation, and tissue repair across the body. Table 1 provides a summary of key myokines induced by endurance exercise, outlining their primary functions, target tissues, and contributions to exercise-induced metabolic and homeostatic adaptations. This compilation emphasizes the critical role of myokines in systemic health and metabolic resilience as part of the body's response to sustained physical activity.

1) Interleukin-6

Both the gene expression and plasma concentration of IL-6, one of the first myokines to be studied extensively, have been shown to increase as a result of endurance exercise. Clinical studies have shown that plasma IL-6 concentrations and skeletal muscle biopsy IL-6 gene expression were elevated after a marathon compared to pre-race measurements [30]. Increased IL-6 expression is considered a mediator of the effects of endurance exercise. Administration of recombinant human IL-6 can elevate plasma IL-6 to concentrations similar to those measured postexercise and induces an anti-inflammatory response by inhibiting tumor necrosis factor production. Additionally, IL-6 has been confirmed to contribute to increased endogenous glucose production, demonstrating its role in interactions between skeletal muscle and the liver [31-33].

2) FNDC5/Irisin

After researchers first shed light on possible crosstalk between skeletal muscle and other organs, the downstream signaling factor PGC-1α was investigated as a putative mediator (Fig. 1). In response to endurance exercise, the mRNA expression of PGC-1α and of the downstream Fibronectin type III domain containing 5 (FNDC5) increases significantly [27,34-36]. The FNDC5 type I membrane protein is cleaved to form irisin, (named after Iris, the Greek messenger goddess), which is secreted into the extracellular milieu and circulation to be delivered to other organs such as subcutaneous fat, bone, and brain [27,34].

(1) Fat

In subcutaneous adipose tissue, the exercise-induced hormone irisin promotes the expression of thermogenic genes, particularly uncoupling protein 1 (UCP1), a central mediator of mitochondrial heat production. This upregulation of UCP1, along with other thermogenic factors, contributes to an increase in energy expenditure, not only through direct energy consumption during physical activity, but also by enhancing heat generation [27]. Thus, irisin facilitates a key metabolic benefit of exercise through molecular modulation of thermogenesis.
Furthermore, irisin enhances thermogenic capacity in subcutaneous adipose tissue by expanding a specialized subset of adipocyte progenitor cells (APCs) known as beige APCs [37]. These beige APCs, identified by the cell surface marker CD81, play a crucial role in adaptive thermogenesis in response to cold and physical exercise [37]. Notably, CD81 forms a complex with integrins αV/β1 and αV/β5, mediating activation of integrinfocal adhesion kinase (FAK) signaling upon irisin stimulation to drive the proliferation of these progenitor cells [37].
Collectively, irisin enhances basal thermogenesis in subcutaneous adipose tissue by upregulating thermogenic gene expression and promoting the proliferation of thermogenic beige APCs in response to endurance exercise. This process is a critical pathway through which exercise exerts beneficial effects on energy balance and metabolic health as a result of cellular adaptations within adipose tissue.

(2) Bone

The effects of endurance exercise on bone health remain controversial, and the impacts of exercise on bone formation and resorption have not been elucidated fully [5,7-10,12-14]. To determine whether irisin influences bone formation and/or resorption, the impact of FNDC5 knockout in bone was assessed in the lumbar vertebrae of ovariectomized mice, a model that simulates menopause in humans [38,39]. Surprisingly, FNDC5-knockout mice showed resistance to ovariectomy-induced bone loss [38], suggesting that irisin may contribute to bone resorption in response to endurance exercise.
Interestingly, irisin appears to play distinct roles depending on sex and physiological context [40]. Irisin is essential in the development of the male skeleton, protecting it from calcium deficiency. However, this protective effect does not extend to females, in whom irisin may instead serve a unique role by targeting osteocytes to mobilize calcium stores during lactation, ensuring sufficient calcium availability for offspring.
Irisin's role in bone resorption suggest it as a promising therapeutic target for menopause-related osteopenia and osteoporosis. Plasma irisin concentrations were greater in ovariectomized mice than in a control group [38], and FNDC5 overexpression exacerbated bone loss in ovariectomized mice, particularly in calcium-deficient or hormonally altered states such as menopause [41].

(3) Brain

Irisin is also implicated in cognitive enhancement, as demonstrated in studies using FNDC5-knockout mice and Alzheimer disease (AD) models [42-44]. These studies indicate that irisin decreases amyloid β42 and prevents AD-induced neuronal cell death, contributing to the cognitive benefits of exercise [42-44]. Moreover, irisin has been shown to reduce the accumulation of α-synuclein fibrils in models of Parkinson disease [45], suggesting its potential as a treatment for neurodegenerative diseases including AD and Parkinson disease.

(4) Mechanism of action for irisin

The molecular mechanism by which irisin binds to target receptors and transduces signals in target organs is not known. Members of the integrin αV family were identified as irisin receptors in a study using the murine osteocyte like cell line osteocyte cell line after applying quantitative proteomics utilizing mass spectrometry [38]. Osteocytes act as command-control cells to maintain bone homeostasis [15]. To determine the specificity of these receptors for irisin, the irisin-integrin binding assay was performed and confirmed that the integrin αV family directly interacts with irisin. Additionally, irisin-integrin binding motifs were identified through hydrogen-/deuterium-exchange mass spectrometry and mapped at an atomic level using cryogenic electron microscopy (cryo-EM) [46]. Furthermore, the molecular mechanism of irisin-mediated effects of endurance exercise in fat, bone, and brain has been studied through signaling response and function tests upon integrin αV family-irisin interactions [47]. Previous studies of integrin signaling have established phosphorylation of FAK integrin as the major signal transduction event of integrin activation [48-50], and various irisin functional assays have confirmed this activity. The gene expression of sclerostin, which is essential for bone resorption and known to be induced by irisin, has been investigated in irisin-treated osteocytes coincubated with inhibitors of integrin-ligand binding, such as RGDS peptide [51,52] and echistatin [53], and integrin αV-ligand binding inhibitors such as cyclo-RGDyK peptide [54,55]. The results indicated the activity of integrin αV family members as irisin receptors to mediate the effects of endurance exercise through FAK signaling pathways. Furthermore, cell and in vivo studies have shown that CD81 is essential for irisin-integrin αV signaling and its role in beige fat [37].

3) Neurturin

Since irisin was identified, people have tried to identify other exercise-induced hormones [56]. One such compound is neurturin, a myokine secreted by skeletal muscle in response to endurance exercise and has been reported to impact motor neurons [57]. In 1996, a novel neurotrophic factor that contributes to cell survival was discovered during the culture of sympathetic neurons, and this factor was named neurturin [58,59]. In the skeletal muscle of genetically modified mice with muscle-specific overexpression of the PGC-1α gene, which mimics endurance-trained mice, neurturin is secreted [60]. Similarly, the gene expression of neurturin was upregulated in skeletal muscle from mice that underwent chronic endurance exercise training [57]. Increased secretion of muscle-derived neurturin contributes to the crosstalk between skeletal muscle and motor neurons [57]. Neurturin promotes motor neuron remodeling to induce a slow motor neuron phenotype [57], which leads to the transformation of muscle fiber types toward slow-twitch fibers by improving mitochondrial function, promoting angiogenesis, and reprogramming glucose and lipid metabolism for muscle cells to perform fatty acid oxidation [57]. Therefore, the induction of skeletal-muscle-derived neurturin during endurance exercise improves motor coordination and exercise performance.

4) Fibroblast growth factor 21

Fibroblast growth factor 21 (FGF21) is primarily recognized as a hepatokine, originating from the liver, but its expression and function are also well-documented in adipose tissue [61] and the pancreas [61,62]. Overexpression of FGF21 in the liver or pharmacological administration of recombinant FGF21 protein has produced various effects, including increased insulin sensitivity and reduced blood glucose and plasma triglyceride concentrations in mice fed high-fat diets. In addition, FGF21 receptors are the conjugated form between FGF receptor 1c and an essential cofactor Klotho including α-Klotho and β-Klotho [63]. Physiologically, plasma FGF21 concentration increases in response to stressors such as exercise or cold. In skeletal muscle, FGF21 is expressed as a myokine, upregulated in plasma by endurance exercise [35,64,65], and has been shown to influence energy metabolism [66]. Muscle-derived FGF21 acts on white adipose tissue to increase fatty acid oxidation and respiratory capacity [61].

5) β-Aminoisobutyric acid

Intermediate metabolites of signaling proteins can also serve as mediators of exercise effects by being secreted from muscle. The metabolite β-aminoisobutyric acid (BAIBA) is secreted in mice that overexpress the muscle-specific PGC-1α gene as a model of endurance training [67]. The BAIBA metabolite increases the expression of thermogenic genes in subcutaneous white adipose tissue [68]. In addition to its effects on white adipose tissue, BAIBA contributes to glucose homeostasis by inducing β-oxidation in the liver [69,70]. Moreover, BAIBA secreted from skeletal muscle during exercise acts on osteocyte mitochondria, preventing cell death caused by reactive oxygen species and contributing to bone homeostasis [71]. The BABA metabolite may be secreted and act as a hormone to impart the effects of exercise on other organs. Further studies of the roles of other exercise-induced metabolite hormones, such as N-lactoyl-phenylalanine [72] and succinate [73], offer a deeper understanding of exercise.

5. Secretory factors induced by resistance exercise

In addition to endurance exercise, resistance exercise also prompts the secretion of distinct myokines from muscle tissue. 1 presents an overview of key myokines stimulated by resistance exercise, describing their primary roles and target tissues to underscore their impact on the body's adaptive response to strength training.

1) Myostatin

Myostatin, identified in the early 2000s as a myokine directly involved in the regulation of muscle size by direct interaction with activin type II receptors [74-79], has been a primary target for research regarding muscle hypertrophy induced by resistance exercise [80] and therapeutic agents for muscle-wasting diseases [81-83]. Research has highlighted that the expression and secretion of myostatin decrease during resistance exercise, leading to increased muscle size. However, a proportional increase in muscle strength has not been reported, and the strength per unit muscle area was found to be less than in wild-type mouse models [81-84]. Various interpretations have been proposed for this phenotype. From an energy metabolism perspective, it is thought that the reduction in mitochondrial respiration in muscle lowers the beta-oxidation of fatty acids, which is a major energy supply factor [85]. The role of myostatin as a myokine has also been studied with respect to its interactions with adipose tissue and the liver. Myostatin inhibits beta-oxidation of fatty acids and brown adipose tissue formation [86,87]. In the liver, myostatin is associated with reduced glucose uptake and insulin sensitivity, potentially increasing insulin resistance [88].

2) Meteorin-like

Research on the PGC-1α4 isoform led to the discovery of a distinct myokine known as meteorin-like (Metrnl) [89]. Muscle-cell-specific overexpression of PGC-1α4 in mice resulted in increased skeletal muscle mass and strength, as well as elevated expression of thermogenic genes, including UCP1, in both subcutaneous and visceral adipose tissues. Similarly, gene microarray and mass spectrometry analyses identified Metrnl in primary myotubes overexpressing PGC-1α4. It was also confirmed that Metrnl secretion increases in the skeletal muscle of exercise-trained mice [90]. Secreted Metrnl promotes activation of adipose tissue macrophages through IL-4 and IL-13 secreted from eosinophils [90]. The IL-4/IL-13 signaling pathway induces the expression of thermogenic genes in subcutaneous and visceral white adipose tissue, inducing browning and contributing to enhanced energy expenditure

Conclusion

Skeletal muscle, as the organ most directly impacted by physical exercise, undergoes specific structural and biochemical changes depending on the type and intensity of activity. Different exercise modalities influence muscle fiber composition distinctly, creating a mosaic of possible adaptations that extend to various body systems through the secretion of myokines and metabolic hormones [2-4]. These muscle-derived factors such as protein hormones like irisin [27] and neurturin [57] and metabolic compounds including BAIBA [67], N-lactoyl-phenylalanine [72], and succinate [73] are mediators that extend the effects of exercise beyond muscle tissue itself, promoting metabolic, musculoskeletal, and neurological health benefits (Fig. 2). However, while the biochemical understanding of exercise-induced benefits is evolving, many molecular mechanisms remain unclear, and targeted therapeutic strategies based on obtained insights are under development.
The potential of exercise as a therapeutic intervention has been demonstrated across various health conditions, with strong evidence supporting its role in preventing metabolic disorders (e.g., obesity, diabetes, nonalcoholic fatty liver disease), musculoskeletal diseases (e.g., sarcopenia, muscular dystrophy, osteoporosis), and neurodegenerative conditions (e.g., AD, Parkinson disease). Additionally, exercise has shown promise in reducing the risk of certain cancers, such as colorectal and breast cancer, and may even delay progression in some cases. To harness these benefits fully, there is an urgent need to understand the molecular processes involved in exercise tolerance and plasticity, particularly through myokine research. Notably, further studies into myokine expression and secretion across muscle fiber types could reveal crucial biochemical pathways driving exercise adaptations. For instance, examining the roles of FNDC5 and irisin secretion in specific muscle fiber types may provide valuable insights into the effects of distinct muscle characteristics on whole-body health outcomes.
Biochemical studies of exercise should include a comprehensive range of analyses—from genetic to protein-level changes and their post-translational modifications—considering the unique responses of each fiber type to physical stimuli. This knowledge will be instrumental in advancing therapeutic strategies that replicate the effects of exercise, particularly for populations where direct exercise interventions are challenging due to age or physical limitations. With an aging society facing an increasing incidence of exercise-modifiable diseases, the development of therapeutic agents that can mimic the effects of physical exercise is critical. Such agents could not only facilitate effective management of age-associated metabolic, musculoskeletal, and neurological disorders, but also contribute to cancer prevention strategies. Ultimately, research that deepens our understanding of muscle fiber-specific myokine expression and secretion will pave the way for next-generation therapeutics that target the unique biochemical effects of each muscle fiber type, providing tailored interventions that mimic the multifaceted benefits of exercise.

Notes

Conflicts of interest

Hyeonwoo Kim holds a patent related to irisin (WO2019157495A3). Except for that, no potential conflict of interest relevant to this article was reported.

Funding

This study was supported by a grant from the National Research Foundation of Korea (NRF; RS-2023-00213512).

Author contribution

Conceptualization: HK; Data curation: HK, JH, SS; Funding acquisition: HK; Project administration: HK; Visualization: HK, JH, SS; Writing - original draft: HK, JH, SS; Writing - review & editing: HK, JH, SS

References

1. Four types of exercise can improve your health and physical ability [Internet]. Bethesda (MD): National Institue on Aging (NIA); c2021 [2024 Aug 30]. Available from: https://www.nia.nih.gov/health/exercise-and-physical-activity/four-types-exercise-can-improve-your-health-andphysical#endurance.
2. Howald H, Hoppeler H, Claassen H, Mathieu O, Straub R. Influences of endurance training on the ultrastructural composition of the different muscle fiber types in humans. Pflugers Arch. 1985; 403:369–76.
crossref
3. Ortenblad N, Nielsen J, Boushel R, Soderlund K, Saltin B, Holmberg HC. The muscle fiber profiles, mitochondrial content, and enzyme activities of the exceptionally well-trained arm and leg muscles of elite cross-country skiers. Front Physiol. 2018; 9:1031.
4. Wilson JM, Loenneke JP, Jo E, Wilson GJ, Zourdos MC, Kim JS. The effects of endurance, strength, and power training on muscle fiber type shifting. J Strength Cond Res. 2012; 26:1724–9.
crossref
5. Hong AR, Kim SW. Effects of resistance exercise on bone health. Endocrinol Metab (Seoul). 2018; 33:435–44.
crossref
6. Nomura S, Takano-Yamamoto T. Molecular events caused by mechanical stress in bone. Matrix Biol. 2000; 19:91–6.
crossref
7. Eatemadololama A, Karimi MT, Rahnama N, Rasolzadegan MH. Resistance exercise training restores bone mineral density in renal transplant recipients. Clin Cases Miner Bone Metab. 2017; 14:157–60.
crossref
8. Spindler A, Paz S, Berman A, Lucero E, Contino N, Penalba A, et al. Muscular strength and bone mineral density in haemodialysis patients. Nephrol Dial Transplant. 1997; 12:128–32.
crossref
9. Vincent KR, Braith RW. Resistance exercise and bone turnover in elderly men and women. Med Sci Sports Exerc. 2002; 34:17–23.
crossref
10. Snow-Harter C, Bouxsein ML, Lewis BT, Carter DR, Marcus R. Effects of resistance and endurance exercise on bone mineral status of young women: a randomized exercise intervention trial. J Bone Miner Res. 1992; 7:761–9.
crossref
11. Gargouri N, Walke H, Belbeisi A, Hadadin A, Salah S, Ellis A, et al. Estimated burden of human Salmonella, Shigella, and Brucella infections in Jordan, 2003-2004. Foodborne Pathog Dis. 2009; 6:481–6.
12. Duckham RL, Peirce N, Bailey CA, Summers G, Cameron N, Brooke-Wavell K. Bone geometry according to menstrual function in female endurance athletes. Calcif Tissue Int. 2013; 92:444–50.
crossref
13. Scofield KL, Hecht S. Bone health in endurance athletes: runners, cyclists, and swimmers. Curr Sports Med Rep. 2012; 11:328–34.
14. Pickering ME, Simon M, Sornay-Rendu E, Chikh K, Carlier MC, Raby AL, et al. Serum sclerostin increases after acute physical activity. Calcif Tissue Int. 2017; 101:170–3.
crossref
15. Bonewald LF. The amazing osteocyte. J Bone Miner Res. 2011; 26:229–38.
crossref
16. van Bezooijen RL, Roelen BA, Visser A, van der Wee-Pals L, de Wilt E, Karperien M, et al. Sclerostin is an osteocyte-expressed negative regulator of bone formation, but not a classical BMP antagonist. J Exp Med. 2004; 199:805–14.
crossref
17. Kirwan JP, Sacks J, Nieuwoudt S. The essential role of exercise in the management of type 2 diabetes. Cleve Clin J Med. 2017; 84(7 Suppl 1):S15–21.
crossref
18. Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Mootha V, et al. Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1. Cell. 1999; 98:115–24.
crossref
19. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 2003; 34:267–73.
crossref
20. Patti ME, Butte AJ, Crunkhorn S, Cusi K, Berria R, Kashyap S, et al. Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: potential role of PGC1 and NRF1. Proc Natl Acad Sci U S A. 2003; 100:8466–71.
21. Mootha VK, Handschin C, Arlow D, Xie X, St Pierre J, Sihag S, et al. Erralpha and Gabpa/b specify PGC-1alpha-dependent oxidative phosphorylation gene expression that is altered in diabetic muscle. Proc Natl Acad Sci U S A. 2004; 101:6570–5.
22. Schreiber SN, Emter R, Hock MB, Knutti D, Cardenas J, Podvinec M, et al. The estrogen-related receptor alpha (ERRalpha) functions in PPARgamma coactivator 1alpha (PGC-1alpha)-induced mitochondrial biogenesis. Proc Natl Acad Sci U S A. 2004; 101:6472–7.
23. Handschin C, Rhee J, Lin J, Tarr PT, Spiegelman BM. An autoregulatory loop controls peroxisome proliferator-activated receptor gamma coactivator 1alpha expression in muscle. Proc Natl Acad Sci U S A. 2003; 100:7111–6.
24. Czubryt MP, McAnally J, Fishman GI, Olson EN. Regulation of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1 alpha ) and mitochondrial function by MEF2 and HDAC5. Proc Natl Acad Sci U S A. 2003; 100:1711–6.
25. Lin J, Wu H, Tarr PT, Zhang CY, Wu Z, Boss O, et al. Transcriptional co-activator PGC-1 alpha drives the formation of slow-twitch muscle fibres. Nature. 2002; 418:797–801.
crossref
26. Handschin C, Choi CS, Chin S, Kim S, Kawamori D, Kurpad AJ, et al. Abnormal glucose homeostasis in skeletal muscle-specific PGC-1alpha knockout mice reveals skeletal muscle-pancreatic beta cell crosstalk. J Clin Invest. 2007; 117:3463–74.
27. Bostrom P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, et al. A PGC1-alpha-dependent myokine that drives brownfat-like development of white fat and thermogenesis. Nature. 2012; 481:463–8.
28. Yang S, Loro E, Wada S, Kim B, Tseng WJ, Li K, et al. Functional effects of muscle PGC-1alpha in aged animals. Skelet Muscle. 2020; 10:14.
crossref
29. Pedersen BK, Steensberg A, Fischer C, Keller C, Keller P, Plomgaard P, et al. Searching for the exercise factor: is IL-6 a candidate? J Muscle Res Cell Motil. 2003; 24:113–9.
30. Ostrowski K, Rohde T, Zacho M, Asp S, Pedersen BK. Evidence that interleukin-6 is produced in human skeletal muscle during prolonged running. J Physiol. 1998; 508(Pt 3):949–53.
crossref
31. Starkie R, Ostrowski SR, Jauffred S, Febbraio M, Pedersen BK. Exercise and IL-6 infusion inhibit endotoxin-induced TNF-alpha production in humans. FASEB J. 2003; 17:884–6.
32. Kistner TM, Pedersen BK, Lieberman DE. Interleukin 6 as an energy allocator in muscle tissue. Nat Metab. 2022; 4:170–9.
crossref
33. Keller C, Steensberg A, Pilegaard H, Osada T, Saltin B, Pedersen BK, et al. Transcriptional activation of the IL-6 gene in human contracting skeletal muscle: influence of muscle glycogen content. FASEB J. 2001; 15:2748–50.
crossref
34. Jedrychowski MP, Wrann CD, Paulo JA, Gerber KK, Szpyt J, Robinson MM, et al. Detection and Quantitation of Circulating Human Irisin by Tandem Mass Spectrometry. Cell Metab. 2015; 22:734–40.
crossref
35. Lee P, Linderman JD, Smith S, Brychta RJ, Wang J, Idelson C, et al. Irisin and FGF21 are cold-induced endocrine activators of brown fat function in humans. Cell Metab. 2014; 19:302–9.
crossref
36. Pekkala S, Wiklund PK, Hulmi JJ, Ahtiainen JP, Horttanainen M, Pollanen E, et al. Are skeletal muscle FNDC5 gene expression and irisin release regulated by exercise and related to health? J Physiol. 2013; 591:5393–400.
37. Oguri Y, Shinoda K, Kim H, Alba DL, Bolus WR, Wang Q, et al. CD81 controls beige fat progenitor cell growth and energy balance via FAK signaling. Cell. 2020; 182:563–77.e20.
crossref
38. Kim H, Wrann CD, Jedrychowski M, Vidoni S, Kitase Y, Nagano K, et al. Irisin Mediates Effects on Bone and Fat via alphaV Integrin Receptors. Cell. 2018; 175:1756–68.e17.
crossref
39. Almeida M, Laurent MR, Dubois V, Claessens F, O'Brien CA, Bouillon R, et al. Estrogens and androgens in skeletal physiology and pathophysiology. Physiol Rev. 2017; 97:135–87.
crossref
40. Shimonty A, Pin F, Prideaux M, Peng G, Huot J, Kim H, et al. Deletion of FNDC5/irisin modifies murine osteocyte function in a sex-specific manner. Elife. 2024; 12:RP92263.
crossref
41. Estell EG, Le PT, Vegting Y, Kim H, Wrann C, Bouxsein ML, et al. Irisin directly stimulates osteoclastogenesis and bone resorption in vitro and in vivo. Elife. 2020; 9:e58172.
crossref
42. Lourenco MV, Frozza RL, de Freitas GB, Zhang H, Kincheski GC, Ribeiro FC, et al. Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer's models. Nat Med. 2019; 25:165–75.
crossref
43. Islam MR, Valaris S, Young MF, Haley EB, Luo R, Bond SF, et al. Exercise hormone irisin is a critical regulator of cognitive function. Nat Metab. 2021; 3:1058–70.
crossref
44. Kim E, Kim H, Jedrychowski MP, Bakiasi G, Park J, Kruskop J, et al. Irisin reduces amyloid-beta by inducing the release of neprilysin from astrocytes following downregulation of ERK-STAT3 signaling. Neuron. 2023; 111:3619–33.e8.
crossref
45. Kam TI, Park H, Chou SC, Van Vranken JG, Mittenbuhler MJ, Kim H, et al. Amelioration of pathologic alpha-synuclein-induced Parkinson's disease by irisin. Proc Natl Acad Sci U S A. 2022; 119:e2204835119.
crossref
46. A M, Wales TE, Zhou H, Draga-Coletă SV, Gorgulla C, Blackmore KA, et al. Irisin acts through its integrin receptor in a two-step process involving extracellular Hsp90α. Mol Cell. 2023; 83:1903–20.e12.
crossref
47. Farmer SR. Boning up on irisin. N Engl J Med. 2019; 380:1480–2.
crossref
48. D'Amico M, Hulit J, Amanatullah DF, Zafonte BT, Albanese C, Bouzahzah B, et al. The integrin-linked kinase regulates the cyclin D1 gene through glycogen synthase kinase 3beta and cAMP-responsive element-binding protein-dependent pathways. J Biol Chem. 2000; 275:32649–57.
49. Giancotti FG, Ruoslahti E. Integrin signaling. Science. 1999; 285:1028–32.
crossref
50. Schaller MD, Hildebrand JD, Shannon JD, Fox JW, Vines RR, Parsons JT. Autophosphorylation of the focal adhesion kinase, pp125FAK, directs SH2-dependent binding of pp60src. Mol Cell Biol. 1994; 14:1680–8.
crossref
51. Plow EF, Pierschbacher MD, Ruoslahti E, Marguerie G, Ginsberg MH. Arginyl-glycyl-aspartic acid sequences and fibrinogen binding to platelets. Blood. 1987; 70:110–5.
crossref
52. Plow EF, Haas TA, Zhang L, Loftus J, Smith JW. Ligand binding to integrins. J Biol Chem. 2000; 275:21785–8.
crossref
53. Kumar CC, Nie H, Rogers CP, Malkowski M, Maxwell E, Catino JJ, et al. Biochemical characterization of the binding of echistatin to integrin alphavbeta3 receptor. J Pharmacol Exp Ther. 1997; 283:843–53.
54. Chen X, Tohme M, Park R, Hou Y, Bading JR, Conti PS. Micro-PET imaging of alphavbeta3-integrin expression with 18F-labeled dimeric RGD peptide. Mol Imaging. 2004; 3:96–104.
crossref
55. Dechantsreiter MA, Planker E, Matha B, Lohof E, Holzemann G, Jonczyk A, et al. N-Methylated cyclic RGD peptides as highly active and selective alpha(V)beta(3) integrin antagonists. J Med Chem. 1999; 42:3033–40.
56. Leal LG, Lopes MA, Batista ML Jr. Physical exercise-induced myokines and muscle-adipose tissue crosstalk: a review of current knowledge and the implications for health and metabolic diseases. Front Physiol. 2018; 9:1307.
crossref
57. Correia JC, Kelahmetoglu Y, Jannig PR, Schweingruber C, Shvaikovskaya D, Zhengye L, et al. Muscle-secreted neurturin couples myofiber oxidative metabolism and slow motor neuron identity. Cell Metab. 2021; 33:2215–30.e8.
crossref
58. Kotzbauer PT, Lampe PA, Heuckeroth RO, Golden JP, Creedon DJ, Johnson EM Jr, et al. Neurturin, a relative of glial-cell-line-derived neurotrophic factor. Nature. 1996; 384:467–70.
crossref
59. Baudet C, Pozas E, Adameyko I, Andersson E, Ericson J, Ernfors P. Retrograde signaling onto Ret during motor nerve terminal maturation. J Neurosci. 2008; 28:963–75.
crossref
60. Mills R, Taylor-Weiner H, Correia JC, Agudelo LZ, Allodi I, Kolonelou C, et al. Neurturin is a PGC-1alpha1-controlled myokine that promotes motor neuron recruitment and neuromuscular junction formation. Mol Metab. 2018; 7:12–22.
crossref
61. Kharitonenkov A, Shiyanova TL, Koester A, Ford AM, Micanovic R, Galbreath EJ, et al. FGF-21 as a novel metabolic regulator. J Clin Invest. 2005; 115:1627–35.
crossref
62. Moyers JS, Shiyanova TL, Mehrbod F, Dunbar JD, Noblitt TW, Otto KA, et al. Molecular determinants of FGF-21 activity-synergy and cross-talk with PPARgamma signaling. J Cell Physiol. 2007; 210:1–6.
crossref
63. Goetz R, Beenken A, Ibrahimi OA, Kalinina J, Olsen SK, Eliseenkova AV, et al. Molecular insights into the klotho-dependent, endocrine mode of action of fibroblast growth factor 19 subfamily members. Mol Cell Biol. 2007; 27:3417–28.
crossref
64. Cuevas-Ramos D, Almeda-Valdes P, Meza-Arana CE, Brito-Cordova G, Gomez-Perez FJ, Mehta R, et al. Exercise increases serum fibroblast growth factor 21 (FGF21) levels. PLoS One. 2012; 7:e38022.
crossref
65. Kim KH, Kim SH, Min YK, Yang HM, Lee JB, Lee MS. Acute exercise induces FGF21 expression in mice and in healthy humans. PLoS One. 2013; 8:e63517.
crossref
66. Hojman P, Pedersen M, Nielsen AR, Krogh-Madsen R, Yfanti C, Akerstrom T, et al. Fibroblast growth factor-21 is induced in human skeletal muscles by hyperinsulinemia. Diabetes. 2009; 58:2797–801.
crossref
67. Roberts LD, Bostrom P, O'Sullivan JF, Schinzel RT, Lewis GD, Dejam A, et al. beta-Aminoisobutyric acid induces browning of white fat and hepatic beta-oxidation and is inversely correlated with cardiometabolic risk factors. Cell Metab. 2014; 19:96–108.
crossref
68. Jeremic N, Chaturvedi P, Tyagi SC. Browning of white fat: novel insight into factors, mechanisms, and therapeutics. J Cell Physiol. 2017; 232:61–8.
crossref
69. Ginter E, Simko V. Recent data on obesity research: beta-aminoisobutyric acid. Bratisl Lek Listy. 2014; 115:492–3.
70. Jung TW, Park HS, Choi GH, Kim D, Lee T. beta-aminoisobutyric acid attenuates LPS-induced inflammation and insulin resistance in adipocytes through AMPK-mediated pathway. J Biomed Sci. 2018; 25:27.
71. Kitase Y, Vallejo JA, Gutheil W, Vemula H, Jähn K, Yi J, et al. β-aminoisobutyric acid, l-BAIBA, is a muscle-derived osteocyte survival factor. Cell Rep. 2018; 22:1531–44.
crossref
72. Li VL, He Y, Contrepois K, Liu H, Kim JT, Wiggenhorn AL, et al. An exercise-inducible metabolite that suppresses feeding and obesity. Nature. 2022; 606:785–90.
crossref
73. Reddy A, Bozi LH, Yaghi OK, Mills EL, Xiao H, Nicholson HE, et al. pH-Gated succinate secretion regulates muscle remodeling in response to exercise. Cell. 2020; 183:62–75.e17.
crossref
74. Lee SJ, McPherron AC. Regulation of myostatin activity and muscle growth. Proc Natl Acad Sci U S A. 2001; 98:9306–11.
crossref
75. McPherron AC, Lee SJ. Double muscling in cattle due to mutations in the myostatin gene. Proc Natl Acad Sci U S A. 1997; 94:12457–61.
76. Hamrick MW, McPherron AC, Lovejoy CO, Hudson J. Femoral morphology and cross-sectional geometry of adult myostatin-deficient mice. Bone. 2000; 27:343–9.
crossref
77. Grobet L, Martin LJ, Poncelet D, Pirottin D, Brouwers B, Riquet J, et al. A deletion in the bovine myostatin gene causes the double-muscled phenotype in cattle. Nat Genet. 1997; 17:71–4.
crossref
78. Grobet L, Poncelet D, Royo LJ, Brouwers B, Pirottin D, Michaux C, et al. Molecular definition of an allelic series of mutations disrupting the myostatin function and causing double-muscling in cattle. Mamm Genome. 1998; 9:210–3.
crossref
79. Kambadur R, Sharma M, Smith TP, Bass JJ. Mutations in myostatin (GDF8) in double-muscled Belgian Blue and Piedmontese cattle. Genome Res. 1997; 7:910–6.
80. Willoughby DS. Effects of heavy resistance training on myostatin mRNA and protein expression. Med Sci Sports Exerc. 2004; 36:574–82.
crossref
81. Latres E, Pangilinan J, Miloscio L, Bauerlein R, Na E, Potocky TB, et al. Myostatin blockade with a fully human monoclonal antibody induces muscle hypertrophy and reverses muscle atrophy in young and aged mice. Skelet Muscle. 2015; 5:34.
crossref
82. St Andre M, Johnson M, Bansal PN, Wellen J, Robertson A, Opsahl A, et al. A mouse anti-myostatin antibody increases muscle mass and improves muscle strength and contractility in the mdx mouse model of Duchenne muscular dystrophy and its humanized equivalent, domagrozumab (PF-06252616), increases muscle volume in cynomolgus monkeys. Skelet Muscle. 2017; 7:25.
83. Latres E, Mastaitis J, Fury W, Miloscio L, Trejos J, Pangilinan J, et al. Activin A more prominently regulates muscle mass in primates than does GDF8. Nat Commun. 2017; 8:15153.
crossref
84. Jang J, Park S, Kim Y, Jung J, Lee J, Chang Y, et al. Myostatin inhibition-induced increase in muscle mass and strength was amplified by resistance exercise training, and dietary essential amino acids improved muscle quality in mice. Nutrients. 2021; 13:1508.
crossref
85. Mouisel E, Relizani K, Mille-Hamard L, Denis R, Hourdé C, Agbulut O, et al. Myostatin is a key mediator between energy metabolism and endurance capacity of skeletal muscle. Am J Physiol Regul Integr Comp Physiol. 2014; 307:R444–54.
crossref
86. Hamrick MW, Pennington C, Webb CN, Isales CM. Resistance to body fat gain in 'double-muscled' mice fed a high-fat diet. Int J Obes (Lond). 2006; 30:868–70.
crossref
87. Guo T, Jou W, Chanturiya T, Portas J, Gavrilova O, McPherron AC. Myostatin inhibition in muscle, but not adipose tissue, decreases fat mass and improves insulin sensitivity. PLoS One. 2009; 4:e4937.
crossref
88. Watts R, Ghozlan M, Hughey CC, Johnsen VL, Shearer J, Hittel DS. Myostatin inhibits proliferation and insulin-stimulated glucose uptake in mouse liver cells. Biochem Cell Biol. 2014; 92:226–34.
crossref
89. Ruas JL, White JP, Rao RR, Kleiner S, Brannan KT, Harrison BC, et al. A PGC-1α isoform induced by resistance training regulates skeletal muscle hypertrophy. Cell. 2012; 151:1319–31.
crossref
90. Rao RR, Long JZ, White JP, Svensson KJ, Lou J, Lokurkar I, et al. Meteorin-like is a hormone that regulates immune-adipose interactions to increase beige fat thermogenesis. Cell. 2014; 157:1279–91.
crossref
91. Scheller J, Chalaris A, Schmidt-Arras D, Rose-John S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta. 2011; 1813:878–88.
crossref
92. Tu C, Chen YB, Lai SQ, Yu YP, Huang ZW, Li HZ, et al. Accumulation of beta-aminoisobutyric acid mediates hyperalgesia in ovariectomized mice through Mas-related G protein-coupled receptor D signaling. Biochim Biophys Acta Mol Basis Dis. 2024; 1870:167269.
93. Aoi W, Hirano N, Lassiter DG, Bjornholm M, Chibalin AV, Sakuma K, et al. Secreted protein acidic and rich in cysteine (SPARC) improves glucose tolerance via AMP-activated protein kinase activation. FASEB J. 2019; 33:10551–62.
94. Aoi W, Naito Y, Takagi T, Tanimura Y, Takanami Y, Kawai Y, et al. A novel myokine, secreted protein acidic and rich in cysteine (SPARC), suppresses colon tumorigenesis via regular exercise. Gut. 2013; 62:882–9.
crossref
95. Watanabe-Takano H, Ochi H, Chiba A, Matsuo A, Kanai Y, Fukuhara S, et al. Mechanical load regulates bone growth via periosteal Osteocrin. Cell Rep. 2021; 36:109380.
crossref
96. Subbotina E, Sierra A, Zhu Z, Gao Z, Koganti SR, Reyes S, et al. Musclin is an activity-stimulated myokine that enhances physical endurance. Proc Natl Acad Sci U S A. 2015; 112:16042–7.
crossref

Fig. 1.
Scheme of PGC-1α-mediated interorgan crosstalk between skeletal muscle and other organs during exercise. The activity of PGC-1α is not limited to skeletal muscle, but is also evident in other organs including the pancreas and fat. PGC-1α, peroxisome proliferatoractivated receptor γ coactivator-1α; TFAM, mitochondrial transcription factor A; ERRs, estrogen-related receptors; FNDC5, fibronectin type III domain containing 5.
apem-2448218-109f1.tif
Fig. 2.
Secretion of skeletal-muscle-derived protein and metabolite hormones during different types of exercise induces systemic effects on many organs. IL-6, interleukin-6; FGF21, Fibroblast growth factor 21; BAIBA, β-aminoisobutyric acid; Metrnl, meteorin-like.
apem-2448218-109f2.tif
Table 1.
Factors secreted during endurance exercise and resistance exercise and their functions and known receptors
Exercise type Secretory factors Functions Receptor/signaling
Endurance exercise Interleukin-6 Induces anti-inflammatory response by inhibiting tumor necrosis factor production [30,32] IL-6R, gp130 [32]/PI3K, AMPK, JAK-STAT3 pathway [91]
Contributes to increased endogenous glucose production [31]
Fibronectin type III domain containing 5 (Shed form: Irisin) Upregulates thermogenesis and proliferation of thermogenic fat cells [28] Integrin αV
Contributes to bone resorption [37] Family [38]/Integrin-FAK-CREB pathway [37,38]
Reduced amyloid-β24 in an Alzheimer disease model and reduced accumulation of α-synuclein fibrils in a Parkinson disease model [40,41]
Neurin Promotes transformation of muscle fibers toward slow-twitch fiber phenotype, enhances mitochondrial function, and promotes angiogenesis [55] RET, GFRα2/RET pathway [57]
Fibroblast growth factor Increases fatty acid oxidation and respiratory capacity in mitochondria [34,62] FGFR, Klotho/ APK pathway [63]
β-aminoisobutyric acid Upregulates thermogenic gene expression [66] MrgD/MrgD pathway [92]
Contributes to glucose homeostasis by inducing β-oxidation [67,68]
Contributes to bone homeostasis by preventing ROS-induced cell death [69]
Secreted protein acidic and rich in cysteine Participates in AMPK-mediated glucose regulation and improves glucose tolerance [93,94] AMPK pathway [91]
Musclin, Osteocrin Stimulate osteogenic differentiation and bone formation [95] NPR/CNP pathway [92]
Promote mitochondrial biogenesis [96]
Resistance exercise Myostatin Inhibits fatty acid oxidation and respiratory capacity in mitochondria ActRIIA, ActRIIB/mTORC1 pathway [84]
Inhibits fatty acid oxidation and brown adipocyte formation [84,85]
Reduces glucose uptake and insulin sensitivity [86]
Meteorin-like Upregulates thermogenic gene expression [88] IL-4/IL-13 pathway [90]

IL-6R, Interleukin 6 receptor; PI3K, Phosphatidylinositol 3-kinase; AMPK, AMP-activated protein kinase; JAK-STAT3, Janus kinase-signal transducer and activator of transcription 3; Integrin-FAK-CREB, Integrin-focal adhesion kinase-cAMP responsive element binding protein; RET, Ret proto-oncogene; GFRα2/RET, GDNF family receptor alpha 2/Ret proto-oncogene; MrgD/MrgD, Mas-related G protein coupled receptor; NPR/CNP, Natriuretic peptide receptor/C-type natriuretic peptide.

TOOLS
Similar articles