Back to Journals » Clinical Interventions in Aging » Volume 20
Exploration of Pathogenesis and Cutting-Edge Treatment Strategies of Sarcopenia: A Narrative Review
Authors Gao Y, Liu D, Xiao Q, Huang S, Li L , Xie B, Zhou L, Qi Y, Liu Y
Received 21 January 2025
Accepted for publication 12 May 2025
Published 23 May 2025 Volume 2025:20 Pages 659—684
DOI https://doi.org/10.2147/CIA.S517833
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 4
Editor who approved publication: Dr Nandu Goswami
Yin Gao,1,2,* Di Liu,1,2,* Qixian Xiao,1,* Shan Huang,1,2 Li Li,2 Baocheng Xie,3 Limin Zhou,3 Yi Qi,2 Yanzhi Liu1
1Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Institute of Clinical Medicine, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China; 2Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China; 3Department of Pharmacy, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People’s Hospital), Dongguan, 52305, People’s Republic of China
*These authors contributed equally to this work
Correspondence: Yanzhi Liu, Email [email protected] Yi Qi, Email [email protected]
Abstract: Sarcopenia a progressive and multifactorial musculoskeletal syndrome characterized by loss of muscle mass and function, poses a significant global health challenge, particularly in aging populations. Epidemiological studies reveal that sarcopenia affects approximately 5– 10% of the general population, with prevalence rates escalating dramatically after age 60 to reach 10– 27% in older adults. This age-associated increase contributes significantly to healthcare burdens by elevating risks of disability, frailty, and mortality. Despite its profound impact, current clinical approaches to sarcopenia remain limited. While resistance exercise and protein supplementation form the cornerstone of management, their efficacy is often constrained by poor long-term adherence and variable individual responses, highlighting the urgent need for more comprehensive and personalized treatment strategies. The pathogenesis of sarcopenia is complex and influenced by various factors, including aging, inflammation, nutritional deficits, physical inactivity, and mitochondrial dysfunction. However, the precise molecular mechanisms underlying this condition are still not fully understood. Recent research has made significant strides in elucidating the intricate mechanisms contributing to sarcopenia, revealing novel insights into its molecular and cellular underpinnings. Notably, emerging evidence points to the pivotal role of mitochondrial dysfunction, altered myokine profiles, and neuromuscular junction degeneration in sarcopenia progression. Additionally, breakthroughs in stem cell therapy, exosome-based treatments, and precision nutrition offer promising avenues for clinical intervention. This review aims to synthesize the latest advancements in sarcopenia research, focusing on the novel contributions to its pathogenesis and treatment strategies. We explore emerging trends such as the role of cellular senescence, epigenetic regulation, and targeted therapeutic interventions that could reshape future approaches to managing sarcopenia. By highlighting recent breakthroughs and cutting-edge research, we hope to advance the understanding of sarcopenia and foster the translation of these findings into effective clinical therapies.
Keywords: sarcopenia, muscle, aging, skeleton
Introduction
Sarcopenia, first described by Rosenberg in 1997, is a clinical syndrome characterized by the progressive decline in muscle mass and function, predominantly associated with aging.1 The European Working Group on Sarcopenia in Older People (EWGSOP) defines sarcopenia as a gradual, generalized loss of skeletal muscle mass and strength that leads to reduced physical capability, diminished quality of life, and increased mortality.2 Similar definitions have been proposed by the International Sarcopenia Working Group, the Asian Working Group for Sarcopenia (AWGS), and the Chinese Society of Osteoporosis and Bone Mineral Research.3–5 The AWGS introduced regional guidelines tailored for Asian populations in 2014, which were updated in 2019.4,6 China currently adheres mainly to the EWGSOP definition.
Contemporary sarcopenia diagnosis relies on a tripartite assessment framework encompassing muscle mass, muscle strength, and biomechanical performance. The current gold standard quantifies muscle mass index (ASMI) via dual-energy X-ray absorptiometry (DXA), with sex-specific thresholds defining pathology (<7.0 kg/m² males; <5.4 kg/m² females). Dynamometric evaluation of maximal grip strength establishes functional reserve boundaries (<26 kg males; <18 kg females), while ambulatory capacity is objectively measured through standardized gait analysis, where velocities below 0.8 m/s signify clinically relevant impairment. Diagnostic confirmation necessitates concurrent abnormalities in ≥2 domains, as per international consensus guidelines.7 As the understanding of sarcopenia continues to evolve, the Global Leadership Initiative in Sarcopenia (GLIS) recently suggested that muscle mass, muscle strength, and muscle-specific strength should be considered the core components of sarcopenia, while impaired physical performance should be viewed as an outcome rather than a component of the syndrome.8 Epidemiological studies reveal that sarcopenia affects approximately 5–10% of the general population9 and with prevalence rates escalating dramatically after age 60 to reach 10–27% in older adults.10 Another report estimated that 10–16% of the global elderly population is affected by sarcopenia.11 A meta-analysis involving 41 studies and 34,955 participants found that the prevalence of sarcopenia among community-dwelling individuals was 11% in men and 9% in women.12 Systematic analysis suggest that the overall prevalence of sarcopenia in the elderly ranges from 5% to 37%.13 Among specific groups, prevalence rates are reported to be 41–59% in nursing home residents and 12.9–40.4% in community-dwelling older adults.14 Community-dwelling adults exhibit prevalence rates of 8–13% in men and 7–11% in women (EWGSOP/AWGS/IWGS criteria).15 Certain populations demonstrate disproportionately elevated sarcopenia prevalence, including 40.7% in liver transplant recipients, 51% in male nursing home residents and 23–24% in hospitalized populations.15 These populations exhibit robust high-risk to sarcopenia, strongly correlating with adverse clinical outcomes such as prolonged hospitalization and mortality. Previous reports reveal significant variations in sarcopenia prevalence among older populations, with estimates ranging from 10% to 27% across studies employing different diagnostic classifications and cut-off points. Notably, geographical disparities emerge, with Oceania demonstrating the highest rates when applying EWGSOP criteria, while European populations show the lowest prevalence under EWGSOP2 guidelines. Studies reveal an 8.3% prevalence in Beijing16 (AWGS-2019 criteria) and a striking age-dependent increase from 1.5% (60–69 years) to 33.1% (≥80 years) in Chinese men.17 Age-stratified data indicate a progressive increase from around 8% in adults under 60 years to 10–27% in those aged 60 and above. Intriguingly, sex-specific patterns differ substantially based on diagnostic frameworks: while EWGSOP2 identifies higher prevalence in males, IWGS criteria conversely report elevated rates among females.2 The observed heterogeneity in prevalence estimates reflects methodological disparities in diagnostic frameworks. Existing consensus guidelines (AWGS, EWGSOP, IWGS, FNIH) demonstrate variable sensitivity profiles, with particularly limited detection capability for incipient sarcopenia. While advanced imaging techniques (eg, DXA/BIA) represent the current diagnostic gold standard, their implementation barriers in resource-constrained environments necessitate the development of validated, point-of-care assessment tools. This critical gap has prompted international calls for unified diagnostic standards to enhance early identification and targeted therapeutic interventions. In this work, we performed a systematic search across three major databases (PubMed, Web of Science, and Cochrane Library) for collecting relevant publications from January 2015 to December 2023. The inclusion criteria were peer-reviewed clinical trials, meta-analyses, and mechanistic studies published in English. The exclusion criteria contained case reports, non-human studies, and editorials. A three-phase screening process was employed, which involved title and abstract review, full-text evaluation, and quality assessment, with dual independent verification at each stage.
The observed heterogeneity in prevalence estimates primarily reflects methodological disparities in diagnostic frameworks. Existing consensus guidelines (AWGS, EWGSOP, IWGS, FNIH) demonstrate variable sensitivity profiles, with particularly limited detection capability for incipient sarcopenia. While advanced imaging techniques (eg, DXA/BIA) represent the current diagnostic gold standard, their implementation barriers in resource-constrained environments necessitate the development of validated, point-of-care assessment tools. This critical gap has prompted international calls for unified diagnostic standards to enhance early identification and targeted therapeutic interventions.
Sarcopenia manifests as a multisystemic risk multiplier, demonstrating significant pathophysiological linkages to cardiovascular compromise, respiratory insufficiency, neurocognitive deterioration, frailty syndrome.18 This musculoskeletal degeneration also substantially elevates clinical risks including injurious falls (particularly fragility fractures), unplanned hospital admissions, and all-cause mortality.19 The cumulative disease burden poses significant risks to patients’ functional independence and psychosocial well-being. Emerging evidence positions the SarQoL® questionnaire as a validated multidimensional tool20 for quantifying sarcopenia-specific health-related quality of life (HRQoL) impairments.21 Comparative analyses reveal pronounced health-related quality of life deficits in sarcopenic populations versus their non-sarcopenic counterparts.22 A significantly lower HRQoL was observed for sarcopenic individuals compared with non-sarcopenic ones.23,24 A meta-analysis demonstrated that sarcopenia is associated with lower health-related quality of life measured with SarQoL. Subgroup analyses revealed substantial impact of regions, clinical settings, and diagnostic criteria on the difference in health-related quality of life between sarcopenic and non-sarcopenic individuals.24 This metric now facilitates therapeutic monitoring in clinical trials evaluating sarcopenia interventions–from pharmacological agents like metformin to the effect of pathophysiological states like leaky gut.25 Despite this, sarcopenia is still underdiagnosed and poorly managed in clinical. Standardizing diagnostic criteria and improving measurement techniques are crucial steps toward advancing sarcopenia management.26
The primary risk factors for sarcopenia include aging, chronic diseases, physical inactivity and malnutrition (Figure 1). However, a recent study suggests that the risk factors for sarcopenia in younger individuals differ somewhat from those in older adults. In younger populations, sarcopenia is more closely associated with metabolic and endocrine disorders, while genetic predisposition, physical inactivity, vitamin D deficiency, and poor nutrition are common risk factors across all age groups.27
![]() |
Figure 1 Major risk factors of sarcopenia. |
This comprehensive review critically examines recent breakthroughs in sarcopenia research, with particular emphasis on mechanistic insights into disease pathogenesis and evolving therapeutic paradigms. By highlighting recent breakthroughs and cutting-edge research, we hope to advance the understanding of sarcopenia and foster the translation of these findings into effective clinical therapies.
Risk Factors
Aging and Sarcopenia
Muscle physical functions decline with aging, accompany with a decrease in anti-inflammatory cytokines and an accumulation of inflammatory factors. The inflammatory microenvironment contributes to muscle loss, atrophy, and reduced functionality. Aging also triggers reductions in autophagy and endogenous hormone secretion, impairing muscle growth and regeneration (Figure 2).
![]() |
Figure 2 Signals and factors involved in sarcopenia development due to aging. |
Aging-Induced Muscle Mass Decline and Fiber Degradation
Previous studies reported muscle mass declines by 1–2% annually after the age of 50,28 with higher rates observed in individuals over 75—ranging from 0.64–0.70% in women and 0.80–0.98% in men.29 Prolonged inactivity accelerates muscle atrophy, increasing the risk of sarcopenia.
Aging also affects muscle fiber composition, particularly the fast-twitch type II fibers, which are essential for power and speed. Type II fibers shrink in both size and number as people aging, and their functionality is compromised. Clinical studies show a 10–40% reduction in type II fibers among older adults compared to younger populations.30–32
Skeletal muscle harbors a specialized population of adult stem cells known as satellite cells, which predominantly exist in a quiescent state under homeostatic conditions. These cells exhibit limited proliferative activity, primarily responding to replace damaged myofibers resulting from routine physiological stress while simultaneously preserving the progenitor pool necessary for long-term muscle maintenance. Recent evidence indeed suggests that satellite cell depletion in knockout models does not significantly accelerate sarcopenia progression, indicating that impaired myoregeneration through type II myonecrosis may not be the primary driver of age-related muscle loss. Instead, accumulating data support that sarcopenia develops through multifactorial processes including neuromuscular junction remodeling with motor unit reorganization33 (shifting from type II to type I fiber predominance), chronic low-grade inflammation,34 and altered muscle protein turnover.35 While satellite cells may not be essential for maintaining muscle mass during aging under basal conditions, their dysfunction likely contributes to sarcopenia through paracrine mechanisms affecting the muscle microenvironment.36 Emerging research demonstrates that skeletal muscle exhibit altered crosstalk with immune cells (particularly macrophages),37 fibroblasts, and other stromal components,38 disrupting the coordinated signaling required for effective muscle maintenance and repair.39,40 Epigenetic alterations in Polycomb/Trithorax group protein activity impair the temporal regulation of myogenic gene expression during regeneration.41 These reports suggest that satellite cell dysfunction in aging primarily manifests as impaired microenvironmental communication rather than an absolute loss of regenerative potential. (Figure 3).
![]() |
Figure 3 Aging-induced loss of muscle mass and degeneration of muscle fibers. |
Previous analysis highlight five novel hallmarks that are particularly consequential for age-related skeletal muscle decline: chronic low-grade inflammation, progressive neural dysfunction, extracellular matrix remodeling, diminished vascular perfusion, and disrupted ionic homeostasis, each representing promising therapeutic targets for sarcopenia intervention.39 Building upon this framework, newly work on meta-hallmarks of aging revealed the meta-hallmarks of aging and their intersection with cancer biology, particularly their conceptual expansion of the original hallmarks of aging to include impaired macroautophagy, sustained inflammaging, and gut microbiome dysbiosis - all highly relevant to sarcopenia pathophysiology - interact to drive muscle deterioration.42 These works collectively provide a comprehensive roadmap for developing targeted interventions that address both local tissue changes and organism-wide aging processes contributing to sarcopenia progression.
In conclusion, the combined effects of reduced muscle fiber number, atrophy, and satellite cell dysfunction are central contributors to sarcopenia.
Aging and Excitation-Contraction Coupling (E-CC)
Aging not only reduces muscle mass but also weakens muscle strength at both the single-fiber and whole-muscle levels.43,44 A key reason for this is the disruption of excitation-contraction coupling (E-CC), the process by which neural signals trigger muscle contractions. Impaired Ca2+ release from the sarcoplasmic reticulum (SR) is a potential mechanism behind this decline.45 Additionally, aging decreases the expression of the voltage-gated Ca2+ channel α1 subunit (Cav1.1), which is essential for E-CC. Research by Zhang et al found that genetic knockout of fast skeletal muscle troponin T3 (TnT3) downregulates Cav1.1. In aged sedentary mice, preventing TnT3 fragmentation led to a 20–30% increase in gastrocnemius muscle mass.46 These findings suggest that targeting E-CC abnormalities could help reverse age-related declines in muscle strength and mass.
Aging and Autophagy Reduction
Aging reduces the number of MuSCs in skeletal muscle and disrupts their regulation. This results in impaired muscle regeneration and increased vulnerability to age-related decline.47 Autophagy plays a key role in muscle regeneration, supporting both stem cells and muscle fibers.48 Aged muscle stem cells often exhibit autophagy deficiencies, leading to elevated reactive oxygen species (ROS) levels, which impair regenerative capacity.49 Additionally, excessive ROS damages muscle tissue integrity, further contributing to muscle decline. A decrease in Myf5-positive cells, can lead to muscle defects. Research by Sakuma et al found an increase in autophagy-related molecules, such as p62/SQSTM1 and Beclin-1, in aged mice. However, there was no significant change in the autophagy protein LC3, suggesting a defect in autophagic capacity in aged mice.50 This deficiency contributes to impaired muscle regeneration, metabolic defects, and the progression of sarcopenia.
Aging and Inflammation
Chronic low-grade inflammation is a major driver of sarcopenia. As the body ages, increased fat tissue leads to the accumulation of pro-inflammatory macrophages and immune cells. This imbalance reduces anti-inflammatory cytokines and elevates pro-inflammatory molecules such as leptin, TNF-α, IL-1, and IL-6.50,51 Markers of inflammation, including C-reactive protein and erythrocyte sedimentation rate, are elevated in sarcopenic patients, particularly those with hip fractures.52 Pro-inflammatory cytokines accelerate muscle protein degradation, leading to a loss of muscle mass, strength, and function.53 The ratio of IL-6 to IL-10 is used as an indicator of inflammation in sarcopenia elderly patients.54 This imbalance promotes muscle degradation through pathways such as the ubiquitin-proteasome system and the lysosomal autophagy system. TNF-α could inhibit the Akt/mTOR pathway, triggering muscle degradation via the NF-κB signaling pathway.55 Therefore, chronic inflammation is a significant factor contributing to sarcopenia in the elderly.
Aging and Hormonal Decline
Hormonal changes play a key role in muscle loss with aging. Declining levels of hormones such as growth hormone, testosterone, thyroid hormone, and insulin-like growth factor reduce muscle regeneration and promote atrophy.56 Muscle loss is further exacerbated by increased catabolic signaling mediated by pro-inflammatory cytokines like TNF-α and IL-6.57 Hormonal deficiencies, including lower levels of insulin, estrogen, and androgens, are major contributors to the development of sarcopenia (Table 1).
![]() |
Table 1 The Relationship Between Hormones and Sarcopenia |
Physical Inactivity and Sarcopenia
Physical inactivity is a key risk factor for sarcopenia.66 Muscle fiber decline typically begins around age 50, and sedentary individuals experience more pronounced reductions in muscle mass and strength than those who are physically active.67 Exercise activates various signaling pathways that promote muscle regeneration, enhancing both strength and mass.68 It has been shown to transiently elevate IL-6, which may stimulate the chronic expression of anti-inflammatory mediators such as IL-1 receptor antagonists and IL-10, while suppressing pro-inflammatory mediators like TNFα and IL-1β.69 In a study by Reid et al70 total daily sedentary time was found to be positively correlated with lower lean body mass percentage (−1.70% [95% CI: −2.30, −1.10]) and higher fat mass (2.92 kg [95% CI: 1.94, 3.30]) among 123 elderly individuals living independently. Frequent interruptions in sitting time were associated with a 45% reduction in the risk of pre-sarcopenia (OR = 0.55, 95% CI: 0.34, 0.91), indicating a strong association between sedentary behavior and greater declines in muscle mass and strength. Another cross-sectional study in a Japanese population used logistic regression analysis to explore the relationship between exercise habits and sarcopenia. The study considered factors such as skeletal muscle mass index (SMI), grip strength, walking speed, chair stand time, and single-leg standing time as dependent variables, while middle-aged exercise habits served as independent variables. The results showed significant positive correlations between middle-aged exercise habits and grip strength, walking speed, and single-leg standing time in older adults.71 This suggests that regular exercise during middle age acts as a protective factor against sarcopenia later in life. In summary, individuals who engage in less physical activity are more likely to experience reductions in muscle mass and strength, significantly increasing the risk of sarcopenia.
Mitochondrial Dysfunction and Oxidative Stress in Sarcopenia
Mitochondrial quantity, function, and morphology are closely linked to muscle mass and performance.72 The accumulation of damaged mitochondria and impaired mitochondrial autophagy are common during aging and strongly associated with sarcopenia development.73
In sarcopenia, mitochondrial function declines, with reduced capacity to eliminate defective mitochondria and decreased adenosine triphosphate (ATP) synthesis. This results in lower ATP production and increased oxidative stress. These physiopathological changes are key contributors to muscle weakness and fatigue.74
As aging progresses, the mitochondrial quality-control system undergoes complex molecular changes, including downregulation of genes related to mitochondrial biogenesis, slowed biogenesis rates, mitochondrial DNA mutations, deletions, or copy number variations, and impaired mitochondrial respiratory chain function, leading to increased ROS production.75 Studies suggest that endurance exercise can improve muscle regeneration by modulating mitochondrial metabolism, which promotes satellite cell self-renewal and activation.76 Maintaining a functional mitochondrial network is essential for skeletal muscle health throughout life. Tamaki et al observed that mitochondrial dysfunction occurs at the onset of muscle mass and function decline, highlighting its central role in sarcopenia.77 Oxidative stress and mitochondrial dysfunction lead to DNA fragmentation,78 accelerate cellular denervation, decrease cellular regeneration and differentiation, and ultimately contribute to muscle loss (Figure 4).
![]() |
Figure 4 Mitochondria dysfunction and aging. |
Growing evidence highlights the critical role of mitochondria in sarcopenia progression. Key features of sarcopenia include reduced muscle fiber diameter, decreased myosin heavy chain expression, increased activation of the ubiquitin-proteasome system (UPS), and elevated ROS levels.79 Mitochondria are the primary source of ROS,80 and oxidative phosphorylation (OXPHOS) during intense skeletal muscle contractions. With aging, mitochondrial function declines, leading to bioenergetic failure, increased fatigue, and excessive ROS production.81 This disrupts redox homeostasis and impairs cellular signaling.82 Mitochondrial quality control can be enhanced through mitophagy, the selective degradation of damaged mitochondria, which can be promoted by bioactive components or lifestyle interventions like exercise.83
Defective mitochondrial autophagy and the accumulation of damaged mitochondria also induce inflammation, leading to persistent energy imbalances and increased muscle protein catabolism, which further contribute to sarcopenia (Figure 5). 84 During aging, the reduced capacity of mitochondria in muscle cells is a major factor in the development of sarcopenia,85 and targeting mitochondrial dysfunction may offer new therapeutic approaches for treating sarcopenia.
![]() |
Figure 5 Mitochondrial dysfunction in aging: ROS accumulation, inflammation, and muscle senescence. |
Malnutrition and Sarcopenia
Adequate protein intake and optimal vitamin D levels are essential for maintaining skeletal muscle. Nutritional deficiencies, particularly low protein intake, are major contributors to muscle loss and sarcopenia.
Insufficient Protein Intake
Research indicates that muscle mass declines at rates of 3–8% per decade after age 30, accelerating to 1.1–1.4% per year after 50, and reaching 2.3% per year after 60. Muscle strength and power decline at a rate of 1.5% per year.86 The key factors behind this loss include reduced energy availability and inadequate protein intake. Protein consumption is crucial for preserving skeletal muscle mass, yet malnutrition is common among the elderly. In a study of 836 elderly individuals over 60, Hai et al87 found a strong association between poor nutritional status and sarcopenia. Aging leads to physiological deterioration, reduced protein intake and synthesis, and the accumulation of non-contractile proteins like lipofuscin and cross-linked proteins in muscle. To maintain muscle health, the elderly is recommended to consume 1.0–1.2 g/kg of high-quality protein daily.88 Insufficient protein intake is a significant risk factor for sarcopenia.
Vitamin D Deficiency
Vitamin D deficiency is associated with reduced muscle strength, especially in the elderly, who often experience decreased vitamin D synthesis due to limited sun exposure and metabolic disorders. Studies have shown that treating muscle cells with 100 nM Vitamin D3 for 1–12 days enhances the expression of myogenic regulatory factors, promoting muscle fiber differentiation.89 Nakamura et al demonstrated that low vitamin D levels lead to muscle fiber reduction and muscle atrophy in mice.90 Vitamin D plays a critical role in muscle tone and contraction.91 Overexpression of the vitamin D receptor (VDR) in rats has been shown to promote muscle hypertrophy, increasing muscle cross-sectional area and enhancing anabolic signaling pathways such as the mTOR complex 1 (mTORC1), which stimulates protein synthesis.92 Prolonged vitamin D deficiency primarily affects type II muscle fibers, leading to muscle atrophy, reduced strength, and an elevated risk of falls in the elderly.93 Harvey et al94 reported that vitamin D supplementation reduces the risk of falls by 8%–22%. Additionally, vitamin D has been shown to increase serum IGF-1, which may mitigate muscle atrophy by inhibiting NF-κB and Smad signaling, thereby enhancing muscle growth.95,96 Extensive research has established a significant association between suboptimal serum 25-hydroxyvitamin D (25(OH)D) levels (<50 nmol/L) and impaired muscle protein synthesis, reduced muscle strength, and accelerated progression of sarcopenia, as documented in multiple clinical studies.97–100
Chronic Diseases and Sarcopenia
Many older adults suffer from chronic diseases such as chronic kidney disease, heart failure, lung dysfunction, cancer, diabetes, and hyperthyroidism. These conditions contribute to muscle deterioration, leading to atrophy and functional decline.101–103
Diabetes Mellitus (DM) and Dyslipidemia in Sarcopenia
High blood glucose levels are a risk factor for age-related muscle loss and dysfunction104 Insulin resistance, elevated glucose levels, and increased glucocorticoids accelerate muscle atrophy.
Chronic Kidney Disease and Sarcopenia
Chronic kidney disease impairs muscle precursor (satellite) cell function, reducing myogenesis and contributing to sarcopenia.105 Other factors, such as prolonged dialysis, comorbid diabetes, elevated phosphate levels, and malnutrition, further increase the prevalence of sarcopenia.106
Cancers and Sarcopenia
In cancer patients, oncogenic drivers (eg, MYC, RAS, HIF-1α) promote tumor progression through metabolic reprogramming and oxidative damage.107 Concurrently, aging-associated epigenetic alterations (aberrant DNA methylation, dysregulated histone modifications) and genetic defects (telomere attrition, mitochondrial DNA mutations) further exacerbate genomic instability.42 These synergistic mechanisms not only drive tumorigenesis but also induce metabolic imbalance, directly impairing muscle protein homeostasis and suppressing muscle regenerative capacity, thereby accelerating the development of sarcopenia. Sarcopenia in this population is driven by cancer-related anorexia, malignancy, surgical stress, and the adverse effects of chemotherapy.108 Inflammatory cytokines triggered by cancer can induce muscle breakdown, exacerbating muscle loss.109 A study using tumor mouse models revealed that activation of the receptor for advanced glycation end-products (RAGE) by S100B induces muscle atrophy, suggesting that chronic RAGE activation in cancer patients is linked to systemic inflammation and muscle degradation. Anticancer treatments also worsen physical frailty, contributing to sarcopenia.110
Heart and Lung Dysfunction and Sarcopenia
In heart failure, continuous metabolic demand depletes skeletal muscle protein, increasing the risk of sarcopenia.111 In postoperative non-small cell lung cancer patients, obstructive ventilatory impairment (FEV1% <70%) is an independent risk factor for skeletal muscle loss.112
Hyperthyroidism and Sarcopenia
Hyperthyroidism is a well-known cause of sarcopenia.113 Animal studies show that hyperthyroid mice experience greater muscle fatigability, and the muscle mass and tone decrease in Slc16a2 knockout mice (Mct8KO), which may due to elevated muscle thyroid hormone levels.114,115 Clinically, hyperthyroid patients often present with exercise intolerance, cramps, muscle pain (myalgia), and muscle weakness, likely due to increased calcium ion circulation.116
Genetic Factors and Sarcopenia
The pathogenesis of sarcopenia is multifactorial, with genetic factors playing a key role alongside environmental influences. Recent studies emphasize the impact of genetic variations in structural and metabolic proteins, growth factors, hormones, and inflammatory cytokines.117 Heritability accounts for up to 64% of muscle strength variability in aging populations.118 Studies have identified the COL15A1 gene as one of the genes with shared genetic co-localization evidence related to sarcopenia, and increased expression of the COL15A1 contributed to sarcopenia development.119 Specific gene polymorphisms, such as in the ACTN3 gene, which influences fast-twitch muscle fibers, are associated with differences in muscle mass and performance in older adults.120 Emerging evidence underscores the critical involvement of epigenetic regulation in the pathogenesis and progression of sarcopenia.121 Age-related hypermethylation of DNA and elevated histone deacetylase (HDAC) activity have been shown to suppress myogenic gene expression and impair protein biosynthesis pathways essential for muscle homeostasis.122 Epigenome-wide analyses have revealed 176 differentially methylated CpG sites and 141 distinct methylomic regions significantly associated with sarcopenic phenotypes, with functional enrichment observed in pathways governing myotube fusion dynamics, mitochondrial oxidative phosphorylation, and voltage-gated calcium channels.123 Simultaneously, epigenetic regulators including DNA methylation and HDACs reciprocally modulate miRNA expression, forming an integrated regulatory network wherein miRNAs cooperate with other epigenetic factors to coordinately govern skeletal muscle development and functional adaptation.124 Myogenic miRNAs (including the pro-myogenic miR-1/133/206 cluster and muscle-specific miR-208b) exert central regulatory functions in skeletal muscle development by targeting and suppressing inhibitory factors such as HDAC4 and Pax7, while establishing positive feedback loops with key myogenic transcription factors MyoD and MEF2.125 These genetically related pathophysiological mechanisms interact to exacerbate muscle atrophy and increase the risk of sarcopenia. Clinically significant methylation alterations have been identified in sarcopenic cohorts, including 17 differentially methylated CpG sites and 7 differentially methylated regions (DMRs) were confirmed between patients with sarcopenia and control individuals; hypomethylation at specific FGF2_30 that demonstrates progressive reduction correlating with disease severity. Such epigenetic signatures hold promise as diagnostic biomarkers for sarcopenia risk stratification and therapeutic monitoring.126 Emerging therapeutic strategies focus on epigenetic reprogramming interventions targeting these dysregulated pathways to restore musculoskeletal integrity, offering novel approaches for sarcopenia management.127
Hydration and Muscle–Water Balance and Sarcopenia
Hydration plays a vital role in muscle function and health. Intracellular water (ICW) and extracellular water (ECW) are crucial to maintaining muscle integrity. ICW supports intracellular homeostasis, protein synthesis, and metabolic function. ECW, distributed in interstitial spaces and blood, facilitates nutrient transport and metabolic waste removal.128 The balance between ICW and ECW is regulated by aquaporins, ion channels, and transporters et.al. Disruption of this balance can lead to muscle atrophy, reduced contractility, and weakness—hallmarks of sarcopenia.129
Neuromuscular Junction Transmission and Sarcopenia
Recent studies highlight changes in the neuromuscular junction (NMJ) as a central factor in sarcopenia pathogenesis.130 The NMJ, comprising presynaptic motor neuron terminals, the synaptic cleft, and postsynaptic muscle membranes, relies on acetylcholine (ACh) release for efficient neurotransmission. ACh binds to nicotinic acetylcholine receptors (nAChRs) on the muscle membrane, initiating muscle contraction.131 The C-terminal Agrin fragment(CAF) plays a crucial role in the function of NMJ. Studies have shown that the CAF has higher concentrations in older adults and patients with muscular dystrophy, it is associated with decreased muscle strength and function.132,133 CAF levels correlate with age-related sarcopenia and pathological conditions (eg, diabetes, chronic inflammation).134 Previous analysis demonstrated CAF has been identified as a marker for early diagnosis and monitoring of sarcopenia.135 The differential impact of CAF is associated with sarcopenia subtypes the primary (age-related) and secondary sarcopenia (eg, disuse atrophy, cachexia) demonstrated different utilization value on CAF’s diagnostic and prognostic utility. Exercise and pharmacological agents that may modulate CAF levels, highlighting its potential as a therapeutic target. As age progresses, the loss of motor neurons leads to muscle fiber denervation and NMJ fragmentation, impairing neurotransmission. Additionally, mitochondrial dysfunction and oxidative stress damage NMJ components.136 At the molecular level, the agrin-muscle-specific kinase (MuSK) pathway is critical for NMJ stability. Aging decreases agrin levels and MuSK activation, disrupting NMJ integrity. Similarly, the neuregulin-1 (NRG1)-ErbB2/4 signaling axis, essential for motor neuron and muscle fiber function, diminishes with age, contributing to NMJ degeneration.137
These findings uncover molecular mechanisms of NMJ degeneration and suggest potential therapeutic targets, such as agrin and MuSK regulation for sarcopenia. However, translating these insights into clinical practice requires further research to evaluate their safety and efficacy in humans.
Gut Microbiota and Sarcopenia
Emerging evidence has elucidated the critical involvement of gut microbiota dysbiosis in sarcopenia pathogenesis via the gut-muscle axis,138 with recent mechanistic investigations revealing novel therapeutic targets. Microbial-derived metabolites, particularly short-chain fatty acids (SCFAs)139 and quorum-sensing peptides (QSPs), have emerged as key mediators influencing skeletal muscle homeostasis. SCFAs, generated through bacterial fermentation of dietary fibers, exhibit dual regulatory functions: they promote muscle protein synthesis via mTOR pathway activation140 while simultaneously enhancing mitochondrial biogenesis and metabolic flexibility through AMPK/PGC-1α signaling.141 Interestingly, pediatric studies suggest gut microbial composition and fecal SCFA levels correlate with musculoskeletal parameters in a body fat-dependent manner.142 Preclinical models reveal butyrate supplementation mitigates cachexia-related muscle wasting through gut barrier stabilization and macrophage polarization modulation.143 Previous study systematic screening of 75 microbial quorum-sensing molecules identified 30 bioactive compounds affecting C2C12 myocyte dynamics, substantiating microbiota-derived QSPs as gut-muscle axis mediators.144 Specific QSPs, like iAM373,145 impair muscle regeneration through concurrent suppression of myogenic differentiation and activation of ubiquitin-proteasome degradation. Notably, multi-omics analyses demonstrate that fecal microbiota transplantation from juvenile rodents preserves gut barrier function, enhances mitochondrial performance, and attenuates age-related sarcopenia in senescent recipients, suggesting microbiota-modulating approaches may offer viable therapeutic strategies for musculoskeletal aging.146 These findings collectively underscore the therapeutic potential of modulating specific microbial metabolites while highlighting the need for further research to establish causality in human populations and develop targeted interventions.
Sarcopenia Therapies
The primary goal in treating sarcopenia is to alleviate or reverse the decline in muscle mass and function. Traditional approaches have focused on nutritional interventions, exercise, and pharmacotherapy. Recently, innovative strategies such as stem cell and exosome therapy have been explored, offering new avenues for sarcopenia treatment. The evolving therapeutic approaches for sarcopenia are summarized as follows (Figure 6 and Table 2).
![]() |
Table 2 Advantages and Disadvantages of Sarcopenia Treatment Methods |
![]() |
Figure 6 Treatments for sarcopenia. |
Exercise Intervention
Among the various sarcopenia interventions, physical activity is a cornerstone in both the prevention and treatment of sarcopenia. Resistance training and aerobic exercise are particularly effective in reducing the risk of chronic conditions and enhancing functional mobility in older adults. Current evidence indicates that individualized exercise prescriptions adhering to the FITT framework (Frequency, Intensity, Time, and Type) demonstrate therapeutic efficacy in ameliorating sarcopenia-related muscle weakness and functional decline in older adults.147 Dual-task, and power training may be particularly effective for reducing the C-terminal Agrin fragment—improving neuromuscular junction integrity, and suppressing sarcopenia progression.133 Studies have shown that aerobic exercise is mostly performed in the form of walking, stepping, and bicycle ergometry. For people with musculoskeletal problems, stretching of the upper, trunk, and lower limbs should be recommended to reduce the chance of injury in older adults.148 The most effective strategies for clinically meaningful increase in muscle mass of sarcopenic individuals involve either progressive resistance training protocols or targeted nutritional interventions combining extra energy with adequate protein intake. Current evidence strongly supports structured resistance exercise programs, particularly when combined with leucine-fortified essential amino acid supplementation or whey protein administration in cases of dietary protein inadequacy, as foundational therapeutic approaches for sarcopenia management.149 Importantly, comprehensive exercise interventions incorporating progressive resistance training as a core component demonstrate consistent efficacy in improving muscle strength and physical function among older adults with both frailty and sarcopenia, with meta-analytic data confirming superior outcomes compared to single-modality approaches.150 These multimodal strategies work synergistically by simultaneously stimulating muscle protein synthesis through mechanical loading while providing the necessary substrate availability for tissue remodeling, thereby addressing both the anabolic resistance and nutritional deficiencies characteristic of age-related muscle loss. Resistance training, a cornerstone in managing muscle-wasting diseases,151 has been shown to improve muscle strength, mass, and function—key factors in mitigating sarcopenia.152 Evidence suggests that a resistance training regimen consisting of upper- and lower-body exercises, performed with moderate to high effort for 1–3 sets of 6–12 repetitions, can be highly effective.153 These exercises may involve resistance machines, free weights, bodyweight movements, or resistance bands.154 Importantly, resistance training can be both enjoyable and tolerable for older adults, further reinforcing its effectiveness in sarcopenia treatment. This is exemplified by the Reablement Strategies targeting Sarcopenia (ReStart-S) program, a intervention specifically developed for long-term care residents. The ReStart-S framework integrates current evidence on sarcopenia pathophysiology with practical considerations for frail populations, offering healthcare providers an evidence-based template for developing tailored rehabilitation programs, have yielded promising results that warrant further investigation through large-scale randomized controlled trials to establish its effectiveness in mitigating age-related muscle decline.155 The epidemiological profile of sarcopenia demonstrates significant variability across clinical and community settings, with the highest prevalence rates consistently observed among institutionalized older adults in long-term care facilities. Current diagnostic criteria established by AWGS provide a standardized framework for classifying disease severity, enabling clinicians to distinguish between preclinical, established, and severe sarcopenia stages. This stratification is particularly crucial for developing targeted exercise interventions, as emerging evidence indicates that prescriptive parameters including intensity, frequency, and modality must be precisely calibrated to an individual’s diagnostic category and functional capacity to achieve optimal therapeutic outcomes. The ReStart-S program’s methodological framework offers particularly valuable insights into implementing targeted exercise interventions tailored to different stages of sarcopenia severity. Exercise interventions significantly improve muscle strength (SMD=0.62), muscle mass (SMD=0.28), and physical performance (gait speed +0.08 m/s, SPPB +1.2 points) in older adults with sarcopenia. High-intensity progressive resistance training (>70% 1-RM for ≥12 weeks) demonstrates optimal efficacy, while combined training yields particularly significant functional benefits, with favorable safety profile (AE rate <5%).156 Importantly, These exercises are both pleasant and tolerable for the elderly, further strengthening their effectiveness in treating sarcopenia.157,158 Additionally, physical stimulation, such as Whole Body Vibration Training (WBVT), has proven beneficial in the early stages of sarcopenia. WBVT stimulates bone progenitor cell activity, enhances osteoblast function, and increases levels of sex hormones and cytokines, ultimately improving bone density and muscle function.159,160 In conclusion, physical activity and physical stimulation therapies, are vital components for the prevention and management of sarcopenia.
Nutritional Intervention: Protein and Vitamin D Supplementation
Protein Intake
Malnutrition, particularly inadequate protein intake, is a key contributor to sarcopenia. Insufficient dietary protein impairs the synthesis of muscle proteins, exacerbating muscle loss. While the current recommended dietary allowance (RDA) for protein in adults is 0.8 g/kg of body weight per day,161 this is often insufficient for the elderly to maintain muscle mass and function. Recent expert guidelines suggest that elders should consume 1.0–1.5 g/kg/day of high-quality protein, especially from animal sources such as eggs and meat, to support muscle hypertrophy when combined with resistance training.162 Higher protein intake has been linked to better physical function, with positive associations between protein consumption and physical activity, as well as negative correlations with sedentary behavior.163 Leucine-rich protein supplements, such as whey protein, can help prevent muscle loss, particularly during periods of bed rest or inactivity.164 Given the common decline in protein intake with age,165 it is recommended that older adults aim for around 1.6 g/kg/day to combat sarcopenia.166 Nutrient intake can also be affected by oral health. Logistic regression analysis indicated that the prevalence of sarcopenia is increased among patients diagnosed with reduced oral function (odds ratio: 1.59, 95% confidence interval: 1.02–2.47).167 A cross-sectional study targeting community-dwelling older adults reveals that the prevalence of reduced oral function is 34.3% (n = 427) in the robust group and 65.2% (n = 178) in the sarcopenic group.168 With advancing age, poor oral health status exerts a significant negative impact on food intake, which in turn leads to muscle loss and a decline in muscle strength.169 It is recommended to engage in daily oral function training and undergo regular oral examinations to ensure adequate protein intake.170 Ensuring attention to oral health status provide adequate protein intake, are essential for maintaining muscle health and mitigating sarcopenia in older adults.
Vitamin D Supplementation
Vitamin D deficiency is a major risk factor for muscle atrophy, falls, and fractures in the elderly. Individuals with insufficient vitamin D levels often experience muscle weakness and reduced mobility. This deficiency is particularly common in older adults with limited sun exposure.171 Vitamin D plays a crucial role in muscle metabolism, facilitating calcium absorption and maintaining serum calcium and phosphate levels, which are essential for muscle function.172 Studies suggest that vitamin D supplementation, particularly in combination with leucine, may enhance muscle function and physical performance by promoting protein anabolism. Maintaining optimal serum 25-hydroxyvitamin D levels (50–75 nmol/L) is crucial for preserving muscle strength and supporting muscle protein synthesis. It is recommended that older adults receive 800–1000 international units of vitamin D per day to support musculoskeletal health and reduce the risk of disease.173 Intervention studies indicate that daily supplementation with 1000 IU vitamin D effectively enhances anabolic response, muscle mass, and physical performance in community-dwelling older adults (≥60 years), particularly when baseline levels are deficient, with demonstrated efficacy in elevating 25(OH)D concentrations from suboptimal (28.1 ng/mL) to therapeutic ranges.174 Emerging evidence suggests that combined nutritional strategies incorporating vitamin D3 with whey protein, leucine, calcium, and structured exercise programs yield superior musculoskeletal outcomes compared to monotherapy approaches.173 Importantly, dose-response studies reveal a plateau effect in therapeutic benefits, with high-dose supplementation (3750 IU/day) showing no additional advantage over standard doses (600 IU/day) in overweight elderly populations (>65 years), suggesting the existence of endocrine regulatory mechanisms that maintain vitamin D homeostasis.175
Contrasting these findings, recent randomized controlled trials demonstrate that pharmacological interventions using active vitamin D metabolites fail to produce clinically meaningful improvements in key sarcopenia parameters—including handgrip strength, timed-up-and-go performance, appendicular lean mass, or muscle function—among older adults without severe deficiency.176,177 Notably, even high-dose supplementation (2000 IU/day vitamin D3) showed no significant effects on lower extremity power, muscle strength, or physical performance in functionally impaired elderly individuals (aged 65–89). These contradictory observations highlight critical knowledge gaps regarding the therapeutic application of vitamin D in sarcopenia management, particularly concerning: (1) the necessity of pre-existing deficiency for clinical efficacy, (2) potential differential effects between native vitamin D and active analogs on vitamin D receptor signaling pathways in muscle tissue, and (3) the physiological importance of endogenous 1α-hydroxylation in renal and extrarenal tissues for optimal musculoskeletal outcomes. Resolution of these outstanding questions through carefully designed mechanistic studies will be essential for developing evidence-based vitamin D supplementation strategies tailored to specific sarcopenia phenotypes and metabolic states.
It is important to acknowledge that the Endocrine Society has recently revised its position on defining vitamin D status, as articulated in their latest Guideline Communication.178 After thorough evaluation of existing clinical trial data, the expert panel concluded that current evidence does not support establishing specific 25(OH)D thresholds that reliably predict clinically meaningful benefits from vitamin D supplementation. The Society explicitly withdrew its previous classifications of vitamin D “sufficiency” (≥30 ng/mL [75 nmol/L]) and “insufficiency” (20–30 ng/mL [50–75 nmol/L]), citing substantial limitations in the available evidence and assigning very low certainty to these historical cutoffs. This paradigm shift introduces significant challenges for clinical decision-making regarding vitamin D supplementation in sarcopenia management. The absence of validated biochemical thresholds complicates the identification of patients who might benefit from intervention, particularly given the current lack of consensus on how to interpret serum 25(OH)D levels in the context of musculoskeletal health.
Pharmacological Treatment
Currently, the development of drugs for sarcopenia remains in its early stages. To date, no approved pharmacological agents specifically targeting sarcopenia have been developed, and the management of sarcopenia primarily focuses on strength-training exercises as well as an increased intake of energy-dense foods and a diet rich in protein.179 However, the “Sarcopenia Consensus” highlights several promising agents, including anabolic steroids, vitamin D, growth hormone, β-receptor agonists, angiotensin-converting enzyme inhibitors, myostatin inhibitors, and activin receptor antagonists. These drugs, though not yet widely used for sarcopenia, represent potential therapeutic avenues for future clinical interventions (Table 3).
![]() |
Table 3 Current Status and Pros and Cons of Drug Treatments for Sarcopenia |
Exosome Therapy
In recent years, microvesicles and exosomes have gained significant attention for their promising applications in tissue repair, particularly skeletal muscle regeneration.210 Exosomes secreted by skeletal muscle cells, which carry miRNAs and other regulatory molecules, have been identified as critical regulators of muscle homeostasis.211 For example, a study by Li et al212 demonstrated that exosomes from skeletal muscle cells increased muscle strength by 30% in a mouse model of sarcopenia, underscoring their role in regulating muscle function. Ma et al reported that the use of exosomes derived from human umbilical cord MSCs significantly improved grip strength and increased muscle mass in muscular dystrophy mice.213 Exosomes derived from adipose tissue mesenchymal stem cells (AD-MSCs) have also shown novel paracrine effects on skeletal muscle repair. A study revealed that these exosomes, rich in cytokines, chemokines, and growth factors, enhanced muscle repair by 50% in areas of muscle damage, highlighting their potential in promoting muscle healing.214 Recent research further suggests that exosomes can stimulate muscle regeneration following injury and promote muscle protein synthesis.215 Williams et al216 found that exosome treatment after exercise led to a 25% increase in muscle fiber size and a 20% improvement in muscle strength in aged rats, demonstrating their potential to enhance exercise-induced muscle health.
Although the precise mechanisms through which exosomes exert these effects are not fully understood, ongoing studies are uncovering their therapeutic potential in treating sarcopenia.
Stem Cell Therapy
Mesenchymal stem cells (MSCs) therapy offers a promising approach for substantial diseases due to their self-renew and differentiation potential into various somatic cells.217 Upon injury or stress, MSCs are rapidly activated and quickly proliferate to replenish damaged muscle cells and repair of the muscle fibers structure. Notably, MSCs not only proliferative and differentiation to replace myogenic cells and repair damaged structure, but also secrete paracrine factors that enhance MSC activity and muscle regeneration.218
MSC therapy has demonstrated potential in sarcopenia treatment. For example, Xu et al219 reported a 25% increase in muscle strength and 15% increase in muscle mass in sarcopenic rats treated with MSCs, highlighting their regenerative potential. Additionally, MSC therapy has been shown to restore mitochondrial function in aging muscles.220 Zhao et al221 found a 40% increase in mitochondrial enzyme activity and a 30% reduction in oxidative stress markers in aged muscle tissue following MSC treatment, underscoring its effectiveness in addressing mitochondrial dysfunction associated with sarcopenia.
The extracellular matrix (ECM) and MSCs are crucial for muscle regeneration.222 Disruption of ECM components is associated with impaired muscle regeneration in sarcopenia.223 Previous study suggest that targeting ECM composition—such as using matrix metalloproteinase inhibitors—may improve muscle regeneration.224 Additionally, the myostatin signaling pathway is critical for regulating MSC activity and muscle repair. Myostatin monoclonal antibodies and receptor antagonists can block this pathway, promoting muscle regeneration.225 Ongoing clinical trials are evaluating the efficacy and safety of myostatin inhibitors in elderly sarcopenic patients.
Induced pluripotent stem cells (iPSCs) derived from reprogrammed somatic cells also offer promise, particularly for generating patient-specific stem cells for autologous transplantation, reducing the risk of immune rejection.226
In summary, while traditional treatments for sarcopenia remain limited, emerging evidence supports stem cell therapy as a viable option for enhancing muscle regeneration and restoring mitochondrial function, offering new tools for improved management of sarcopenia.
Critical Unmet Needs and Future Directions
Despite considerable advances in sarcopenia research, several critical challenges persist. First, the absence of universally accepted diagnostic criteria continues to hamper clinical translation, with current tools demonstrating variable sensitivity (particularly for early-stage detection) and limited feasibility in resource-constrained settings. Second, while emerging therapies (eg, exosome-based interventions) show preclinical promise, robust long-term efficacy and safety data from randomized controlled trials remain lacking—a gap further compounded by the paucity of standardized outcome measures. Third, the field would benefit from mechanistic studies elucidating tissue-specific responses to novel interventions across diverse patient populations. Addressing these limitations will require coordinated efforts to: (1) establish validated biomarkers for disease staging; (2) develop pragmatic diagnostic tools for community settings; and (3) implement unified endpoints in therapeutic trials.
Conclusion
The global prevalence of sarcopenia is rising with an aging population, underscoring the urgent need for effective prevention and treatment strategies. Its clinical manifestations are often subtle, making early detection challenging. By the time symptoms become noticeable, significant muscle loss and dysfunction may have already occurred, increasing the risk of falls, fractures, and other complications. Therefore, early screening is crucial. Despite growing awareness, understanding of sarcopenia remains limited, with current diagnostic methods lacking standardization and insufficient evidence on the safety and efficacy of pharmacological treatments.
Emerging therapies, including stem cell and exosome-based approaches, are still under investigation and subject to ongoing debate. While emerging therapeutic modalities—particularly stem cell-derived extracellular vesicles and engineered exosome platforms—show preclinical potential, their translation requires rigorous evaluation through multicenter trials addressing three pivotal questions: (1) long-term safety profiles in metabolically compromised aging cohorts; (2) cost-benefit ratios relative to conventional interventions; and (3) practical implementation barriers across diverse healthcare systems. Concurrently, diagnostic standardization efforts must reconcile sensitivity (particularly for early-stage detection) with clinical feasibility, potentially through multimodal algorithms integrating biochemical markers with functional assessments.
In general, greater emphasis is needed on early screening and the development of comprehensive, multidisciplinary treatment strategies to mitigate the widespread impact of sarcopenia.
Data Sharing Statement
This is a review of the literature, and no research data are reported. Further inquiries can be directed to the corresponding author.
Funding
This research was funded by grants from the National Natural Science Foundation of China (No. 81703584), Guangdong Province Natural Science Foundation of China (No. 2022A1515220166, 2025A1515010196, 2023A1515011091, 2021B1515140054, 2022A1515140138), Discipline construction project of Guangdong Medical University (No. 4SG23002G, and CLP2021B012), the Science and Technology Foundation of Zhanjiang (No. 2022A01099, 2022A01163, and 2022A01170), the Discipline Construction Fund of Central People’s Hospital of Zhanjiang (No. 2022A09), Guangdong medical university research fund (No. FYZM001), and Guangdong Provincial Administration of Chinese Medicine Research Project (No. 20250047, 20245577).
Disclosure
The authors report no conflicts of interest in this work.
References
1. Kim JW, Kim R, Choi H, Lee SJ, Bae GU. Understanding of sarcopenia: from definition to therapeutic strategies. Arch Pharmacal Res. 2021;44(9–10):876–889. doi:10.1007/s12272-021-01349-z
2. Liu S, Zhang L, Li S. Advances in nutritional supplementation for sarcopenia management. Frontiers in Nutrition. 2023;10:1189522. doi:10.3389/fnut.2023.1189522
3. Petermann‐Rocha F, Balntzi V, Gray SR, et al. Global prevalence of sarcopenia and severe sarcopenia: a systematic review and meta‐analysis. J Cachexia Sarcopenia Muscle Nerve. 2022;13(1):86–99. doi:10.1002/jcsm.12783
4. Chen LK, Woo J, Assantachai P, Auyeung TW, Arai H. Asian working group for sarcopenia: 2019 consensus update on sarcopenia diagnosis and treatment. J Am Med Directors Assoc. 2020;21(3):300–307. doi:10.1016/j.jamda.2019.12.012
5. Brzeszczynska J, Meyer A, Mcgregor R, et al. Alterations in the in vitro and in vivo regulation of muscle regeneration in healthy ageing and the influence of sarcopenia. J Cachexia Sarcopenia Muscle. 2018;9(1):93–105. doi:10.1002/jcsm.12252
6. Moore DR. Keeping older muscle “young” through dietary protein and physical activity. Adv Nutr. 2014;5(5):599S–S607. doi:10.3945/an.113.005405
7. Cruz-Jentoft AJ, Bahat G, Bauer J, et al. Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing. 2019;48(4):601. doi:10.1093/ageing/afz046
8. Kirk B, Cawthon PM, Arai H, et al. The conceptual definition of sarcopenia: delphi consensus from the Global Leadership Initiative in Sarcopenia (GLIS). Age Ageing. 2024;53(3):052. doi:10.1093/ageing/afae052
9. Sayer AA, Cooper R, Arai H, et al. Sarcopenia. Nat Rev Dis Prim. 2024;10(1):68. doi:10.1038/s41572-024-00550-w
10. Petermann-Rocha F, Balntzi V, Gray SR, et al. Global prevalence of sarcopenia and severe sarcopenia: a systematic review and meta-analysis. J Cachexia Sarcopenia Muscle. 2022;13(1):86–99.
11. Yuan S, Larsson SC. Epidemiology of sarcopenia: prevalence, risk factors, and consequences. Metabolism. 2023;144:155533. doi:10.1016/j.metabol.2023.155533
12. Papadopoulou S, Tsintavis P, Potsaki G, Papandreou D. Differences in the prevalence of sarcopenia in community-dwelling, nursing home and hospitalized individuals. A systematic review and meta-analysis. J Nutr Health Aging. 2020;24(1):83–90. doi:10.1007/s12603-019-1267-x
13. Huang T, Li C, Chen F, et al. Prevalence and risk factors of osteosarcopenia: a systematic review and meta-analysis. BMC Geriatr. 2023;23(1):369. doi:10.1186/s12877-023-04085-9
14. Damluji AA, Alfaraidhy M, AlHajri N, et al. Sarcopenia and cardiovascular diseases. Circulation. 2023;147(20):1534–1553. doi:10.1161/CIRCULATIONAHA.123.064071
15. Papadopoulou SK, Tsintavis P, Potsaki G, Papandreou D. Differences in the prevalence of sarcopenia in community-dwelling, nursing home and hospitalized individuals. A systematic review and meta-analysis. j nutr health aging. 2020;24(1):83–90.
16. Wang B, Nong C, Zhang J, et al. Prevalence and associated body composition factors of sarcopenia in community-dwelling older adults. Eur J Med Res. 2024;29(1):598. doi:10.1186/s40001-024-02185-9
17. Cao M, Lian J, Lin X, et al. Prevalence of sarcopenia under different diagnostic criteria and the changes in muscle mass, muscle strength, and physical function with age in Chinese old adults. BMC Geriatr. 2022;22(1):889. doi:10.1186/s12877-022-03601-7
18. Zagami F. Overlapping pathophysiological pathways between sarcopenia and chronic diseases. DYSONA - Life Science. 2023;4(1):10–22.
19. Mellen RH, Girotto OS, Marques EB, et al. Insights into pathogenesis, nutritional and drug approach in sarcopenia: a systematic review. Biomedicines. 2023;11(1):136. doi:10.3390/biomedicines11010136
20. Witham MD, Heslop P, Dodds RM, et al. Performance of the SarQoL quality of life tool in a UK population of older people with probable sarcopenia and implications for use in clinical trials: findings from the SarcNet registry. BMC Geriatr. 2022;22(1):368. doi:10.1186/s12877-022-03077-5
21. Demonceau C, Voz B, Bruyère O, Reginster JY, Beaudart C. Content validity of SarQoL, a quality of life questionnaire specific to sarcopenia. Aging Clin Exp Res. 2024;36(1):101. doi:10.1007/s40520-024-02756-0
22. Beaudart C, Reginster JY, Bruyère O, Geerinck A. Quality of life and sarcopenia. Sarcopenia. 2021;21:279–304.
23. Beaudart C, Demonceau C, Reginster JY, et al. Sarcopenia and health-related quality of life: a systematic review and meta-analysis. J Cachexia Sarcopenia Muscle. 2023;14(3):1228–1243. doi:10.1002/jcsm.13243
24. Beaudart C, Tilquin N, Abramowicz P, et al. Quality of life in sarcopenia measured with the SarQoL questionnaire: a meta-analysis of individual patient data. Maturitas. 2024;180:107902. doi:10.1016/j.maturitas.2023.107902
25. Qaisar R, Iqbal MS, Karim A, Muhammad T, Ahmad F. A leaky gut contributes to reduced sarcopenia-related quality of life (SarQoL) in geriatric older adults. Qual Life Res. 2024;33(2):551–559. doi:10.1007/s11136-023-03547-2
26. Alhmly HF, Fielding RA. A critical review of current worldwide definitions of sarcopenia. Calcif Tissue Int. 2024;114(1):74–81. doi:10.1007/s00223-023-01163-3
27. Jung HN, Jung CH, Hwang YC. Sarcopenia in youth. Metabolism. 2023;144:155557. doi:10.1016/j.metabol.2023.155557
28. Kim SW, Park HY, Jung WS, Lim K. Effects of twenty-four weeks of resistance exercise training on body composition, bone mineral density, functional fitness and isokinetic muscle strength in obese older women: a randomized controlled trial. Int J Environ Agric Res. 2022;19(21):14554.
29. Cruz-Jentoft AJ, Sayer AA. Sarcopenia. Lancet. 2019;393(10191):2636–2646. doi:10.1016/S0140-6736(19)31138-9
30. Larsson L, Degens H, Li M, et al. Sarcopenia: aging-related loss of muscle mass and function. Physiol Rev. 2019;99(1):427–511. doi:10.1152/physrev.00061.2017
31. Christian CJ, Benian GM. Animal models of sarcopenia. Aging Cell. 2020;19(10):e13223. doi:10.1111/acel.13223
32. Tanganelli F, Meinke P, Hofmeister F, et al. Type-2 muscle fiber atrophy is associated with sarcopenia in elderly men with Hip fracture. Exp Gerontology. 2021;144:111171. doi:10.1016/j.exger.2020.111171
33. Arnold WD, Clark BC. Neuromuscular junction transmission failure in aging and sarcopenia: the nexus of the neurological and muscular systems. Ageing Res Rev. 2023;89:101966. doi:10.1016/j.arr.2023.101966
34. Kamper RS, Alcazar J, Andersen LL, et al. Associations between inflammatory markers, body composition, and physical function: the Copenhagen sarcopenia study. J Cachexia Sarcopenia Muscle. 2021;12(6):1641–1652. doi:10.1002/jcsm.12832
35. Dowling P, Gargan S, Swandulla D, Ohlendieck K. Fiber-type shifting in sarcopenia of old age: proteomic profiling of the contractile apparatus of skeletal muscles. Int J Mol Sci. 2023;24(3):2415. doi:10.3390/ijms24032415
36. Sousa-Victor P, García-Prat L, Muñoz-Cánoves P. Control of satellite cell function in muscle regeneration and its disruption in ageing. Nat Rev Mol Cell Biol. 2022;23(3):204–226. doi:10.1038/s41580-021-00421-2
37. Pillon NJ, Bilan PJ, Fink LN, Klip A. Cross-talk between skeletal muscle and immune cells: muscle-derived mediators and metabolic implications. Am J Physiol Endocrinol Metab. 2013;304(5):E453–E465. doi:10.1152/ajpendo.00553.2012
38. Johnson AL, Kamal M, Parise G. The role of supporting cell populations in satellite cell mediated muscle repair. Cells. 2023;12(15):1968. doi:10.3390/cells12151968
39. Granic A, Suetterlin K, Shavlakadze T, Grounds MD, Sayer AA. Hallmarks of ageing in human skeletal muscle and implications for understanding the pathophysiology of sarcopenia in women and men. Clin Sci. 2023;137(22):1721–1751. doi:10.1042/CS20230319
40. Farup J, Madaro L, Puri PL, Mikkelsen UR. Interactions between muscle stem cells, mesenchymal-derived cells and immune cells in muscle homeostasis, regeneration and disease. Cell Death Dis. 2015;6(7):e1830–e1830. doi:10.1038/cddis.2015.198
41. Fernández-Lázaro D, Garrosa E, Seco-Calvo J, Garrosa M. Potential satellite cell-linked biomarkers in aging skeletal muscle tissue: proteomics and proteogenomics to monitor sarcopenia. Proteomes. 2022;10(3):29. doi:10.3390/proteomes10030029
42. López-Otín C, Pietrocola F, Roiz-Valle D, Galluzzi L, Kroemer G. Meta-hallmarks of aging and cancer. Cell Metab. 2023;35(1):12–35. doi:10.1016/j.cmet.2022.11.001
43. Monti E, Tagliaferri S, Zampieri S, et al. Effects of a 2‐year exercise training on neuromuscular system health in older individuals with low muscle function. J Cachexia Sarcopenia Muscle Nerve. 2023;14(2):794–804. doi:10.1002/jcsm.13173
44. de Almeida AC, Aily JB, Pedroso MG, et al. A periodized training attenuates thigh intermuscular fat and improves muscle quality in patients with knee osteoarthritis: results from a randomized controlled trial. Clin Rheumatol. 2020;39(4):1265–1275. doi:10.1007/s10067-019-04892-9
45. Xu J, Liao C, Yin -C-C, Li G, Zhu Y, Sun F. In situ structural insights into the excitation-contraction coupling mechanism of skeletal muscle. Sci Adv. 2024;10(12):eadl1126. doi:10.1126/sciadv.adl1126
46. Zhang T, Pereyra AS, Wang ZM, et al. Calpain inhibition rescues troponin T3 fragmentation, increases Cav1.1, and enhances skeletal muscle force in aging sedentary mice. Aging Cell. 2016;15(3):488–498. doi:10.1111/acel.12453
47. Ancel S, Stuelsatz P, Feige JN. Muscle stem cell quiescence: controlling stemness by staying asleep. Trends Cell Biol. 2021;31(7):556–568. doi:10.1016/j.tcb.2021.02.006
48. Lee D, Bareja A, Bartlett D, White J. Autophagy as a therapeutic target to enhance aged muscle regeneration. Cells. 2019;8(2):183. doi:10.3390/cells8020183
49. Revuelta M, Matheu A. Autophagy in stem cell aging. Aging Cell. 2017;16(5):912–915. doi:10.1111/acel.12655
50. Sakuma K, Aizawa M, Wakabayashi H, Yamaguchi A. The autophagy-dependent signaling in skeletal muscle. Plasticity Skelet Musc. 2017;5:93–111.
51. Racine J. The role of nix-mediated mitophagy in muscle stem cell fate decision and muscle regeneration. Université d’Ottawa/University of Ottawa. 2024.
52. Beloosesky Y, Hendel D, Weiss A, et al. Cytokines and C-reactive protein production in hip-fracture-operated elderly patients. J Gerontol Ser A. 2007;62(4):420–426. doi:10.1093/gerona/62.4.420
53. Sharma B, Dabur R. Role of pro-inflammatory cytokines in regulation of skeletal muscle metabolism: a systematic review. Curr Med Chem. 2020;27(13):2161–2188. doi:10.2174/0929867326666181129095309
54. He Y, Duan W, Xu P, et al. Exploring the impact of interleukins on sarcopenia development: a systematic review and meta-analysis. Exp Gerontology. 2024;193:112480. doi:10.1016/j.exger.2024.112480
55. Singh A, Yadav A, Phogat J, Dabur R. Dynamics and interplay between autophagy and ubiquitin-proteasome system coordination in skeletal muscle atrophy. Curr Mol Pharmacol. 2022;15(3):475–486. doi:10.2174/1874467214666210806163851
56. Yong-Zhan Z, Pei-Jie C, Wei-Hua X, Kinesiology SO. Mechanism of the occurrence of sarcopenia in the elderly. Sheng Li xue Bao. 2018;70(4):445–454.
57. Rong Y-D, Bian A-L, Hu H-Y, Ma Y, Zhou X-Z. Study on relationship between elderly sarcopenia and inflammatory cytokine IL-6, anti-inflammatory cytokine IL-10. BMC Geriatr. 2018;18:1–6. doi:10.1186/s12877-018-1007-9
58. Huang LT, Wang JH. The Therapeutic Intervention of Sex Steroid Hormones for Sarcopenia. Front Med. 2021;8:739251. doi:10.3389/fmed.2021.739251
59. Feng LT, Chen ZN, Bian H. Skeletal muscle: molecular structure, myogenesis, biological functions, and diseases. MedCom. 2024;5(7):e649. doi:10.1002/mco2.649
60. Rizk J, Sahu R, Duteil D. An overview on androgen-mediated actions in skeletal muscle and adipose tissue. Steroids. 2023;199:109306. doi:10.1016/j.steroids.2023.109306
61. Hansen M. Female hormones: do they influence muscle and tendon protein metabolism? Proc Nutr Soc. 2018;77(1):32–41. doi:10.1017/S0029665117001951
62. Volkert D, Beck AM, Cederholm T, et al. ESPEN practical guideline: clinical nutrition and hydration in geriatrics. Clin Nutr. 2022;41(4):958–989. doi:10.1016/j.clnu.2022.01.024
63. Liu Y, Wang Z, Cao J, Dong Y, Gao T, Chen Y. Effects of thyroid hormone on monochromatic light combinations mediate skeletal muscle fiber pattern in broilers. Poultr Sci. 2024;103(9):103999. doi:10.1016/j.psj.2024.103999
64. Chen J, Wei L, Zhu X, et al. TT3, a more practical indicator for evaluating the relationship between sarcopenia and thyroid hormone in the euthyroid elderly compared with FT3. Clin Interv Aging. 2023;18:1285–1293. doi:10.2147/CIA.S420558
65. Candow DG, Chilibeck PD, Forbes SC, Fairman CM, Gualano B, Roschel H. Creatine supplementation for older adults: focus on sarcopenia, osteoporosis, frailty and Cachexia. Bone. 2022;162:116467. doi:10.1016/j.bone.2022.116467
66. Chapman-Lopez T, Wilburn D, Fletcher E, et al. The influence of resistance training on adipokines in post-menopausal women: a brief review. Sports Med Health Sci. 2022;4(4):219–224. doi:10.1016/j.smhs.2022.09.001
67. Faulkner JA, Larkin LM, Claflin DR, Brooks SV. Age-related changes in the structure and function of skeletal muscles. Clin Exp Pharmacol Physiol. 2010;34(11):1091–1096. doi:10.1111/j.1440-1681.2007.04752.x
68. Heidari D, Shirvani H, Bazgir B, Shamsoddini A. The resistance training effects on skeletal muscle stem cells in older adult: a systematic review and meta-analysis. Cell Journal. 2023;25(8):513–523. doi:10.22074/cellj.2023.1986679.1206
69. Dalle S, Rossmeislova L, Koppo K. The role of inflammation in age-related sarcopenia. Front Physiol. 2017;8:1045. doi:10.3389/fphys.2017.01045
70. Reid N, Healy GN, Gianoudis J, et al. Association of sitting time and breaks in sitting with muscle mass, strength, function, and inflammation in community-dwelling older adults. Osteoporosis Int. 2018;29(6):1341–1350. doi:10.1007/s00198-018-4428-6
71. Akune T, Muraki S, Oka H, et al. Exercise habits during middle age are associated with lower prevalence of sarcopenia: the ROAD study. Osteoporosis Int. 2014;25(3):1081–1088. doi:10.1007/s00198-013-2550-z
72. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: an expanding universe. Cell. 2023;186(2):243–278. doi:10.1016/j.cell.2022.11.001
73. Marzetti E, Calvani R, Coelho-Júnior HJ, Landi F, Picca A. Mitochondrial quantity and quality in age-related sarcopenia. Int J Mol Sci. 2024;25(4):2052. doi:10.3390/ijms25042052
74. Affourtit C, Carré JE. Mitochondrial involvement in sarcopenia. Acta Physiol. 2024;240(3):e14107. doi:10.1111/apha.14107
75. Amorim JA, Coppotelli G, Rolo AP, Palmeira CM, Ross JM, Sinclair DA. Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat Rev Endocrinol. 2022;18(4):243–258. doi:10.1038/s41574-021-00626-7
76. Abreu P, Kowaltowski AJ. Satellite cell self‐renewal in endurance exercise is mediated by inhibition of mitochondrial oxygen consumption. J Cachexia Sarcopenia Muscle. 2020;11(6):1661–1676. doi:10.1002/jcsm.12601
77. Tamaki M, Miyashita K, Wakino S, Mitsuishi M, Hayashi K, Itoh H. Chronic kidney disease reduces muscle mitochondria and exercise endurance and its exacerbation by dietary protein through inactivation of pyruvate dehydrogenase. Kidney Int. 2014;85(6):1330–1339. doi:10.1038/ki.2013.473
78. Stuart JA, Maddalena LA, Merilovich M, Robb EL. A midlife crisis for the mitochondrial free radical theory of aging. Longevity Healthspan. 2014;3(1):1–15. doi:10.1186/2046-2395-3-4
79. Campos F, Abrigo J, Aguirre F, et al. Sarcopenia in a mice model of chronic liver disease: role of the ubiquitin–proteasome system and oxidative stress. Pflügers Archiv-Euro J Physiol. 2018;470:1503–1519. doi:10.1007/s00424-018-2167-3
80. Chen TH, Koh KY, Lin KM, Chou CK. Mitochondrial dysfunction as an underlying cause of skeletal muscle disorders. Int J Mol Sci. 2022;23(21):12926. doi:10.3390/ijms232112926
81. Nunes-Pinto M, Bandeira de Mello RG, Pinto MN, et al. Sarcopenia and the biological determinants of aging: a narrative review from a geroscience perspective. Ageing Res Rev. 2025;103:102587. doi:10.1016/j.arr.2024.102587
82. Li Y, Li S, Wu H. Ubiquitination-proteasome system (UPS) and autophagy two main protein degradation machineries in response to cell stress. Cells. 2022;11(5):851. doi:10.3390/cells11050851
83. Xie G, Jin H, Mikhail H, et al. Autophagy in sarcopenia: possible mechanisms and novel therapies. Biomed Pharmacother. 2023;165:115147. doi:10.1016/j.biopha.2023.115147
84. Irazoki A, Martinez‐Vicente M, Aparicio P, et al. Coordination of mitochondrial and lysosomal homeostasis mitigates inflammation and muscle atrophy during aging. Aging Cell. 2022;21(4):e13583. doi:10.1111/acel.13583
85. Gonzalez-Freire M, Adelnia F, Moaddel R, Ferrucci L. Searching for a mitochondrial root to the decline in muscle function with ageing. J Cachexia Sarcopenia Muscle. 2018;9(3):435–440. doi:10.1002/jcsm.12313
86. Alcazar J, Rodriguez-Lopez C, Delecluse C, Thomis M, Van Roie E. Ten-year longitudinal changes in muscle power, force, and velocity in young, middle-aged, and older adults. J Cachexia Sarcopenia Muscle. 2023;14(2):1019–1032. doi:10.1002/jcsm.13184
87. Hai S, Cao L, Wang H. Association between sarcopenia and nutritional status and physical activity among community-dwelling Chinese adults aged 60 years and older. Geriatrics Gerontol Int. 2017;17(11):1959–1966. doi:10.1111/ggi.13001
88. Traylor DA, Gorissen SHM, Phillips SM. Perspective: protein requirements and optimal intakes in aging: are we ready to recommend more than the recommended daily allowance? Adv Nutr. 2018;9(3):171–182. doi:10.1093/advances/nmy003
89. Montenegro KR, Carlessi R, Cruzat V, Newsholme P. Effects of vitamin D on primary human skeletal muscle cell proliferation, differentiation, protein synthesis and bioenergetics. J Steroid Biochem Mol Biol. 2019;193:105423. doi:10.1016/j.jsbmb.2019.105423
90. Nakamura S, Sato Y, Kobayashi T, et al. Vitamin D protects against immobilization-induced muscle atrophy via neural crest-derived cells in mice. Sci Rep. 2020;10(1):12242. doi:10.1038/s41598-020-69021-y
91. Dzik KP, Kaczor JJ. Mechanisms of vitamin D on skeletal muscle function: oxidative stress, energy metabolism and anabolic state. Eur J Appl Physiol. 2019;119:825–839. doi:10.1007/s00421-019-04104-x
92. Bass JJ, Nakhuda A, Deane CS, et al. Overexpression of the vitamin D receptor (VDR) induces skeletal muscle hypertrophy. Mol Metabol. 2020;42:101059. doi:10.1016/j.molmet.2020.101059
93. Montenegro KR, Cruzat V, Carlessi R, Newsholme P. Mechanisms of vitamin D action in skeletal muscle. Nutr Res Rev. 2019;32(2):192–204. doi:10.1017/S0954422419000064
94. Harvey NC, Biver E, Kaufman JM, et al. The role of calcium supplementation in healthy musculoskeletal ageing: an expert consensus meeting of the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO) and the International Foundation for Osteoporosis (IOF). Osteoporosis Int. 2017;28(2):447–462. doi:10.1007/s00198-016-3773-6
95. Kord-Varkaneh H, Rinaldi G, Hekmatdoost A, et al. The influence of vitamin D supplementation on IGF-1 levels in humans: a systematic review and meta-analysis. Ageing Res Rev. 2020;57:100996. doi:10.1016/j.arr.2019.100996
96. Yoshida T, Delafontaine P. Mechanisms of IGF-1-mediated regulation of skeletal muscle hypertrophy and atrophy. Cells. 2020;9(9):1970. doi:10.3390/cells9091970
97. Mizuno T, Hosoyama T, Tomida M, et al. Influence of vitamin D on sarcopenia pathophysiology: a longitudinal study in humans and basic research in knockout mice. J Cachexia Sarcopenia Muscle. 2022;13(6):2961–2973. doi:10.1002/jcsm.13102
98. Wang J, Wang X, Gu Y, et al. Vitamin D is related to handgrip strength in adult men aged 50 years and over: a population study from the TCLSIH cohort study. Clin Endocrinol. 2019;90(5):753–765. doi:10.1111/cen.13952
99. Mendoza-Garcés L, Velázquez-Alva MC, Cabrer-Rosales MF, Arrieta-Cruz I, Gutiérrez-Juárez R, Irigoyen-Camacho ME. Vitamin D deficiency is associated with handgrip strength, nutritional status and T2DM in community-dwelling older Mexican women: a cross-sectional study. Nutrients. 2021;13(3):736. doi:10.3390/nu13030736
100. Aspell N, Laird E, Healy M, Lawlor B, O’Sullivan M. Vitamin D deficiency is associated with impaired muscle strength and physical performance in community-dwelling older adults: findings from the English longitudinal study of ageing. Clin Intervent Aging. 2019;14:1751–1761. doi:10.2147/CIA.S222143
101. de Lima EP, Tanaka M, Lamas CB, et al. Vascular impairment, muscle atrophy, and cognitive decline: critical age-related conditions. Biomedicines. 2024;12(9):2096. doi:10.3390/biomedicines12092096
102. Troutman AD, Arroyo E, Sheridan EM, et al. Skeletal muscle atrophy in clinical and preclinical models of chronic kidney disease: a systematic review and meta‐analysis. J Cachexia Sarcopenia Muscle. 2024;15(1):21–35. doi:10.1002/jcsm.13400
103. Zheng Y, Feng J, Yu Y, Ling M, Wang X. Advances in sarcopenia: mechanisms, therapeutic targets, and intervention strategies. Arch Pharmacal Res. 2024;47(4):301–324. doi:10.1007/s12272-024-01493-2
104. Hou L, Liu Y, Li X, et al. Changes and risk factors of skeletal muscle mass and strength in patients with type 2 diabetes over 60 years old: a cross‐sectional study from China. J Diabetes Res. 2020;2020(1):9815485. doi:10.1155/2020/9815485
105. Chatzipetrou V, Bégin MJ, Hars M, Trombetti A. Sarcopenia in chronic kidney disease: a scoping review of prevalence, risk factors, association with outcomes, and treatment. Calcif Tissue Int. 2022;110(1):1–31. doi:10.1007/s00223-021-00898-1
106. Ren H, Gong D, Jia F, Xu B, Liu Z. Sarcopenia in patients undergoing maintenance hemodialysis: incidence rate, risk factors and its effect on survival risk. Renal Failure. 2016;38(3):364–371. doi:10.3109/0886022X.2015.1132173
107. Rungratanawanich W, Qu Y, Wang X, Essa MM, Song BJ. Advanced glycation end products (AGEs) and other adducts in aging-related diseases and alcohol-mediated tissue injury. Exp Mol Med. 2021;53(2):168–188. doi:10.1038/s12276-021-00561-7
108. Zhang FM, Wu HF, Shi HP, Yu Z, Zhuang CL. Sarcopenia and malignancies: epidemiology, clinical classification and implications. Ageing Res Rev. 2023;91:102057. doi:10.1016/j.arr.2023.102057
109. Anjanappa M, Corden M, Green A, et al. Sarcopenia in cancer: risking more than muscle loss. Tech Innov Patient Support Radiat Oncol. 2020;16:50–57. doi:10.1016/j.tipsro.2020.10.001
110. Luo L, Shen X, Fang S, et al. Sarcopenia as a risk factor of progression-free survival in patients with metastases: a systematic review and meta-analysis. BMC Cancer. 2023;23(1):127. doi:10.1186/s12885-023-10582-2
111. Carbone S, Billingsley HE, Rodriguez-Miguelez P, et al. Lean mass abnormalities in heart failure: the role of sarcopenia, sarcopenic obesity, and cachexia. Curr Prob Cardiol. 2020;45(11):100417. doi:10.1016/j.cpcardiol.2019.03.006
112. Takamori S, Toyokawa G, Okamoto T, Shimokawa M, Maehara Y. Clinical impact and risk factors for skeletal muscle loss after complete resection of early non-small cell lung cancer. Ann Surg Oncol. 2018;25(5):1–8. doi:10.1245/s10434-017-6261-0
113. Sheng Y, Ma D, Zhou Q, et al. Association of thyroid function with sarcopenia in elderly Chinese euthyroid subjects. Aging Clin Exp Res. 2019;31:1113–1120. doi:10.1007/s40520-018-1057-z
114. Elnakish MT, Schultz EJ, Gearinger RL, et al. Differential involvement of various sources of reactive oxygen species in thyroxin-induced hemodynamic changes and contractile dysfunction of the heart and diaphragm muscles. Biophys J. 2015;108(2):292a. doi:10.1016/j.bpj.2014.11.1594
115. Di Cosmo C, Liao X-H, Ye H, et al. Mct8-deficient mice have increased energy expenditure and reduced fat mass that is abrogated by normalization of serum T3 levels. Endocrinology. 2013;154(12):4885–4895. doi:10.1210/en.2013-1150
116. Bloise FF, Oliveira TS, Cordeiro A, Ortiga-Carvalho TM. Thyroid hormones play role in sarcopenia and myopathies. Front Physiol. 2018;9:355610. doi:10.3389/fphys.2018.00560
117. Aslam MA, Ma EB, Huh JY. Pathophysiology of sarcopenia: genetic factors and their interplay with environmental factors. Metabolism. 2023;149:155711. doi:10.1016/j.metabol.2023.155711
118. Roth SM. Genetic aspects of skeletal muscle strength and mass with relevance to sarcopenia. Bonekey Rep. 2012;1:58. doi:10.1038/bonekey.2012.58
119. Yin KF, Chen T, Gu XJ, et al. Systematic druggable genome‐wide Mendelian randomization identifies therapeutic targets for sarcopenia. J Cachexia Sarcopenia Muscle. 2024;15(4):1324–1334. doi:10.1002/jcsm.13479
120. Urzi F, Pokorny B, Buzan E. Pilot study on genetic associations with age-related sarcopenia. Front Genetics. 2020;11:615238. doi:10.3389/fgene.2020.615238
121. Du J, Wu Q, Bae EJ. Epigenetics of skeletal muscle atrophy. Int J Mol Sci. 2024;25(15):8362. doi:10.3390/ijms25158362
122. Liang W, Xu F, Li L, et al. Epigenetic control of skeletal muscle atrophy. Cell Mol Biol Lett. 2024;29(1):99.
123. Antoun E, Garratt ES, Taddei A, et al. Epigenome-wide association study of sarcopenia: findings from the Hertfordshire Sarcopenia Study (HSS). J Cachexia Sarcopenia Muscle. 2022;13(1):240–253. doi:10.1002/jcsm.12876
124. Wang Z, Yao H, Lin S, et al. Transcriptional and epigenetic regulation of human microRNAs. Cancer Lett. 2013;331(1):1–10. doi:10.1016/j.canlet.2012.12.006
125. Bianchi M, Renzini A, Adamo S, Moresi V. Coordinated actions of microRNAs with other epigenetic factors regulate skeletal muscle development and adaptation. Int J Mol Sci. 2017;18(4):840. doi:10.3390/ijms18040840
126. Li J-W, Shen Z-K, Lin Y-S, et al. DNA methylation of skeletal muscle function-related secretary factors identifies FGF2 as a potential biomarker for sarcopenia. J Cachexia Sarcopenia Muscle. 2024;15(3):1209–1217. doi:10.1002/jcsm.13472
127. Livshits G, Kalinkovich A. Restoration of epigenetic impairment in the skeletal muscle and chronic inflammation resolution as a therapeutic approach in sarcopenia. Ageing Res Rev. 2024;96:102267. doi:10.1016/j.arr.2024.102267
128. Lorenzo I, Serra-Prat M, Yébenes JC. The role of water homeostasis in muscle function and frailty: a review. Nutrients. 2019;11(8):1857. doi:10.3390/nu11081857
129. Hioka A, Akazawa N, Okawa N, Nagahiro S. Influence of aging on extracellular water-to-total body water ratio in community-dwelling females. Clin Nutr ESPEN. 2024;60:73–78. doi:10.1016/j.clnesp.2024.01.007
130. Miao Y, Xie L, Song J, et al. Unraveling the causes of sarcopenia: roles of neuromuscular junction impairment and mitochondrial dysfunction. Physiol Rep. 2024;12(1):e15917. doi:10.14814/phy2.15917
131. Rodríguez Cruz PM, Cossins J, Beeson D, Vincent A. The neuromuscular junction in health and disease: molecular mechanisms governing synaptic formation and homeostasis. Front Mol Neurosci. 2020;13:610964.
132. Fatima R, Kim Y, Baek S, Suram RP, An SL, Hong Y. C-terminal agrin fragment as a biomarker for sarcopenia: a systematic review and meta-analysis. J Cachexia Sarcopenia Muscle. 2025;16(1):e13707. doi:10.1002/jcsm.13707
133. Kumar P, Nayak K, Umakanth S, Girish N. Effect of targeted intervention on C-terminal agrin fragment and its association with the components of sarcopenia: a scoping review. Aging Clin Exp Res. 2023;35(6):1161–1186. doi:10.1007/s40520-023-02396-w
134. Monti E, Sarto F, Sartori R, et al. C-terminal agrin fragment as a biomarker of muscle wasting and weakness: a narrative review. J Cachexia Sarcopenia Muscle. 2023;14(2):730–744. doi:10.1002/jcsm.13189
135. Pratt J, De Vito G, Narici M, et al. Plasma C-terminal agrin fragment as an early biomarker for sarcopenia: results from the genofit study. J Gerontol a Biol Sci Med Sci. 2021;76(12):2090–2096. doi:10.1093/gerona/glab139
136. Bao Z, Cui C, Chow SK, Qin L, Wong RMY, Cheung WH. AChRs degeneration at NMJ in aging-associated sarcopenia-a systematic review. Front Aging Neurosci. 2020;12:597811. doi:10.3389/fnagi.2020.597811
137. Wu J, Ding P, Wu H, et al. Sarcopenia: molecular regulatory network for loss of muscle mass and function. Frontiers in Nutrition. 2023;10:1037200. doi:10.3389/fnut.2023.1037200
138. Li Z, Wang Q, Huang X, Wu Y, Shan D. Microbiome’s role in musculoskeletal health through the gut-bone axis insights. Gut Microbes. 2024;16(1):2410478. doi:10.1080/19490976.2024.2410478
139. Frampton J, Murphy KG, Frost G, Chambers ES. Short-chain fatty acids as potential regulators of skeletal muscle metabolism and function. Nat Metab. 2020;2(9):840–848. doi:10.1038/s42255-020-0188-7
140. Lefevre C, Bindels LB. Role of the gut microbiome in skeletal muscle physiology and pathophysiology. Curr Osteoporosis Rep. 2022;20(6):422–432. doi:10.1007/s11914-022-00752-9
141. Grosicki GJ, Fielding RA, Lustgarten MS. Gut microbiota contribute to age-related changes in skeletal muscle size, composition, and function: biological basis for a gut-muscle axis. Calcif Tissue Int. 2018;102(4):433–442. doi:10.1007/s00223-017-0345-5
142. Chen F, Li Q, Chen Y, et al. Association of the gut microbiota and fecal short-chain fatty acids with skeletal muscle mass and strength in children. FASEB J. 2022;36(1):e22109. doi:10.1096/fj.202002697RRR
143. Liu H, Xi Q, Tan S, et al. The metabolite butyrate produced by gut microbiota inhibits cachexia-associated skeletal muscle atrophy by regulating intestinal barrier function and macrophage polarization. Int Immunopharmacol. 2023;124:111001. doi:10.1016/j.intimp.2023.111001
144. De Spiegeleer A, Elewaut D, Van Den Noortgate N, et al. Quorum sensing molecules as a novel microbial factor impacting muscle cells. Biochimica Et Biophysica Acta (BBA) - Molecular Basis of Disease. 2020;1866(3):165646. doi:10.1016/j.bbadis.2019.165646
145. De Spiegeleer A, Wynendaele E, Descamps A, et al. The bacterial quorum sensing peptide iAM373 is a novel inducer of sarcopenia. Clin Transl Med. 2022;12(10):e1053. doi:10.1002/ctm2.1053
146. Mo X, Shen L, Cheng R, et al. Faecal microbiota transplantation from young rats attenuates age-related sarcopenia revealed by multiomics analysis. J Cachexia Sarcopenia Muscle. 2023;14(5):2168–2183. doi:10.1002/jcsm.13294
147. Kumar P, Umakanth S, Girish N. A review of the components of exercise prescription for sarcopenic older adults. Eur Geriatric Med. 2022;13(6):1245–1280. doi:10.1007/s41999-022-00693-7
148. Coelho-Júnior HJ, Calvani R, Picca A, Tosato M, Landi F, Marzetti E. Engagement in aerobic exercise is associated with a reduced prevalence of sarcopenia and severe sarcopenia in Italian older adults. J Pers Med. 2023;13(4):655. doi:10.3390/jpm13040655
149. Izquierdo M, Merchant RA, Morley JE, et al. International Exercise Recommendations in Older Adults (ICFSR): expert consensus guidelines. j nutr health aging. 2021;25(7):824–853.
150. Ruiz JG, Dent E, Morley JE, et al. Screening for and managing the person with frailty in primary care: ICFSR consensus guidelines. j nutr health aging. 2020;24(9):920–927. doi:10.1007/s12603-020-1498-x
151. Lee SY, Tung HH, Liu CY, Chen LK. Physical activity and sarcopenia in the geriatric population: a systematic review. J Am Med Directors Assoc. 2018;19(5):378–383. doi:10.1016/j.jamda.2018.02.003
152. Chen N, He X, Feng Y, Ainsworth BE, Liu Y. Effects of resistance training in healthy older people with sarcopenia: a systematic review and meta-analysis of randomized controlled trials. Eur Rev Aging Phys Activity. 2021;18(1):23. doi:10.1186/s11556-021-00277-7
153. Hurst C, Robinson SM, Witham MD, et al. Resistance exercise as a treatment for sarcopenia: prescription and delivery. Age Ageing. 2022;51(2):afac003. doi:10.1093/ageing/afac003
154. Talar K, Hernández-Belmonte A, Vetrovsky T, Steffl M, Kałamacka E, Courel-Ibáñez J. Benefits of resistance training in early and late stages of frailty and sarcopenia: a systematic review and meta-analysis of randomized controlled studies. J Clin Med. 2021;10(8):1630. doi:10.3390/jcm10081630
155. Kumar P, Umakanth S, Marzetti E, Kalra S, G N. Four-step co-designing of the reablement strategies targeting Sarcopenia (ReStart-S): an exercise-based multicomponent program for older adults residing in long-term care settings. J Multidiscip Healthc. 2024;Volume 17:1415–1433. doi:10.2147/JMDH.S452269
156. Wang H, Huang WY, Zhao Y. Efficacy of exercise on muscle function and physical performance in older adults with sarcopenia: an updated systematic review and meta-analysis. Int J Environ Res Public Health. 2022;19(13):8212. doi:10.3390/ijerph19138212
157. Kumar P, Umakanth S, Bruyère O, G N. Effect of Reablement Strategies targeting Sarcopenia (ReStart-S) on sarcopenic older adults residing in long term care settings: a protocol for cluster randomized trial. Heliyon. 2025;11(3):e42174. doi:10.1016/j.heliyon.2025.e42174
158. Xue J, Han X, Zheng Y, Zhang Q, Kong L. Effectiveness of resistance training in modulating inflammatory biomarkers among Asian patients with sarcopenia: a systematic review and meta-analysis of randomized controlled trials. Front Immunol. 2024;15:1385902. doi:10.3389/fimmu.2024.1385902
159. Almeida HC, Lage VKS, Taiar R, et al. Effect of whole-body vibration stimulation on plasma soluble TNF receptors in elderly with sarcopenia: a randomized controlled trial. Braz J Med Biol Res. 2024;57:e13282.
160. Li W, Chen M, Chen F, et al. Vitamin D combined with whole-body vibration training for the treatment of osteo-sarcopenia: study protocol for a randomized controlled trial. Trials. 2024;25(1):638. doi:10.1186/s13063-024-08498-8
161. Richter M, Baerlocher K, Bauer JM, et al. Revised reference values for the intake of protein. Ann Nutr Metabolism. 2019;74(3):242–250. doi:10.1159/000499374
162. Salas-Groves E, Alcorn M, Childress A, Galyean S. The effect of web-based culinary medicine to enhance protein intake on muscle quality in older adults: randomized controlled trial. JMIR Format Res. 2024;8:e49322.
163. Coelho-Júnior HJ, Milano-Teixeira L, Rodrigues B, Bacurau R, Marzetti E, Uchida M. Relative protein intake and physical function in older adults: a systematic review and meta-analysis of observational studies. Nutrients. 2018;10(9):1330. doi:10.3390/nu10091330
164. Ispoglou T, Witard OC, Duckworth LC, Lees MJ. The efficacy of essential amino acid supplementation for augmenting dietary protein intake in older adults: implications for skeletal muscle mass, strength and function. Proc Nutr Soc. 2021;80(2):230–242. doi:10.1017/S0029665120008010
165. Thalacker-Mercer A, Riddle E, Barre L. Protein and amino acids for skeletal muscle health in aging. Adv Food Nutr Res. 2020;91:29–64.
166. McKendry J, Currier BS, Lim C, McLeod JC, Thomas ACQ, Phillips SM. Nutritional supplements to support resistance exercise in countering the sarcopenia of aging. Nutrients. 2020;12(7):2057. doi:10.3390/nu12072057
167. Kugimiya Y, Iwasaki M, Ohara Y, et al. Relationship between oral hypofunction and sarcopenia in community-dwelling older adults: the otassha study. Int J Environ Res Public Health. 2021;18(12):6666. doi:10.3390/ijerph18126666
168. Kugimiya Y, Iwasaki M, Ohara Y, et al. Association between sarcopenia and oral functions in community-dwelling older adults: a cross-sectional study. J Cachexia Sarcopenia Muscle. 2023;14(1):429–438. doi:10.1002/jcsm.13145
169. Kumar P, Varshney S, Darsana G. Relation between components of oral health and sarcopenia in older adults: a narrative review. Muscles Ligaments Tendons J. 2022;12(4):570–593. doi:10.32098/mltj.04.2022.14
170. Celis A, Cáceres B, Escobar B, Barahona P, Dreyer E, Petermann-Rocha F. Impact of oral health interventions on sarcopenia and frailty in older adults: a systematic review. J Clin Med. 2025;14(6):1991. doi:10.3390/jcm14061991
171. Rebelos E, Tentolouris N, Jude E. The role of vitamin D in health and disease: a narrative review on the mechanisms linking vitamin D with disease and the effects of supplementation. Drugs. 2023;83(8):665–685. doi:10.1007/s40265-023-01875-8
172. Gil Á, Plaza-Diaz J, Mesa MD. Vitamin D. classic and novel actions. Ann Nutr Metab. 2018;72:87–95. doi:10.1159/000486536
173. Chang MC, Choo YJ. Effects of whey protein, leucine, and vitamin D supplementation in patients with sarcopenia: a systematic review and meta-analysis. Nutrients. 2023;15(3):521. doi:10.3390/nu15030521
174. Aoki K, Sakuma M, Endo N. The impact of exercise and vitamin D supplementation on physical function in community-dwelling elderly individuals: a randomized trial. J Orthop Sci. 2018;23(4):682–687. doi:10.1016/j.jos.2018.03.011
175. Jabbour J, Rahme M, Mahfoud ZR, El-Hajj Fuleihan G. Effect of high dose vitamin D supplementation on indices of sarcopenia and obesity assessed by DXA among older adults: a randomized controlled trial. Endocrine. 2022;76(1):162–171. doi:10.1007/s12020-021-02951-3
176. Prokopidis K, Giannos P, Katsikas Triantafyllidis K, et al. Effect of vitamin D monotherapy on indices of sarcopenia in community-dwelling older adults: a systematic review and meta-analysis. J Cachexia Sarcopenia Muscle. 2022;13(3):1642–1652. doi:10.1002/jcsm.12976
177. Rosendahl-Riise H, Spielau U, Ranhoff AH, Gudbrandsen OA, Dierkes J. Vitamin D supplementation and its influence on muscle strength and mobility in community-dwelling older persons: a systematic review and meta-analysis. J Hum Nutr Diet. 2017;30(1):3–15. doi:10.1111/jhn.12394
178. McCartney CR, McDonnell ME, Corrigan MD, Lash RW. Vitamin D insufficiency and epistemic humility: an endocrine society guideline communication. J Clin Endocrinol Metab. 2024;109(8):1948–1954. doi:10.1210/clinem/dgae322
179. Rolland Y, Dray C, Vellas B, Barreto PS. Current and investigational medications for the treatment of sarcopenia. Metabolism. 2023;149:155597. doi:10.1016/j.metabol.2023.155597
180. Feike Y, Zhijie L, Wei C. Advances in research on pharmacotherapy of sarcopenia. Aging Med. 2021;4(3):221–233. doi:10.1002/agm2.12168
181. Dias JP, Veldhuis JD, Carlson O, et al. Effects of transdermal testosterone gel or an aromatase inhibitor on serum concentration and pulsatility of growth hormone in older men with age-related low testosterone. Metabolism. 2017;69:143–147. doi:10.1016/j.metabol.2017.01.025
182. Holman ME, Gorgey AS. Testosterone and resistance training improve muscle quality in spinal cord injury. Med Sci Sports Exercise. 2019;51(8):1591–1598. doi:10.1249/MSS.0000000000001975
183. Wright TJ, Dillon EL, Durham WJ, et al. A randomized trial of adjunct testosterone for cancer‐related muscle loss in men and women. J Cachexia Sarcopenia Muscle. 2018;9(3):482–496. doi:10.1002/jcsm.12295
184. Dias JP, Melvin D, Shardell M, et al. Effects of transdermal testosterone gel or an aromatase inhibitor on prostate volume in older men. J Clin Endocrinol Metab. 2016;101(4):1865–1871. doi:10.1210/jc.2016-1111
185. Nam Y-S, Lee G, Yun JM, Cho B. Testosterone replacement, muscle strength, and physical function. World J Mens Health. 2018;36(2):110–122. doi:10.5534/wjmh.182001
186. Batsis JA, Villareal DT. Sarcopenic obesity in older adults: aetiology, epidemiology and treatment strategies. Nat Rev Endocrinol. 2018;14(9):513–537. doi:10.1038/s41574-018-0062-9
187. Fonseca GWPD, Dworatzek E, Ebner N, Von Haehling S. Selective androgen receptor modulators (SARMs) as pharmacological treatment for muscle wasting in ongoing clinical trials. Expert Opin Invest Drugs. 2020;29(8):881–891. doi:10.1080/13543784.2020.1777275
188. Muta Y, Tanaka T, Hamaguchi Y, Hamanoue N, Yanase T. Selective androgen receptor modulator, S42 has anabolic and anti-catabolic effects on cultured myotubes. Biochem Biophys Rep. 2019;17:177–181. doi:10.1016/j.bbrep.2019.01.006
189. Caputo M, Pigni S, Agosti E, et al. Regulation of GH and GH Signaling by Nutrients. Cells. 2021;10(6):1376.
190. Brioche T, Kireev RA, Cuesta S, et al. Growth hormone replacement therapy prevents sarcopenia by a dual mechanism: improvement of protein balance and of antioxidant defenses. J Gerontol Series A. 2014;69(10):1186–1198. doi:10.1093/gerona/glt187
191. Bian A, Ma Y, Zhou X, et al. Association between sarcopenia and levels of growth hormone and insulin-like growth factor-1 in the elderly. BMC Musculoskelet Disord. 2020;21(2020):1–9. doi:10.1186/s12891-020-03236-y
192. Bollen SE, Bass JJ, Fujita S, Wilkinson D, Hewison M, Atherton PJ. The Vitamin D/Vitamin D receptor (VDR) axis in muscle atrophy and sarcopenia. Cell Signal. 2022;96:110355. doi:10.1016/j.cellsig.2022.110355
193. Cheng SH, Chen KH, Chen C, Chu WC, Kang YN. The optimal strategy of vitamin D for sarcopenia: a network meta-analysis of randomized controlled trials. Nutrients. 2021;13(10):3589. doi:10.3390/nu13103589
194. Sakuma K, Hamada K, Yamaguchi A, Aoi W. Current nutritional and pharmacological approaches for attenuating sarcopenia. Cells. 2023;12(19):2422. doi:10.3390/cells12192422
195. Dent E, Morley J, Cruz-Jentoft A, et al. International clinical practice guidelines for sarcopenia (ICFSR): screening, diagnosis and management. j nutr health aging. 2018;22(10):1148–1161. doi:10.1007/s12603-018-1139-9
196. Han H, Zhou X, Mitch WE, Goldberg AL. Myostatin/activin pathway antagonism: molecular basis and therapeutic potential. Int J Biochem Cell Biol. 2013;45(10):2333–2347. doi:10.1016/j.biocel.2013.05.019
197. Dong J, Mitch W, Mitch W, Mitch W, Zhang L, Zhang L. Inhibition of myostatin in mice improves insulin sensitivity via irisin-mediated cross talk between muscle and adipose tissues. Int J Obesity. 2016;40(3):434–442. doi:10.1038/ijo.2015.200
198. Ahmad SS, Ahmad K, Lee EJ, Shaikh S, Choi I. Computational identification of dithymoquinone as a potential inhibitor of myostatin and regulator of muscle mass. Molecules. 2021;26(17):5407. doi:10.3390/molecules26175407
199. Becker C, Lord SR, Studenski SA, et al. Myostatin antibody (LY2495655) in older weak fallers: a proof-of-concept, randomised, Phase 2 trial. Lancet Diabetes Endocrinol. 2015;3(12):948–957. doi:10.1016/S2213-8587(15)00298-3
200. Statland JM, Campbell C, Desai U, et al. Randomized phase 2 study of ACE-083, a muscle-promoting agent, in facioscapulohumeral muscular dystrophy. Muscle Nerve. 2022;66(1):50–62. doi:10.1002/mus.27558
201. Sivakumar K, Cochrane TI, Sloth B, et al. Long-term safety and tolerability of bimagrumab (BYM338) in sporadic inclusion body myositis. Neurology. 2020;95(14):e1971–e1978. doi:10.1212/WNL.0000000000010417
202. Chen L, Zhang H, Chi M, Yang Q, Guo C. Drugs for the treatment of muscle atrophy. Background Manag Muscul Atrophy. 2021;(93503):83–101. doi:10.5772/intechopen.93503
203. Lautherbach N, Gonçalves DAP, Silveira WA, et al. Urocortin 2 promotes hypertrophy and enhances skeletal muscle function through cAMP and insulin/IGF-1 signaling pathways. Mol Metabol. 2022;60:101492. doi:10.1016/j.molmet.2022.101492
204. Higashihara T, Nishi H, Takemura K, et al. β2-adrenergic receptor agonist counteracts skeletal muscle atrophy and oxidative stress in uremic mice. Sci Rep. 2021;11(1):9130. doi:10.1038/s41598-021-88438-7
205. Kim J, Grotegut CA, Wisler JW, et al. The β-arrestin-biased β-adrenergic receptor blocker carvedilol enhances skeletal muscle contractility. Proc Natl Acad Sci. 2020;117(22):12435–12443. doi:10.1073/pnas.1920310117
206. Aguirre F, Abrigo J, Gonzalez F, Gonzalez A, Simon F, Cabello-Verrugio C. Protective effect of angiotensin 1–7 on sarcopenia induced by chronic liver disease in mice. Int J Mol Sci. 2020;21(11):3891. doi:10.3390/ijms21113891
207. Masiero E, Agatea L, Mammucari C, et al. Autophagy is required to maintain muscle mass. Cell Metab. 2009;10(6):507–515. doi:10.1016/j.cmet.2009.10.008
208. Schellino R, Boido M, Vrijbloed JW, Fariello RG, Vercelli A. Synergistically acting on myostatin and agrin pathways increases neuromuscular junction stability and endurance in old mice. Aging Dis. 2024;15(2):893–910. doi:10.14336/AD.2023.0713-1
209. Bano G, Trevisan C, Carraro S, et al. Inflammation and sarcopenia: a systematic review and meta-analysis. Maturitas. 2017;96:10–15. doi:10.1016/j.maturitas.2016.11.006
210. Murphy C, Withrow J, Hunter M, et al. Emerging role of extracellular vesicles in musculoskeletal diseases. Mol Aspect Med. 2018;60:123–128. doi:10.1016/j.mam.2017.09.006
211. Yue B, Yang H, Wang J, et al. Exosome biogenesis, secretion and function of exosomal miRNAs in skeletal muscle myogenesis. Cell Proliferation. 2020;53(7):e12857. doi:10.1111/cpr.12857
212. Kerr HL, Krumm K, Anderson B, et al. Mouse sarcopenia model reveals sex- and age-specific differences in phenotypic and molecular characteristics. J Clin Invest. 2024;134(16):e172890. doi:10.1172/JCI172890
213. Ma H, Jing Y, Zeng J, et al. Human umbilical cord mesenchymal stem cell-derived exosomes ameliorate muscle atrophy via the miR-132-3p/FoxO3 axis. J Orthop Translat. 2024;49:23–36. doi:10.1016/j.jot.2024.08.005
214. Byun SE, Sim C, Chung Y, et al. Skeletal muscle regeneration by the exosomes of adipose tissue-derived mesenchymal stem cells. Curr Issues Mol Biol. 2021;43(3):1473–1488. doi:10.3390/cimb43030104
215. Wan R, Liu S, Feng X, et al. The revolution of exosomes: from biological functions to therapeutic applications in skeletal muscle diseases. J Orthop Translat. 2024;45:132–139. doi:10.1016/j.jot.2024.01.001
216. Wohlwend M, Laurila -P-P, Williams K, et al. The exercise-induced long noncoding RNA CYTOR promotes fast-twitch myogenesis in aging. Sci, trans med. 2021;13(623):eabc7367. doi:10.1126/scitranslmed.abc7367
217. Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past, present, and future. Stem Cell Res Ther. 2019;10(1):1–22. doi:10.1186/s13287-019-1165-5
218. Hassanzadeh A, Rahman HS, Markov A, et al. Mesenchymal stem/stromal cell-derived exosomes in regenerative medicine and cancer; overview of development, challenges, and opportunities. Stem Cell Res Ther. 2021;12(1):297. doi:10.1186/s13287-021-02378-7
219. Jiang X, Yang J, Lin Y, et al. Extracellular vesicles derived from human ESC-MSCs target macrophage and promote anti-inflammation process, angiogenesis, and functional recovery in ACS-induced severe skeletal muscle injury. Stem Cell Res Ther. 2023;14(1):331. doi:10.1186/s13287-023-03530-1
220. Marzetti E, Lozanoska-Ochser B, Calvani R, Landi F, Coelho-Júnior HJ, Picca A. Restoring mitochondrial function and muscle satellite cell signaling: remedies against age-related sarcopenia. Biomolecules. 2024;14(4):415. doi:10.3390/biom14040415
221. Wang C, Zhao B, Zhai J, et al. Clinical-grade human umbilical cord-derived mesenchymal stem cells improved skeletal muscle dysfunction in age-associated sarcopenia mice. Cell Death Dis. 2023;14(5):321. doi:10.1038/s41419-023-05843-8
222. Tu H, Li YL. Inflammation balance in skeletal muscle damage and repair. Front Immunol. 2023;14:1133355. doi:10.3389/fimmu.2023.1133355
223. Cai L, Shi L, Peng Z, Sun Y, Chen J. Ageing of skeletal muscle extracellular matrix and mitochondria: finding a potential link. Annals of Medicine. 2023;55(2):2240707. doi:10.1080/07853890.2023.2240707
224. Barakat N, Jangir H, Gallo L, Grillo M, Guo X, Hickman J. Inhibition of metalloproteinases extends longevity and function of in vitro human iPSC-derived skeletal muscle. Biomedicines. 2024;12(4):856. doi:10.3390/biomedicines12040856
225. Lee SJ, Bhasin S, Klickstein L, Krishnan V, Rooks D. Challenges and future prospects of targeting myostatin/activin a signaling to treat diseases of muscle loss and metabolic dysfunction. J Gerontol a Biol Sci Med Sci. 2023;78(Suppl 1):32–37. doi:10.1093/gerona/glad033
226. Caron L, Testa S, Magdinier F. Induced pluripotent stem cells for modeling physiological and pathological striated muscle complexity. J Neuromuscul Disord. 2023;10(5):761–776. doi:10.3233/JND-230076
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.