Myokines are cytokines and peptides produced, expressed, and released by skeletal muscle cells (myocytes) in response to muscular contractions or exercise, functioning as signaling molecules with autocrine, paracrine, and endocrine effects that mediate communication between skeletal muscle and other organs.[1][2]The term "myokine" was first coined in 2003 by exercise physiologist Bengt Saltin to describe these muscle-derived factors, building on the 1997 discovery of myostatin as the inaugural myokine by Se-Jin Lee and colleagues, which inhibits muscle growth.[1] Over 600 myokines have since been identified through secretome analyses of human myocyte cultures, highlighting skeletal muscle's role as an endocrine organ.[1] These molecules are synthesized via transcriptional and translational processes triggered by mechanical stress from contractions, enabling rapid release into circulation to influence distant tissues.[1][2]Key functions of myokines include regulating skeletal muscle mass and function by promoting hypertrophy, proliferation, and differentiation while counteracting atrophy; they also modulate metabolism, inflammation, and tissue regeneration across the body.[1] Through inter-organ crosstalk, myokines affect adipose tissue (e.g., inducing lipolysis and fat browning), the liver (e.g., enhancing glucose uptake), bone (e.g., stimulating formation), the brain (e.g., supporting neurogenesis), and the vascular system (e.g., improving endothelial function).[2] Notable examples include interleukin-6 (IL-6), which boosts fat oxidation and insulin sensitivity while exerting anti-inflammatory effects; irisin, which converts white adipose tissue to energy-expending brown fat; brain-derived neurotrophic factor (BDNF), which enhances cognitive function and neuroplasticity; and myostatin, which limits muscle growth to prevent excessive hypertrophy.[1][2] Dysregulation of myokine signaling is implicated in conditions like sarcopenia, obesity, and metabolic disorders, underscoring their therapeutic potential in exercise-based interventions.[1][2]
Discovery and History
Initial Identification of Muscle-Derived Factors
Myokines are defined as cytokines or peptide hormones that are produced and released by skeletal muscle cells, known as myocytes, primarily in response to muscle contraction.[3] This secretion enables skeletal muscle to act as an endocrine organ, influencing systemic physiological processes beyond local muscle function.[4]The discovery of myokines began with the identification of myostatin in 1997 by McPherron et al. and Se-Jin Lee, a member of the transforming growth factor beta (TGF-β) superfamily that acts as a negative regulator of skeletal muscle growth, marking it as the first recognized myokine.[5] During the 1990s and early 2000s, researchers began to recognize skeletal muscle's role as an endocrine organ through observations that muscle-derived factors enhance glucose uptake in peripheral tissues and promote fat metabolism during exercise.[6] These early insights stemmed from studies showing that physical activity improves insulin sensitivity and lipid oxidation, suggesting the involvement of humoral signals from contracting muscle.[7] The foundational shift occurred with the demonstration that skeletal muscle actively secretes signaling molecules akin to those from traditional endocrine glands.[8]A pivotal experiment in 2000 by Pedersen and colleagues directly identified interleukin-6 (IL-6) as the first muscle-derived factor secreted during muscle contraction in humans.[6] In their study, IL-6 mRNA expression and protein release were measured in skeletal muscle biopsies before and after prolonged exercise, revealing a marked increase in circulating IL-6 levels attributable to production within contracting myocytes.[9] This work established IL-6 as the inaugural exercise-induced myokine, with human exercise trials confirming its release specifically from skeletal muscle to support glucose homeostasis.[6] Subsequent analyses indicated that IL-6 is primarily released via exocytosis from myocytes, while other early-recognized factors may involve membrane shedding for proteolytic liberation.[10] These findings laid the groundwork for viewing muscle contraction as a trigger for endocrine-like signaling.[3]
Key Milestones and Research Evolution
The concept of myokines gained significant traction in 2012 with the identification of irisin, a muscle-derived peptide that promotes the browning of white adipose tissue, thereby linking skeletal muscle activity to systemic metabolic regulation.[11] This discovery, reported by Boström et al., marked a pivotal shift toward recognizing myokines as key mediators in inter-tissue communication, building on earlier observations of muscle-secreted factors like IL-6 identified in 2000.[12] By 2014, research had formalized the paradigm of muscle-organ crosstalk, with studies demonstrating how exercise-induced myokines facilitate signaling between skeletal muscle and distant organs such as the liver, adipose tissue, and pancreas to influence metabolic homeostasis.[13]Over the subsequent decade, myokine research evolved from a primary emphasis on exercise-stimulated secretion—initially highlighted in seminal works defining myokines in 2003—to broader investigations into their production under resting conditions and in pathological states.[2] This progression revealed myokines' roles beyond acute physical activity, including chronic dysregulation in metabolic disorders, where altered profiles contribute to disease progression rather than solely beneficial adaptations.[14] Recent bibliometric analyses from 2023 to 2025 underscore emerging trends, with heightened focus on myokines in sarcopenia, where they modulate muscle atrophy pathways; obesity, via anti-inflammatory and insulin-sensitizing effects; and cancer, particularly cachexia models involving tumor-muscle interactions.[15][14][16]In 2024, a systematic review synthesized evidence on myokines' influence on bone metabolism, highlighting their anabolic effects on osteoblasts and potential therapeutic implications for osteoporosis through exercise-mimetic interventions.[17] This built momentum into 2025, when a comprehensive review proposed the "Myokine-mediated Multi-organ Metabolic Network" theory, positing myokines as central hubs in a dynamic web of endocrine signaling that coordinates metabolic flux across organs like the heart, liver, and adipose tissue during health and stress.[18] Addressing prior research gaps, studies from 2023 onward have expanded investigations into how myokines from skeletal muscle influence cardiac protection, such as against ischemia-reperfusion injury via factors like myonectin, and vascular function, including endothelial health and atherosclerosis prevention.[19][20] This interdisciplinary broadening promises to refine therapeutic strategies targeting myokine networks in multifaceted diseases.
Secretion and Regulation
Mechanisms of Myokine Secretion
Myokines are primarily triggered for secretion through repetitive skeletal muscle contractions during physical exercise, which initiate intracellular signaling cascades leading to altered gene expression. These contractions stimulate the activation of transcription factors, notably peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), a master regulator of mitochondrial biogenesis and metabolic adaptation in muscle fibers. PGC-1α binds to promoters of myokine-encoding genes, promoting their transcription without being secreted itself.[21][22]At the molecular level, muscle contractions elevate intracellular calcium levels, which activate signaling pathways including AMP-activated protein kinase (AMPK). This calcium-AMPK axis, often in concert with PGC-1α, upregulates the expression of specific myokine genes, such as fibronectin type III domain-containing protein 5 (FNDC5), the precursor to irisin. For instance, AMPK is essential for maintaining FNDC5 expression in skeletal muscle, ensuring sustained myokine production in response to contractile stress.[23][24]Myokines are released via diverse secretion pathways tailored to their molecular structure. Larger peptide myokines, such as interleukin-6 (IL-6), are typically exported through the classical exocytosis route, involving synthesis in the endoplasmic reticulum, processing in the Golgi apparatus, and vesicular fusion with the plasma membrane. Alternative non-classical pathways include encapsulation within exosomes—small extracellular vesicles (30–150 nm) shed from muscle cells—that transport myokines like irisin and IL-15 to distant tissues, with exosome release amplified by acute exercise bouts. Additionally, certain myokines arise from membrane-bound precursors through proteolytic shedding; for example, irisin is generated by enzymatic cleavage of the transmembrane FNDC5 protein on the muscle cell surface.[25][21]The secretion of myokines exhibits dose-dependency on exercise intensity, where higher workloads elicit proportionally greater release to meet metabolic demands. Even in the absence of exercise, skeletal muscle maintains basal myokine secretion at low levels to support tissue homeostasis, including regulation of local inflammation and energy balance.[26][27]
Factors Influencing Secretion
The secretion of myokines is profoundly influenced by exercise parameters, with distinct profiles emerging from aerobic versus resistance training modalities. Aerobic exercise, such as endurance activities, acutely elevates interleukin-6 (IL-6) levels up to 100-fold post-exercise, serving as an energy sensor to enhance glucose uptake and fat oxidation.[14] In contrast, resistance training promotes the release of immunoregulatory myokines like interleukin-15 (IL-15) and myonectin, supporting muscle hypertrophy and repair.[28] Acute bouts of exercise trigger rapid increases in IL-6 and meteorin-like protein (Metrnl), while chronic training sustains elevated levels of myonectin and brain-derived neurotrophic factor (BDNF), leading to adaptive improvements in metabolic health.[28]Non-exercise factors also modulate myokine production, including age, sex, nutrition, and inflammation. Aging is associated with diminished secretion of beneficial myokines such as IL-15 and irisin, contributing to reduced muscle regenerative capacity in conditions like sarcopenia.[29] Sex differences arise through hormonal influences, with testosterone in males inhibiting myostatin release to favor muscle anabolism.[28] Chronic inflammation shifts myokine profiles toward catabolic states, elevating myostatin while suppressing anti-inflammatory factors.[28]Pathological conditions further alter myokine secretion, often impairing beneficial profiles. In obesity, irisin levels are reduced and proinflammatory myokines are dysregulated, exacerbating insulin resistance and adipose inflammation.[24] Sedentary lifestyles diminish the release of protective myokines like IL-6 and irisin, fostering a pro-inflammatory environment and muscle atrophy.[14] Conversely, training-adapted muscle in active individuals shows enhanced myokine output, mitigating these deficits. Recent evidence from a 2025 randomized clinical trial demonstrates that 36 weeks of personalized resistance training in older women with possible sarcopenic obesity significantly increased circulating insulin-like growth factor-1 (IGF-1) by 7.73 ng/mL and decreased myostatin by 0.49 ng/mL, alongside improvements in muscle strength and function.[30]Hormonal regulators exert direct control over myokine dynamics, integrating endocrine signals with muscle responses. Insulin enhances glucose uptake in muscle while suppressing proinflammatory myokine release, thereby supporting anabolic processes.[28] Elevated cortisol, as in stress states, upregulates myostatin to promote muscle catabolism and atrophy.[28]
Physiological Functions
Metabolic and Endocrine Regulation
Myokines function as endocrine signals secreted by skeletal muscle to regulate systemic metabolism, including the promotion of glucose uptake in skeletal muscle and peripheral tissues. For instance, interleukin-6 (IL-6), a prototypical myokine released during exercise, enhances glucose disposal by stimulating AMP-activated protein kinase (AMPK) pathways, thereby improving insulin-independent glucose uptake in muscle cells.[31] Irisin, another key myokine, similarly boosts glucose uptake while inducing lipolysis in adipose tissue, contributing to better energy homeostasis.[31] These actions help maintain euglycemia during physical activity and in resting states.[3]In addition to glucose regulation, myokines drive lipolysis and the browning of white adipose tissue (WAT), transforming energy-storing white adipocytes into thermogenic beige adipocytes that increase energy expenditure. Irisin, derived from fibronectin type III domain-containing protein 5 (FNDC5), activates uncoupling protein 1 (UCP1) expression in WAT, promoting mitochondrial biogenesis and fat oxidation to counteract obesity.[32] This process exemplifies muscle-fat crosstalk, where myokines like irisin and IL-6 reduce adiposity by inhibiting white fat accumulation and enhancing lipid mobilization.[4] Recent 2024 analyses highlight myokines' anti-diabetic potential in obesity trends, showing that exercise-induced elevations in IL-6 and irisin improve insulin sensitivity and mitigate type 2 diabetes risk through these mechanisms.[31]Myokines also influence insulin sensitivity and mitochondrial biogenesis across organs, fostering adaptive metabolic responses. For example, IL-6 signaling during exercise enhances fat oxidation in muscle and liver by activating AMPK, which suppresses hepatic glycogenolysis and promotes fatty acid utilization, preventing excessive glucose release.[3] Paracrine effects on adjacent adipocytes further amplify this by increasing lipolytic enzyme activity, while endocrine actions extend to distant sites like the liver to regulate energy partitioning.[33] Collectively, these pathways underscore myokines' role in integrating muscle-derived signals for whole-body metabolic balance and endocrine harmony.[34]
Cardiovascular and Musculoskeletal Effects
Myokines play a crucial role in regulating cardiac structure and function by mitigating pathological remodeling processes. Certain myokines exert protective effects against cardiac hypertrophy and fibrosis, key contributors to heart failure progression. For instance, they inhibit the development of hypertrophy in cardiomyocytes and reduce fibrotic deposition in the myocardium, thereby preserving ventricular function during stress conditions such as ischemia.[35] Additionally, myokines promote angiogenesis in cardiac tissue, enhancing vascularization and oxygen supply to support myocardial repair and adaptation to exercise-induced demands.[36]In the context of heart regulation, decorin, a myokine secreted by skeletal muscle in response to exercise, facilitates collagen organization within cardiac extracellular matrix. By modulating collagen fibril assembly and stabilizing the matrix, decorin prevents excessive fibrosis and supports proper tissue remodeling post-injury, such as myocardial infarction.[37] This anti-fibrotic action involves inhibition of transforming growth factor-beta (TGF-β) signaling, which otherwise drives collagen overproduction and scar formation. Autocrine effects of myokines on cardiomyocytes further contribute to these benefits, enabling local metabolic regulation and protection against adverse remodeling directly within heart muscle cells.[38]Shifting to musculoskeletal impacts, myokines help maintain muscle integrity by inhibiting wasting processes, particularly under conditions of disuse or injury. They counteract atrophy through pathways that promote protein synthesis and satellite cell activation, ensuring skeletal muscle mass preservation and functional recovery.[39] In bone tissue, myokines stimulate osteoblast activity, fostering bone formation and enhancing overall density. This anabolic influence counters resorption, supporting structural integrity and reducing fracture risk via increased mineralization and matrix deposition.[40]Recent 2024 research highlights advances in understanding myokine-driven bone metabolism, emphasizing their role in exercise-mediated osteoblast proliferation and bone mass augmentation. Studies demonstrate that myokine secretion during physical activity correlates with 1-2% improvements in bone density after structured training regimens, underscoring their therapeutic potential for maintaining skeletal health. Paracrine signaling from myokines to osteocytes further amplifies these effects, coordinating cellular responses that optimize bone remodeling and adaptation to mechanical loads.[41]
Neurological and Immunomodulatory Roles
Myokines exert significant influence on neurological function through endocrine signaling pathways that enable certain molecules to cross the blood-brain barrier (BBB), thereby promoting neuroplasticity and cognitive processes. For instance, exercise-induced myokines such as irisin and brain-derived neurotrophic factor (BDNF) traverse the BBB to enhance synaptic plasticity in regions like the hippocampus, facilitating neuronal adaptation and connectivity.[42] This crosstalk supports memory consolidation by activating signaling cascades, including the cAMP/PKA/CREB pathway, which strengthens long-term potentiation and information retention in the brain.[43] Additionally, muscle-derived BDNF directly contributes to hippocampal neurogenesis, increasing neuron proliferation in the dentate gyrus and counteracting age-related cognitive decline.[44] These effects extend to mood regulation, where elevated BDNF levels mitigate depressive symptoms by modulating neurotransmitter systems and reducing neuroinflammation.[45]Beyond cognition, myokines participate in sleep regulation via endocrine signaling to the hypothalamus, a key integrator of circadian rhythms and homeostatic balance. Physical activity triggers myokine release that influences hypothalamic nuclei, promoting restorative sleep patterns and alleviating disruptions associated with sedentary lifestyles. This hypothalamic modulation underscores the broader muscle-brain axis, where myokines like IL-6 and BDNF act as intermediaries to synchronize neural activity with peripheral metabolic demands, enhancing overall brain resilience.[4]In the realm of immunomodulation, myokines exhibit anti-inflammatory properties by balancing pro- and anti-inflammatory cytokines, thereby mitigating excessive immune responses. Interleukin-6 (IL-6), a prototypical myokine, demonstrates a dual role: during acute exercise, it promotes an anti-inflammatory milieu by stimulating the release of IL-10 and other suppressors, which dampen systemic inflammation without promoting tissue damage.[46] In contrast, chronic elevation of IL-6, often linked to inactivity, can exacerbate pro-inflammatory states by sustaining cytokine storms.[47] This balance is crucial for maintaining immune homeostasis, as myokines like IL-6 and IL-15 regulate the recruitment and polarization of immune cells.[48]Mechanistically, myokines facilitate paracrine modulation of T-cells and macrophages within the muscle microenvironment, directing immune cell trafficking and function to support resolution of inflammation. For example, exercise-induced myokines attract macrophages to sites of microdamage, shifting them from pro-inflammatory M1 to anti-inflammatory M2 phenotypes, which aids in tissue repair and prevents chronic immune activation.[27] Similarly, they influence T-cell recruitment by altering chemokine gradients, ensuring targeted immune surveillance without overactivation.[49] Through these paracrine interactions, myokines integrate local immune responses with systemic endocrine signals, fostering an adaptive immunomodulatory network.[50]
Emerging Roles in Organ Crosstalk
Myokines play a pivotal role in the interorgan network, particularly through the muscle-liver axis, where they regulate hepatic gluconeogenesis to maintain glucose homeostasis during physical activity.[51] For instance, exercise-induced myokines such as interleukin-6 (IL-6) signal to the liver to enhance glucose output, counterbalancing increased muscular glucose uptake and preventing hypoglycemia.[4] Similarly, in the muscle-kidney axis, myokines like irisin and IL-6 mediate renal protection by suppressing metabolic reprogramming and fibrosis in damaged kidneys, thereby preserving renal function under stress conditions.[52]Recent developments have advanced the understanding of myokine functions, including the 2025 proposal of the "Myokine-mediated Multi-organ Metabolic Network" theory, which posits that myokines orchestrate dynamic inter-organ communication to sustain metabolic balance across tissues.[18] This theory highlights how myokines integrate signals from multiple organs, influencing systemic energy distribution and adaptation to physiological demands. Additionally, myokine exosomes facilitate long-range signaling by encapsulating bioactive molecules for targeted delivery to distant organs, enabling sustained endocrine effects beyond direct secretion.[53]Specific crosstalk mechanisms include myokine interactions with the pancreas, where factors like decorin and irisin protect beta-cell function by mitigating inflammation and enhancing insulin secretion in response to metabolic challenges.[54] In the gut-muscle axis, myokines modulate the microbiome by promoting beneficial microbial shifts through exercise, which in turn influences muscle metabolism via short-chain fatty acid production and reduced inflammation.[55]Bidirectional signaling further underscores myokine involvement in organ crosstalk, as adipokines from adipose tissue can reciprocally regulate myokine release from skeletal muscle, amplifying anti-inflammatory and metabolic effects across the endocrine network.[56] This interplay ensures coordinated responses to maintain homeostasis, with examples like leptin modulating IL-6 production in muscle to fine-tune energy partitioning.[57]
Specific Myokines
Myostatin
Myostatin, also known as growth differentiation factor 8 (GDF8), is a secreted protein belonging to the transforming growth factor-beta (TGF-β) superfamily, primarily produced by skeletal muscle cells.[58] It is synthesized as a precursor protein that undergoes proteolytic processing to form the mature, active dimer, which circulates as a myokine to exert autocrine and paracrine effects.[59] Exercise, particularly aerobic and resistancetraining, suppresses myostatin secretion and reduces its circulating levels, thereby alleviating its inhibitory influence on muscle growth.[60][61]As a potent negative regulator, myostatin primarily functions to limit skeletal musclehypertrophy by inhibiting myoblast proliferation and differentiation through activation of the Smad2/3 signaling pathway.[62] In the cardiac context, myostatin contributes to remodeling processes by modulating cardiomyocyte growth and energy homeostasis, potentially preventing excessive hypertrophy under stress conditions.[63][64]Loss-of-function mutations in the myostatin gene lead to pronounced muscle overgrowth, as exemplified by the double-muscling phenotype in Belgian Blue cattle, where a 11-base pair deletion in the coding region disrupts protein function and results in up to 20% increased muscle mass.[65][66] This discovery has inspired therapeutic strategies targeting myostatin inhibition for muscular dystrophies, such as Duchenne muscular dystrophy, where monoclonal antibodies and gene therapies have shown promise in preclinical models by enhancing muscle mass and strength, though clinical trials have faced challenges in efficacy translation.[67][68]Dysregulation of myostatin, characterized by elevated circulating levels, is implicated in sarcopenia, where it correlates with reduced muscle mass and function in older adults.[69][70] Recent 2025 studies have further positioned myostatin as a potential biomarker for sarcopenia risk, with higher serum levels independently associated with disease prevalence and progression in cohorts including rheumatoid arthritis patients.[71] This elevation may be antagonized by follistatin, highlighting myostatin's role in muscle homeostasis.[72]
Interleukin Family
The interleukins represent a key family of cytokines that function as myokines, primarily secreted by skeletal muscle cells in response to contraction during physical exercise.[73] Among these, interleukin-6 (IL-6) is recognized as the prototypical myokine, with its expression and release markedly upregulated in skeletal muscle during aerobic and resistance activities.[33] This secretion occurs independently of immune cell activation, distinguishing exercise-induced IL-6 from its inflammatory roles in other contexts.[74]IL-6 exerts beneficial metabolic effects by promoting lipolysis in adipose tissue and enhancing glucose uptake in muscle, thereby facilitating energy mobilization during prolonged exercise.[75] It also displays anti-inflammatory properties, counteracting pro-inflammatory cytokines like tumor necrosis factor-alpha (TNF-α) and stimulating the production of anti-inflammatory mediators such as interleukin-10 (IL-10).[76] In parallel, interleukin-15 (IL-15), another prominent member of this family, supports muscle hypertrophy by enhancing protein synthesis and inhibiting proteolysis, contributing to adaptive responses in skeletal muscle following resistance training.[77]A hallmark of IL-6 as a myokine is its rapid and substantial elevation in plasma levels, which can increase up to 100-fold immediately following intense exercise, before returning to baseline within hours.[78] Recent 2025 research has further highlighted IL-6's potential in cancer suppression, demonstrating that exercise-induced elevations in IL-6 levels, as part of broader myokine responses, correlate with reduced tumor cell proliferation in vitro, suggesting synergies with therapeutic modalities like radiotherapy.[79]At the molecular level, interleukin myokines such as IL-6 and IL-15 primarily signal through the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway. Upon binding to their respective receptors on target cells, these ligands activate receptor-associated JAK kinases, leading to phosphorylation and nuclear translocation of STAT proteins, which then regulate gene expression involved in metabolic and anabolic processes.[74] This pathway underscores the endocrine-like communication of muscle-derived interleukins to distant tissues.[80]
Irisin
Irisin is a myokine produced through the proteolytic cleavage of fibronectin type III domain-containing protein 5 (FNDC5), a type I transmembrane precursor protein predominantly expressed in skeletal muscle.[81] The mature irisin peptide consists of approximately 112 amino acids, forming a structure with a fibronectin type III domain that enables its secretion into circulation following enzymatic processing by proteases such as furin.[82] Its secretion is regulated by peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) in response to exercise stimuli.[81]Discovered in 2012, irisin was initially identified in mice as an exercise-induced factor derived from FNDC5 that mimics some metabolic benefits of physical activity.[81] Early reports suggested its role in promoting metabolic adaptations, but controversy arose regarding its existence and physiological relevance in humans, primarily due to challenges in detecting low circulating levels and concerns over antibody specificity in immunoassays.[83] This debate was largely resolved by 2019 studies employing mass spectrometry and validated assays, which confirmed detectable irisin in human plasma and cerebrospinal fluid, with levels influenced by age and sex.[84]The primary function of irisin centers on metabolic adaptation, particularly by inducing the "browning" of white adipose tissue, where it drives the differentiation of white adipocytes into beige fat cells capable of thermogenesis.[81] This process enhances energy expenditure and fat oxidation, as irisin directly activates uncoupling protein 1 (UCP1) in adipocytes, uncoupling mitochondrial respiration from ATP production to generate heat.[81] Additionally, irisin improves insulin sensitivity by facilitating glucose uptake in skeletal muscle and adipose tissue, thereby mitigating insulin resistance associated with sedentary lifestyles.[85]Exercise acutely elevates circulating irisin levels, with the rise correlating positively with increased resting energy expenditure and overall metabolic rate in humans.[86] In 2024 research, recombinant irisin administration in high-fat diet-fed mice reversed obesity-related metabolic dysfunction, including reduced body weight and improved glucose homeostasis, through enhanced brown adipose tissue activation.[87] These findings underscore irisin's potential as a mediator of exercise-induced metabolic benefits.
Brain-Derived Neurotrophic Factor (BDNF)
Brain-derived neurotrophic factor (BDNF) serves as a key myokine secreted by skeletal muscle, primarily in its precursor pro-BDNF form, which differs from the mature BDNF isoform predominantly produced in the brain. This muscle-derived pro-BDNF is expressed at high levels in human type I muscle fibers and is significantly upregulated during acute and chronic exercise, with contractions inducing its local release within muscle tissue to support neuromuscular adaptations. Unlike brain-derived mature BDNF, which circulates more readily, pro-BDNF from muscle exhibits limited systemic release but can influence peripheral and central nervous system functions through endocrine signaling.[88]As a myokine, BDNF promotes neuronal survival by enhancing the viability of neurons in response to stress and injury, while also driving synaptic plasticity through mechanisms that strengthen connections between neurons, particularly in regions involved in learning and memory. In the context of mood stabilization, elevated BDNF levels counteract depressive states by modulating neurotransmitter systems and reducing neuroinflammation, thereby fostering resilience against mood disorders. These functions are especially relevant in exercise-induced scenarios, where muscle-derived BDNF contributes to neuroprotection and cognitive benefits.[89]Exercise-mediated increases in BDNF have been linked to a reduced risk of depression, as physical activity elevates circulating and tissue BDNF levels, correlating with improved mood and lower incidence of depressive symptoms in both healthy individuals and those with mood disorders. Recent 2023 studies further highlight BDNF's role in sleep regulation, demonstrating that altered BDNF expression disrupts slow-wave sleep and contributes to insomnia, while exercise-induced BDNF may restore sleep architecture by enhancing hippocampal activity. The primary mechanism underlying these effects involves activation of the TrkB receptor in the hippocampus, where BDNF binding triggers downstream signaling cascades, including phosphorylation of CREB and enhancement of long-term potentiation, essential for neuroplasticity and emotional regulation.[90][91][92]
Decorin
Decorin is a small leucine-rich proteoglycan (SLRP) that functions as an exercise-induced myokine secreted by skeletal muscle cells during contraction.[93] As part of the SLRP family, it consists of a core protein with attached glycosaminoglycan chains, enabling its interactions with extracellular matrix components and signaling molecules.[94]In tissue remodeling, decorin plays a key role in regulating collagen assembly by promoting fibrillogenesis and organizing collagen fibers in the extracellular matrix.[95] This function is mediated through direct binding to collagen type I and sequestration of transforming growth factor-β (TGF-β), which inhibits excessive extracellular matrix production and fibrosis.[96] By modulating TGF-β signaling, decorin helps maintain structured tissue architecture, particularly in response to mechanical stress from muscle activity.[97]Decorin also exhibits anti-cancer properties by inhibiting tumor growth through interference with epidermal growth factor receptor (EGFR) signaling. It binds to EGFR, attenuating downstream pathways such as ERK that promote cell proliferation and survival in tumor cells.[98] This suppression extends to invasion and metastasis, as demonstrated in models of prostate and breast cancers where decorin overexpression reduced tumor progression.[99] A 2025 study on breast cancer survivors found that a single bout of resistance or high-intensity interval training acutely increased circulating decorin levels, which in turn suppressed proliferation of MDA-MB-231 cancer cells in vitro by up to 30%.[79] Additionally, decorin contributes to cardiovascular matrix stability by similar ECM regulatory mechanisms.[96]
SPARC (Osteonectin)
SPARC, also known as osteonectin or secreted protein acidic and rich in cysteine, is a matricellular glycoprotein expressed in various tissues, including skeletal muscle, where it functions as an exercise-inducible myokine.[100] Its secretion from muscle cells increases in response to acute and chronic exercise, contributing to tissue remodeling processes.[101] As a key component of the extracellular matrix (ECM), SPARC plays a central role in bone-muscle crosstalk by influencing matrix assembly and cellular interactions at the osteo-muscular interface.[102]In bone metabolism, SPARC modulates mineralization by regulating the deposition and organization of mineralized ECM, particularly through its high affinity for calcium and hydroxyapatite.[103] It promotes osteoblast differentiation and bone formation while inhibiting excessive matrix calcification, thereby maintaining skeletal integrity.[104] Recent 2024 research using multi-omics approaches has identified SPARC as a conserved osteokine secreted by osteoblasts, highlighting its role in promoting bone formation and noting that its expression declines with aging, potentially exacerbating bone mass loss.[105] Additionally, SPARC supports angiogenesis within bone and muscle tissues by modulating vascular endothelial growth factor (VEGF) signaling, which facilitates nutrient delivery and tissue repair during exercise-induced adaptations.[106]Mechanistically, SPARC interacts directly with collagen, acting as a chaperone to facilitate proper fibril assembly and prevent premature collagen interactions with cell surfaces, which is essential for organized ECM in bone and muscle.[102] It also binds VEGF, thereby fine-tuning angiogenic responses to ensure balanced vascularization without excessive proliferation in mineralizing environments.[107] These interactions underscore SPARC's protective role against pathological ECM remodeling, such as fibrosis in cardiac contexts indirectly linked to musculoskeletal health, by preserving matrix compliance and endothelial integrity.[108]
Follistatin and Other Emerging Myokines
Follistatin, a secreted glycoprotein produced by skeletal muscle cells, acts as a potent antagonist to myostatin, a member of the transforming growth factor-β (TGF-β) superfamily that inhibits muscle growth. By binding directly to myostatin, follistatin neutralizes its inhibitory effects on muscle cellproliferation and differentiation, thereby promoting skeletal musclehypertrophy and increasing muscle mass. This antagonistic interaction has been demonstrated in preclinical models where follistatin overexpression leads to significant muscle enlargement independent of myostatin levels in some contexts. Additionally, follistatin binds with high affinity to activins, other TGF-β family members, inhibiting their interaction with receptors and suppressing downstream signaling pathways that limit muscle development.As of 2025, studies have highlighted follistatin's potential as a biomarker for sarcopenia, with higher serum levels associated with physical functional impairment and disease severity in conditions like rheumatoid arthritis in aging populations.[109] In multiorgan networks, follistatin contributes to endocrine signaling that coordinates muscle-liver and muscle-adipose interactions, influencing systemic metabolism and tissue remodeling.Among emerging myokines identified between 2023 and 2025, fibroblast growth factor 21 (FGF21) stands out for its involvement in the metabolic stress response. Secreted by skeletal muscle under conditions like endoplasmic reticulum stress or psychological strain, FGF21 acts as an endocrine signal to enhance glucose uptake, promote fatty acid oxidation, and protect against insulin resistance. This myokine's production surges in response to exercise or nutritional challenges, facilitating adaptations in distant organs such as the liver and adipose tissue.Meteorin-like protein (Metrnl), another recently characterized myokine, exerts neuroprotective effects and drives adipose tissue browning. Induced in muscle by aerobic exercise, Metrnl circulates to stimulate neuronal survival pathways in the brain, reducing inflammation and supporting cognitive function during aging or neurodegenerative stress. Concurrently, it promotes the conversion of white adipose tissue to beige fat, increasing thermogenesis and energy expenditure via endocrine signaling that activates uncoupling protein 1 in adipocytes. These actions position Metrnl as a key player in muscle-brain and muscle-adipose crosstalk within evolving multiorgan frameworks.
Clinical and Therapeutic Implications
Myokines in Sarcopenia and Aging
Sarcopenia, characterized by progressive loss of skeletal muscle mass and function, is closely linked to alterations in myokine secretion during aging. Declining levels of beneficial myokines such as irisin and follistatin contribute to this pathophysiology by impairing muscle regeneration and promoting atrophy. Irisin, derived from fibronectin type III domain-containing protein 5 (FNDC5), decreases in skeletal muscle mRNA and protein expression with advancing age, exacerbating muscle wasting and metabolic dysfunction in aged models. Similarly, serum follistatin concentrations diminish over time, reducing its antagonistic effect on myostatin and thereby accelerating sarcopenic progression. In contrast, pro-atrophic myokines like myostatin often elevate in sarcopenic individuals, further disrupting muscle homeostasis. Interleukin-15 (IL-15), another key myokine, shows reduced plasma levels in older adults with sarcopenia, correlating with diminished muscle strength and increased risk of frailty.Exercise interventions effectively counteract sarcopenia by stimulating myokine release to preserve muscle mass and function. Physical activity, particularly resistance training, upregulates secretion of anabolic myokines such as irisin, follistatin, and IL-15, which enhance myogenesis and mitigate inflammatory pathways. A 2025 randomized controlled trial demonstrated that 36 weeks of personalized resistance training in older women with possible sarcopenic obesity significantly improved muscle function and elevated circulating myokine levels, including irisin and IL-6, leading to better grip strength and reduced fat mass.[110] These findings underscore exercise's role in restoring myokine balance, with resistance protocols showing dose-dependent benefits on physical performance in sarcopenic populations.Myostatin inhibition emerges as a promising anti-aging therapeutic target for sarcopenia, given its role in limiting muscle growth. Pharmacological blockade of myostatin, such as with anti-myostatin antibodies, has reversed age-related muscle loss in preclinical models by increasing muscle mass and strength without adverse effects on other tissues. Clinical translation of this approach is ongoing, supported by evidence that myostatin levels rise in chronic aging conditions, making it a viable intervention to halt sarcopenic decline.In clinical contexts, myokines serve as biomarkers for sarcopenia detection and monitoring. A 2025 review highlights four key myokines—myostatin, irisin, follistatin, and BDNF—as potential diagnostic tools, with their dysregulated profiles reflecting muscle wasting severity in older adults.[111] Low IL-15 levels, in particular, predict sarcopenia risk in community-dwelling elderly, offering a non-invasive marker for early intervention. These biomarkers enable personalized assessments, guiding therapies like exercise or myostatin inhibitors to improve outcomes in aging populations.
Applications in Metabolic Disorders and Cancer
Myokines have emerged as promising therapeutic targets in metabolic disorders, particularly obesity and type 2 diabetes, due to their roles in interorgan crosstalk that regulates energy homeostasis and insulin sensitivity. In obesity, exercise-induced myokines such as irisin facilitate the browning of white adipose tissue, promoting thermogenesis and fat oxidation, which contributes to weight loss and improved metabolic profiles. Recent bibliometric analyses from 2024 highlight a surge in research on myokines like irisin and interleukin-6 (IL-6), emphasizing their mediation of muscle-adipose interactions to combat obesity-related inflammation and enhance interorgan health, with publication volumes increasing 12.5-fold since the prior decade.[112] For type 2 diabetes, myokine mimetics, including synthetic analogs of irisin and brain-derived neurotrophic factor (BDNF), have shown potential to enhance glucose uptake and mitochondrial function in preclinical models, mimicking exercise benefits to improve insulin resistance.Exercise protocols specifically designed to boost irisin levels offer a non-pharmacological approach to weight management in metabolic disorders. High-intensity interval training (HIIT) has been demonstrated to elevate circulating irisin more effectively than moderate continuous exercise, leading to greater reductions in body fat and improvements in insulin sensitivity in obese individuals. Long-term moderate aerobic exercise also significantly increases irisin in both obese and healthy populations, correlating with sustained weight loss and metabolic improvements over months. These protocols underscore irisin's role as a key myokine linking physical activity to adipose tissue remodeling and glycemic control.In oncology, certain myokines exhibit anti-tumor effects by modulating the tumor microenvironment and immune responses. IL-6, when secreted by exercising muscle, can induce apoptosis and inhibit proliferation in specific breast cancer subtypes, contrasting its pro-inflammatory role in other contexts. Decorin, another muscle-derived myokine, suppresses tumor growth by inhibiting epidermal growth factor receptor (EGFR) signaling, reducing angiogenesis, and preventing epithelial-mesenchymal transition in various cancer models. Emerging 2025 studies on exercise interventions in breast cancer survivors reveal that myokine elevation, particularly through HIIT or resistance training, suppresses tumor cell growth in vitro in breast cancer survivors.[79]Therapeutic strategies targeting myostatin hold promise for managing cancer cachexia, a severe muscle-wasting syndrome affecting up to 80% of advanced cancer patients. Myostatin inhibitors, such as monoclonal antibodies like bimagrumab, preserve muscle mass and improve physical function in preclinical cancer models by blocking the TGF-β pathway that drives atrophy. Clinical trials have explored these agents to counteract cachexia in pancreatic and lung cancers, showing modest gains in lean body mass without exacerbating tumor progression.Despite these advances, translating myokine-based therapies faces significant challenges, including delivery methods and specificity in clinical trials. Protein-based myokines suffer from short half-lives, proteolytic degradation, and off-target effects due to ubiquitous receptors, complicating targeted administration to muscle or adipose tissues. Strategies like PEGylation and nanoparticle encapsulation aim to improve stability and tissue specificity, but trials often encounter immunogenicity and dosing inconsistencies, limiting efficacy in heterogeneous patient populations. Ongoing research emphasizes the need for precise biomarkers to monitor myokine responses and refine trial designs for metabolic and oncologic applications.