Next Article in Journal
Long Non-Coding RNAs Contribute to Glucose Starvation-Induced Dedifferentiation in Lung Adenocarcinoma
Previous Article in Journal
The Identification of a Sub-Micromolar Peptide-Based Protein Arginine Methyltransferase 1 (PRMT1) Inhibitor from a Plate-Based Screening Assay
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cisplatin-Induced Skeletal Muscle Atrophy: Biomolecular Mechanisms and the Protective Role of Exercise-Induced Myokines

1
College of Physical Education, Guangdong University of Education, Guangzhou 510800, China
2
College of Sports and Health, Guangzhou Sport University, Guangzhou 510500, China
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(11), 1495; https://doi.org/10.3390/biom15111495
Submission received: 21 September 2025 / Revised: 19 October 2025 / Accepted: 22 October 2025 / Published: 23 October 2025
(This article belongs to the Section Molecular Biology)

Abstract

Cisplatin is a widely used chemotherapy drug for the treatment of various cancers; however, its clinical use is often accompanied by skeletal muscle atrophy, which not only impacts patients’ physical health but also significantly diminishes their quality of life. The mechanisms underlying cisplatin-induced muscle atrophy are complex and involve a series of molecular biological processes, including oxidative stress, inflammation, protein degradation, and muscle cell apoptosis. Recent studies have suggested that exercise intervention can significantly alleviate cisplatin-induced muscle damage by modulating exercise-induced myokines. Myokines, such as muscle-derived cytokines (e.g., IL-6, irisin) and other related factors, can mitigate muscle atrophy through anti-inflammatory, antioxidative, and muscle-synthesis-promoting mechanisms. This review explores the molecular mechanisms of cisplatin-induced skeletal muscle atrophy, examines the potential protective effects of exercise intervention, and highlights the role of exercise-induced myokines in this process. The findings suggest that exercise not only alleviates chemotherapy-induced muscle atrophy by improving metabolic and immune status but also activates myokines to promote muscle regeneration and repair, offering a promising adjunctive therapy for cisplatin-treated patients.

1. Introduction

Skeletal muscle, which accounts for nearly 40% of total body mass, is fundamental to locomotion, metabolic regulation, and systemic homeostasis [1,2]. The maintenance of skeletal muscle mass and function is therefore critical for health and survival [3,4]. Muscle atrophy, defined as the loss of muscle mass and strength, is a debilitating condition that occurs in diverse clinical contexts, including aging [5], immobilization [6], chronic diseases [7,8], and cancer therapy [9]. In oncology, muscle wasting has attracted particular concern because it worsens fatigue, reduces treatment tolerance, compromises immune competence, and is associated with poor prognosis [10,11,12].
Chemotherapy is a major contributor to secondary muscle atrophy [13,14,15]. Cisplatin, a platinum-based chemotherapeutic drug widely used against solid tumors [16], is especially notorious for inducing skeletal muscle loss [17,18]. Both preclinical and clinical studies demonstrate that cisplatin treatment rapidly accelerates muscle wasting [19,20,21,22,23]. Mechanistically, this process involves the overactivation of proteolytic systems such as the ubiquitin–proteasome [24] and autophagy–lysosome pathways [25], inhibition of protein synthesis due to dysregulated the protein kinase B (Akt)/mechanistic target of rapamycin (mTOR) signaling pathway [26,27], mitochondrial dysfunction with excessive reactive oxygen species (ROS) production [16,26], and chronic inflammatory activation [18]. Together, these alterations disrupt protein homeostasis and cellular metabolism, leading to progressive muscle degradation.
In this review, it is important to differentiate among cachexia, sarcopenia, and atrophy. Cachexia is a multifactorial metabolic syndrome associated with chronic diseases such as cancer, characterized by severe weight and muscle loss driven by systemic inflammation and metabolic imbalance [10]. Sarcopenia refers to the age-related, progressive decline in skeletal muscle mass and function, primarily due to reduced physical activity, hormonal alterations, and impaired protein synthesis [28]. In contrast, atrophy denotes localized muscle loss caused by disuse, denervation, or direct cytotoxic injury, such as that induced by chemotherapy [6,26]. Understanding these distinctions provides a conceptual framework for interpreting chemotherapy-induced muscle wasting within its unique pathological context.
Given the lack of effective pharmacological therapies [13,29], exercise has emerged as a promising non-pharmacological intervention to counteract chemotherapy-induced muscle wasting [30,31,32,33,34,35]. Different modes of exercise act through distinct biomolecular pathways. Endurance exercise enhances mitochondrial quality control and reduces systemic inflammation [36,37,38,39], whereas resistance training promotes muscle hypertrophy by stimulating protein synthesis and inhibiting proteolysis [40,41,42]. Importantly, exercise also induces the secretion of myokines—muscle-derived cytokines and peptides—that mediate local and systemic protective effects [43,44]. For example, irisin exerts anti-inflammatory and antioxidative actions that alleviate ROS-mediated damage [45,46,47]; interleukin-15 (IL-15) stimulates protein synthesis and suppresses catabolic signaling [48,49]; and brain-derived neurotrophic factor (BDNF) supports mitochondrial biogenesis and neuromuscular junction integrity [50,51]. Through these mechanisms, exercise-induced myokines provide a molecular link between physical activity and the preservation of muscle mass under chemotherapeutic stress.
The purpose of this review is to summarize the molecular mechanisms of chemotherapy-induced skeletal muscle atrophy, with a particular focus on cisplatin-related pathways, and to discuss how exercise counteracts these detrimental changes. We highlight the roles of different exercise modalities and their associated myokine responses, and we provide perspectives on future research and translational implications. Understanding these biomolecular interactions may guide the development of integrative therapeutic strategies aimed at preserving skeletal muscle health in patients undergoing chemotherapy.

2. Methods

This review aims to provide a comprehensive synthesis of recent advances in understanding cisplatin-induced skeletal muscle atrophy and the protective effects of exercise-based interventions. A systematic literature search was conducted to identify relevant studies published from January 2020 to September 2025 across PubMed, Web of Science, Scopus, and Google Scholar.
The search strategy combined controlled vocabulary (e.g., MeSH terms) and free-text keywords to identify relevant studies on chemotherapy-induced muscle wasting, cisplatin toxicity, and exercise interventions. The search focused on studies exploring the molecular mechanisms underlying cisplatin-induced skeletal muscle atrophy and the protective effects of exercise, particularly how exercise-induced myokines and related pathways contribute to muscle preservation. Key terms included “Cisplatin,” “Chemotherapy-induced muscle atrophy,” “Cachexia,” “Skeletal muscle wasting,” “Exercise,” “Endurance training,” “Resistance training,” “Combined exercise,” “Myokines,” “IL-6,” “Irisin,” “Mitochondrial dysfunction,” “Oxidative stress,” “Proteostasis,” “Inflammation,” “Muscle regeneration,” “Muscle synthesis,” “Apoptosis,” “Muscle injury repair,” “Cisplatin toxicity mechanism,” “Aerobic exercise,” and “Anaerobic exercise.” To provide a more complete molecular perspective on muscle atrophy and adaptation, additional signaling-related keywords such as Akt, mTOR, MuRF-1, LC3B, and PGC1α were also considered. Boolean operators (AND, OR) and truncation techniques were applied to optimize sensitivity and specificity.
Studies were included if they met the following criteria: (1) peer-reviewed publications in English; (2) human or animal studies, including randomized controlled trials, observational studies, cohort studies, and preclinical experimental studies; (3) primary focus on cisplatin-induced muscle atrophy, chemotherapy-related cachexia, or exercise-based protective mechanisms; and (4) relevance to molecular pathways such as protein turnover, mitochondrial biology, oxidative stress, or inflammation, as well as exercise modalities including endurance, resistance, or combined training. Exclusion criteria comprised non-English publications, preprints or grey literature, in vitro studies without in vivo validation, and articles lacking clear relevance to the review objectives.
Although the review was limited to English-language publications, it is acknowledged that relevant studies may also exist in other languages such as Chinese, Russian, French, or German. Many non-English publications include English abstracts, so this language restriction may have excluded some valuable evidence and should be considered a potential limitation of the study.
A total of 246 articles were identified and screened based on relevance and inclusion criteria. The screening process involved an initial review of titles and abstracts, followed by full-text screening to confirm eligibility. A PRISMA-like flow diagram outlining the study selection process is provided below to increase transparency (Figure 1).

3. Chemotherapy-Induced Skeletal Muscle Atrophy

3.1. Clinical Relevance of Chemotherapy-Associated Muscle Wasting

Cancer patients frequently experience muscle wasting during the course of treatment, which severely affects their functional capacity, quality of life, and overall survival [9,52]. Chemotherapy has been identified as one of the main contributors to secondary muscle atrophy, independent of tumor burden itself [53,54,55]. The phenomenon is often described as chemotherapy-induced cachexia or sarcopenia, characterized by decreased muscle mass, weakness, and metabolic dysfunction [56,57]. Clinical observations demonstrate that patients undergoing platinum-based chemotherapy, including cisplatin, exhibit accelerated loss of lean body mass compared with non-treated cancer patients [58,59].
This muscle wasting is clinically significant for several reasons. First, reduced muscle mass compromises treatment tolerance, leading to increased dose-limiting toxicities, treatment delays, or discontinuation [56,60,61]. Second, skeletal muscle depletion negatively impacts the pharmacokinetics of chemotherapeutic drugs, potentially altering efficacy and increasing systemic toxicity [62,63]. Third, muscle wasting worsens cancer-related fatigue, a major determinant of patient well-being [64,65,66]. Finally, low muscle mass is consistently associated with poor overall survival and disease-free survival in multiple cancer types [67,68,69,70,71,72]. These findings underscore the urgent need for strategies to mitigate chemotherapy-induced skeletal muscle atrophy in clinical settings.

3.2. Cisplatin and Skeletal Muscle Atrophy: Clinical and Preclinical Evidence

Among various chemotherapeutic agents, cisplatin has been extensively studied for its muscle-wasting effects [16,18]. Clinically, cisplatin-based regimens are associated with rapid loss of lean mass, even within the first few cycles of treatment [56,73]. Cross-sectional and longitudinal studies show that cancer patients treated with cisplatin present with reduced muscle cross-sectional area and decreased grip strength, both of which predict poor clinical outcomes [57,74].
Preclinical studies using rodent models have provided valuable insights into the phenotypic changes associated with cisplatin-induced muscle wasting [21,75,76]. Both Liu et al. and Seo et al. found that cisplatin administration leads to significant reductions in body weight and muscle mass, particularly in fast-twitch muscles such as the gastrocnemius and tibialis anterior [18,77]. Histological analysis reveals muscle fiber atrophy and an increase in the expression of atrophy-related genes, including Atrogin-1 (also known as FBXO32) and muscle RING finger-1 (MuRF1), both of which play key roles in muscle degradation [18,56,73]. Additionally, cisplatin induces mitochondrial dysfunction and oxidative stress [78]. In mice, cisplatin impairs mitochondrial respiratory capacity, decreases adenosine triphosphate (ATP) production, and elevates ROS levels, which contribute to oxidative damage in muscle fibers [18,77,79].
In addition to these well-established catabolic pathways, recent studies have highlighted the pivotal role of glycogen synthase kinase 3 beta (GSK3β) in cisplatin-induced muscle atrophy [16]. GSK3β, which can be activated by cisplatin treatment, contributes to skeletal muscle loss by suppressing ribosomal RNA synthesis, impairing mitochondrial function, and promoting muscle fiber degradation [16,77,80,81]. Moreover, GSK3β acts as a key regulator in the Wingless/Integrated–β-catenin signaling cascade, where its overactivation accelerates β-catenin degradation, thereby inhibiting myogenic differentiation and protein synthesis [82,83]. Exercise interventions and GSK3β-targeted therapies have been reported to attenuate GSK3β activity and restore β-catenin signaling, improving muscle regeneration and function [84,85].
Although the molecular mechanisms underlying muscle atrophy share several common pathways across different etiologies, cisplatin-induced skeletal muscle wasting exhibits unique features that distinguish it from other forms such as disuse, denervation, or cancer cachexia [86]. Specifically, cisplatin directly induces mitochondrial DNA damage and oxidative stress through cisplatin–DNA adduct formation [17], which rarely occurs in other atrophy models [18]. In addition, chemotherapy-related systemic inflammation (elevated IL-6 and TNF-α) [77,87] and suppressed anabolic myokine secretion (e.g., IL-15, irisin) [88] collectively exacerbate protein degradation [20]. Moreover, cisplatin markedly inhibits proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) expression and mitochondrial biogenesis [16], leading to energy metabolism failure and impaired muscle regeneration [75]. These features reflect a dual mechanism involving both direct cytotoxic injury to myofibers and systemic metabolic dysregulation, highlighting that cisplatin-induced atrophy represents a distinct biological entity, with its major comparative characteristics summarized in Table 1 [89].
Taken together, clinical and preclinical evidence strongly supports the role of cisplatin in promoting skeletal muscle atrophy through multifactorial biomolecular mechanisms [20,92,93,94] (Table 2). Understanding these mechanisms provides a foundation for exploring targeted interventions, such as exercise, to counteract cisplatin-induced muscle wasting [16,95].

3.3. Chemotherapy-Induced Decreased Motility and Secondary Muscle Disturbance

Beyond the direct cytotoxic effects of cisplatin on skeletal muscle, chemotherapy frequently reduces patients’ physical activity due to fatigue, nausea, and overall malaise [64]. This treatment-induced decline in mobility promotes a secondary form of disuse atrophy, which synergistically exacerbates cisplatin-induced muscle loss [96]. Reduced mechanical loading further impairs mitochondrial turnover and oxidative capacity [97], suppresses muscle protein synthesis [98], and activates proteolytic systems such as the ubiquitin–proteasome and autophagy–lysosome pathways [41]. In addition, inactivity-driven metabolic inflexibility amplifies the pre-existing energy deficit caused by cisplatin-mediated mitochondrial dysfunction [79], resulting in accelerated muscle catabolism [20]. Collectively, the combined effects of chemotherapy-related inactivity and cisplatin toxicity create a feed-forward cycle of muscular and metabolic deterioration [35]. These insights emphasize the importance of incorporating early-stage physical rehabilitation or mild exercise interventions to mitigate both primary and secondary muscle atrophy during chemotherapy [99].

4. Molecular Mechanisms Underlying Muscle Atrophy

Skeletal muscle mass is determined by the dynamic balance between protein synthesis and degradation [100,101]. Chemotherapy-induced muscle wasting, particularly that caused by cisplatin, results from a disruption of this balance through multiple converging molecular pathways [17]. The major mechanisms include activation of proteolytic systems, suppression of anabolic signaling, mitochondrial dysfunction, oxidative stress, and inflammatory dysregulation [98,102]. Figure 2 illustrates these key molecular mechanisms and how they interact to contribute to cisplatin-induced muscle atrophy.

4.1. Protein Degradation Pathways

One of the hallmarks of cisplatin-induced muscle atrophy is the activation of protein degradation systems [103,104]. The ubiquitin–proteasome system (UPS) plays a central role in the breakdown of myofibrillar proteins [16]. Two muscle-specific E3 ubiquitin ligases, Atrogin-1 and MuRF1, are consistently upregulated in cisplatin-treated muscle [105,106]. Their activation enhances ubiquitination and subsequent proteasomal degradation of structural proteins such as myosin heavy chain, leading to muscle fiber shrinkage [107,108].
In parallel, the autophagy–lysosome system is also stimulated [109]. Cisplatin increases autophagy-related proteins, including microtubule-associated protein 1A/1B-light chain 3-II (LC3-II) and Beclin-1, promoting excessive autophagic flux [77,110]. While basal autophagy is essential for cellular homeostasis, sustained overactivation contributes to muscle loss by degrading organelles and cytoplasmic components [111,112]. Together, these pathways shift protein turnover toward catabolism, thereby driving muscle wasting.

4.2. Suppression of Protein Synthesis

In addition to enhanced degradation, cisplatin suppresses protein synthesis by inhibiting the Akt/mTOR signaling pathway [17,29,105,113,114]. Under normal conditions, Akt activation stimulates mammalian target of rapamycin complex 1 (mTORC1), which in turn promotes ribosomal biogenesis and translation initiation [115,116]. However, cisplatin reduces Akt phosphorylation and mTOR activity, leading to decreased expression of downstream effectors such as p70S6 kinase and eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) [17,117]. This impairment of anabolic signaling reduces the ability of muscle fibers to maintain protein mass.
Furthermore, cisplatin induces endoplasmic reticulum (ER) stress, which activates eukaryotic translation initiation factor 2 subunit alpha (eIF2α) phosphorylation and suppresses global protein translation [118,119,120]. The combined effects of reduced mTOR signaling and ER stress-mediated translational arrest exacerbate the imbalance between synthesis and degradation [121].

4.3. Mitochondrial Dysfunction and Oxidative Stress

Mitochondrial impairment is another critical factor in cisplatin-induced muscle atrophy [76,86]. Cisplatin damages mitochondrial deoxyribonucleic acid (DNA), disrupts the electron transport chain, and decreases ATP production [77,122,123]. This energy deficit compromises muscle contractility and cellular metabolism [20]. At the same time, cisplatin increases the generation of ROS, which causes oxidative damage to lipids, proteins, and DNA [17,124,125].
Oxidative stress also activates catabolic signaling pathways [126]. For instance, ROS can stimulate the transcription factor forkhead box O (FoxO), which upregulates Atrogin-1 and MuRF1 [41,127]. In addition, mitochondrial dysfunction triggers mitophagy, contributing to excessive organelle degradation [97,128]. The vicious cycle of mitochondrial damage and ROS overproduction thus exacerbates muscle wasting [79].

4.4. Inflammatory Signaling

Cisplatin administration is associated with systemic and local inflammation [129]. Elevated levels of pro-inflammatory cytokines such as TNF-α, IL-6, and interferon-gamma (IFN-γ) are commonly observed [10,18,20,74]. These cytokines activate intracellular pathways including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) [130] and the janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) signaling pathway [131,132], which promote proteolysis and inhibit protein synthesis [41]. For example, NF-κB activation enhances the expression of Atrogin-1 and MuRF1 [1], while STAT3 activation contributes to mitochondrial dysfunction and metabolic reprogramming [133].
Inflammation also disrupts the function of regulatory immune cells such as Tregs, reducing their ability to maintain an anti-inflammatory environment [10,134]. The resulting imbalance between pro- and anti-inflammatory signaling accelerates muscle loss and contributes to systemic cachexia [133,135].

4.5. Crosstalk Between These Pathways

These molecular mechanisms do not operate in isolation but rather form a complex regulatory network [136,137]. For example, oxidative stress can activate NF-κB signaling, linking mitochondrial dysfunction to inflammation [138,139,140]. Similarly, inflammatory cytokines suppress Akt/mTOR signaling, thereby coupling inflammation with reduced protein synthesis [41,141]. ER stress also interacts with ROS generation and autophagic responses, further amplifying catabolic processes [142,143].
This extensive crosstalk suggests that chemotherapy-induced skeletal muscle atrophy is a multifactorial process, in which proteostasis imbalance, mitochondrial dysfunction, oxidative stress, and inflammation reinforce each other [35,97]. Targeting multiple nodes of this network, such as through exercise interventions, may therefore offer more effective protection against muscle wasting than single-pathway approaches [13,144].

5. Exercise as a Protective Intervention

Exercise has emerged as a promising non-pharmacological approach to counteract chemotherapy-induced skeletal muscle atrophy [13,145]. Unlike pharmacological interventions that typically target single molecules, exercise exerts multi-systemic effects that simultaneously influence protein turnover, mitochondrial function, oxidative stress, and inflammatory signaling [146,147]. Different exercise modalities, including endurance and resistance training, as well as their combination, confer distinct but complementary molecular benefits [148,149,150,151]. In addition, exercise induces the secretion of myokines, which mediate local and systemic protective effects [43,152,153].

5.1. Endurance Exercise and Mitochondrial Adaptations

Endurance exercise, such as treadmill running or swimming, enhances oxidative metabolism and mitochondrial quality control [154,155,156,157,158]. Preclinical studies show that endurance training improves mitochondrial biogenesis through activation of the PGC-1α pathway, thereby restoring ATP production and reducing cisplatin-induced mitochondrial dysfunction [19,105,159]. Enhanced mitochondrial turnover also decreases ROS accumulation, alleviating oxidative damage to muscle fibers [102,160,161].
Endurance exercise further exerts systemic anti-inflammatory effects [162,163]. It downregulates pro-inflammatory cytokines such as TNF-α and IL-6, while promoting anti-inflammatory mediators including IL-10 [164,165]. These changes help to restore immune balance and attenuate the NF-κB and JAK/STAT3 pathways that drive catabolism [165,166]. Thus, endurance exercise addresses two major mechanisms of cisplatin-induced muscle wasting: mitochondrial dysfunction and chronic inflammation [77,105] (Figure 3).

5.2. Resistance Training and Anabolic Signaling

Resistance exercise is the most effective stimulus for promoting muscle hypertrophy and maintaining lean mass [167,168,169,170]. Mechanistically, resistance training activates the Akt/mTOR pathway, leading to increased phosphorylation of mTORC1 downstream effectors such as p70 ribosomal S6 kinase (p70S6K) and 4E-BP1 [42,171]. This activation enhances ribosomal biogenesis and translation initiation, counteracting cisplatin-induced suppression of protein synthesis [172,173].
Resistance training also inhibits proteolysis [174,175]. It reduces the expression of Atrogin-1 and MuRF1, thereby attenuating ubiquitin–proteasome-mediated degradation [102,176,177]. Additionally, resistance exercise has been shown to suppress excessive autophagy by normalizing LC3-II and Beclin-1 levels [178,179]. Together, these adaptations shift the balance toward protein anabolism, providing structural protection against muscle atrophy (Figure 3).

5.3. Combined Exercise and Synergistic Effects

While endurance and resistance training exert distinct benefits, their combination may produce synergistic effects [180,181,182]. Clinical studies in cancer survivors demonstrate that combined training improves both aerobic capacity and muscle strength [183,184]. In preclinical models, concurrent exercise has been shown to simultaneously enhance mitochondrial function, reduce ROS, activate mTOR signaling, and suppress catabolic gene expression [185,186].
Importantly, combined training may provide superior functional outcomes by integrating systemic metabolic improvements with structural muscle preservation [183,187] (Figure 3). This suggests that multimodal exercise prescriptions could be particularly beneficial for patients undergoing cisplatin-based chemotherapy [188,189].

5.4. Myokines as Mediators of Exercise-Induced Protection

Exercise protects skeletal muscle through the secretion of myokines, muscle-derived cytokines and peptides with endocrine-like actions [43,190,191]. These myokines circulate in the blood and not only support muscle cells but also influence other organs such as the liver, fat tissue, and brain [192,193]. Classic myokines, like irisin, IL-15, and BDNF, have been shown to reduce oxidative stress, stimulate protein synthesis, and promote mitochondrial biogenesis, forming a solid foundation for exercise-induced muscle protection [49,51,194]. Figure 4 shows how these myokines, such as irisin, IL-15, and BDNF, help reduce oxidative stress, promote protein synthesis, and improve mitochondrial function, providing protection against muscle atrophy. Irisin, in particular, is released during endurance exercise, enhancing mitochondrial function and energy production, which helps buffer energy deficits and prevent muscle degradation [46]. It also reduces oxidative stress and inflammation, offering protection against muscle wasting conditions, such as those induced by chemotherapy [45]. IL-15 is known to stimulate satellite cell activity, promote protein synthesis, and prevent muscle atrophy, while also reducing oxidative stress and inflammation [48]. BDNF increases after exercise and supports mitochondrial activity, protein synthesis, and muscle regeneration, helping to maintain muscle integrity under stress [195]. These classic myokines play a crucial role in muscle adaptation to exercise and offer potential therapeutic benefits for conditions like chemotherapy-induced muscle wasting.
Beyond these traditional factors, recent studies have identified novel myokines with potential relevance to chemotherapy-induced muscle wasting [89,196] (Figure 4). Fibroblast growth factor 21 (FGF21), a stress-inducible myokine elevated by exercise, enhances mitochondrial function and metabolic flexibility, thereby buffering the energy deficits associated with cisplatin treatment [197,198]. Apelin, whose secretion increases after endurance exercise, improves angiogenesis, activates AMPK/PGC-1α signaling, and stimulates satellite cell activity, facilitating muscle regeneration under wasting conditions [199,200,201]. Decorin, induced by resistance exercise, antagonizes transforming growth factor-beta (TGF-β) signaling while promoting hypertrophy, thus offering both antifibrotic and anabolic protection against chemotherapy-related muscle degeneration [202,203,204]. Emerging candidates such as myonectin (CTRP15) and musclin (osteocrin) have been implicated in lipid metabolism, vascular regulation, and endurance enhancement, though their precise roles in chemotherapy-induced muscle loss remain to be clarified [205,206].
Together, these classical and novel myokines highlight the ability of exercise to orchestrate a multi-system defense network, integrating redox control, proteostasis, angiogenesis, and regenerative pathways [191,207]. This perspective not only deepens mechanistic insights but also points to myokines as potential biomarkers and therapeutic targets to mimic the systemic benefits of exercise [201,207,208,209].

5.5. Summary of Protective Mechanisms

In summary, exercise mitigates chemotherapy-induced skeletal muscle atrophy through multiple converging mechanisms [210,211,212]. Endurance exercise improves mitochondrial function and reduces inflammation [196,213]; resistance training restores anabolic signaling and inhibits proteolysis [31,146]; combined training integrates these benefits to optimize outcomes [31,196]; and myokines serve as systemic mediators that amplify local adaptations [89,146]. These multifaceted effects highlight the unique potential of exercise as an integrative intervention to preserve skeletal muscle health in cancer patients.

6. Future Directions and Challenges

Although numerous studies have investigated the effects of cisplatin and exercise interventions using animal models [214,215], these findings may not fully translate to human physiology. Animal experiments allow for precise control of dosage, environment, and genetic background, but they cannot fully replicate the complexity of human responses, including interindividual variability, comorbidities, and lifestyle factors [214,216]. For example, several protective mechanisms of exercise observed in rodents—such as enhanced mitochondrial biogenesis and antioxidative enzyme activity—may differ in magnitude or even direction in humans [154,217]. Therefore, outcomes derived exclusively from animal studies, such as histological recovery patterns or molecular biomarkers (e.g., PGC-1α, sirtuin 1, or oxidative stress markers), should be interpreted cautiously when inferring potential clinical benefits. Future research should emphasize translational approaches, including well-controlled clinical studies, to validate these findings and clarify species-specific responses.
Recognizing these translational limitations helps to better define the next steps for future research. Building upon these considerations, substantial progress has been made in understanding the mechanisms of cisplatin-induced muscle atrophy and the protective effects of exercise, several gaps remain to be addressed [218,219]. Future research should focus on the following directions (Table 3):
Although patient heterogeneity necessitates personalized exercise prescriptions, the lack of standardized exercise protocols reflects the current gap between individualized practice and evidence-based framework development [227,228]. Bridging this gap through flexible standardized models should be a key goal for future research.

6.1. Standardization of Preclinical Models

Current evidence on cisplatin-induced muscle wasting is largely derived from heterogeneous animal models, with variations in drug dose, treatment duration, and muscle groups analyzed [221,225]. This lack of standardization complicates cross-study comparisons and limits translation to clinical settings [229]. Establishing standardized protocols for cisplatin administration, coupled with reproducible exercise interventions, will be essential to improve the reliability of preclinical findings [230].

6.2. Mechanistic Integration of Exercise Modalities

Most studies focus on either endurance or resistance training, whereas few directly compare or combine modalities in the context of chemotherapy-induced muscle atrophy [35,231]. Moreover, the underlying molecular crosstalk between endurance- and resistance-induced pathways remains poorly defined [10]. Future investigations should explore how combined training orchestrates proteostasis, mitochondrial function, and inflammation in an integrated manner, potentially revealing synergistic mechanisms beyond additive effects.

6.3. Translational Biomarkers and Myokine Profiling

Although several myokines such as irisin, IL-15, and FGF21 have been implicated in exercise-mediated protection, their roles as circulating biomarkers in chemotherapy patients remain to be validated [43,232]. Large-scale studies integrating omics approaches (proteomics [233], metabolomics [234], single-cell transcriptomics [235]) are needed to identify biomarker panels that reliably reflect muscle health and therapeutic responsiveness. This would also accelerate the development of pharmacological mimetics that reproduce the systemic benefits of exercise [236,237].

6.4. Clinical Implementation and Personalized Exercise Prescriptions

Despite encouraging preclinical evidence [189], clinical trials testing exercise interventions during cisplatin-based chemotherapy remain scarce [189,220]. Challenges include patient heterogeneity, treatment side effects, and adherence to exercise programs [220]. Future clinical studies should prioritize the development of personalized exercise prescriptions, accounting for cancer type, treatment regimen, baseline physical status, and genetic background. Integrating digital health tools such as wearables and tele-rehabilitation may enhance feasibility and patient compliance [222,223,224].
Moreover, recent studies highlight that skeletal muscle is not a uniform tissue but comprises distinct molecular subtypes exhibiting differential vulnerability to catabolic stress [87,238]. For example, Bhatt et al. identified molecular subtypes of human skeletal muscle in cancer cachexia, characterized by unique metabolic and inflammatory signatures that influence susceptibility to atrophy [87]. This molecular heterogeneity may also exist in chemotherapy-induced muscle wasting, potentially explaining interindividual variability in treatment response and exercise efficacy [29,35]. Incorporating this concept into future translational research could guide the development of more precise and subtype-specific exercise prescriptions.

6.5. Dietary Modulation in Chemotherapy-Induced Skeletal Muscle Atrophy

Recent findings emphasize that nutritional modulation plays a critical role in counteracting cisplatin-induced skeletal muscle wasting [239]. High-quality protein and branched-chain amino acid (BCAA) supplementation can enhance muscle protein synthesis through activation of the mTOR pathway and suppression of proteolysis [240]. Omega-3 polyunsaturated fatty acids and antioxidant vitamins (C and E) mitigate oxidative stress and inflammation, helping to preserve mitochondrial integrity and function [241]. Additionally, micronutrients such as selenium and zinc strengthen antioxidant defense systems [242,243], while polyphenolic compounds like resveratrol stimulate PGC-1α–mediated mitochondrial biogenesis and muscle regeneration [244]. Recent preclinical data also suggest that combining dietary support with exercise synergistically improves muscle recovery and metabolic adaptation in cisplatin-treated models [239]. Therefore, dietary intervention—particularly amino acid, antioxidant, and micronutrient supplementation—represents a promising adjunct strategy to attenuate chemotherapy-induced muscle atrophy [245].

6.6. Future Directions

In summary, future research should aim to (1) standardize preclinical models, (2) clarify the integrated mechanisms of different exercise modalities, (3) establish reliable biomarkers such as myokines, and (4) translate preclinical insights into personalized clinical interventions. Addressing these challenges will not only deepen mechanistic understanding but also accelerate the implementation of exercise-based strategies to preserve skeletal muscle health in patients undergoing cisplatin chemotherapy [43,226,246].

7. Conclusions

Cisplatin-induced skeletal muscle atrophy represents a major clinical challenge that compromises treatment tolerance, immune competence, and quality of life in cancer patients. Current evidence indicates that exercise is a uniquely effective intervention, capable of targeting multiple molecular pathways simultaneously, including proteostasis, mitochondrial function, oxidative stress, and inflammatory signaling. Both endurance and resistance training, alone or in combination, confer protective benefits, while exercise-induced myokines provide a molecular bridge linking local muscle adaptation to systemic protection. Despite these advances, significant gaps remain in standardizing preclinical models, identifying translational biomarkers, and implementing personalized exercise prescriptions in clinical practice. Addressing these challenges will be critical for transforming exercise from a supportive measure into an evidence-based therapeutic strategy. Ultimately, integrating exercise into cancer care has the potential not only to preserve skeletal muscle health but also to improve overall patient outcomes during cisplatin-based chemotherapy.

Author Contributions

Writing—original draft, M.X. and X.L.; writing—review and editing, M.X. and X.L.; methodology, M.X. and X.L.; supervision, M.X.; conceptualization, X.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the (Ministry of Education in China) Project of Humanities and Social Sciences (No. 22YJC890014), National Natural Science Foundation of China (No. 32300964), Guangdong Basic and Applied Basic Research Foundation (No. 2022A1515111105), Guangzhou Basic and Applied Basic Research Foundation (No. 2023A04J0552), and Guangdong Provincial Sports Bureau 2024-2025 Science and Technology Innovation and Sports Culture Development Research Project (No. GDSS2024N010).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

During the preparation of this manuscript, the authors used Grammarly (premium version) for language polishing and grammar checking only. The authors have reviewed and edited all text and take full responsibility for the content of this publication.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sartori, R.; Romanello, V.; Sandri, M. Mechanisms of Muscle Atrophy and Hypertrophy: Implications in Health and Disease. Nat. Commun. 2021, 12, 330. [Google Scholar] [CrossRef]
  2. Wong, J.P.H.; Ng, Y.-K.; Kjærgaard, J.; Blazev, R.; Deshmukh, A.S.; Parker, B.L. Skeletal Muscle Proteomics: Considerations and Opportunities. npj Metab. Health Dis. 2025, 3, 30. [Google Scholar] [CrossRef]
  3. Bernabe-Ortiz, A.; Carrillo-Larco, R.M.; Gilman, R.H.; Smeeth, L.; Checkley, W.; Miranda, J.J. Skeletal Muscle Mass and All-Cause Mortality: Findings from the CRONICAS Cohort Study. Trop. Med. Int. Health 2023, 28, 107–115. [Google Scholar] [CrossRef] [PubMed]
  4. Cruz-Jentoft, A.J.; Dawson Hughes, B.; Scott, D.; Sanders, K.M.; Rizzoli, R. Nutritional Strategies for Maintaining Muscle Mass and Strength from Middle Age to Later Life: A Narrative Review. Maturitas 2020, 132, 57–64. [Google Scholar] [CrossRef] [PubMed]
  5. Janssen, T.A.H.; Lowisz, C.V.; Phillips, S. From Molecular to Physical Function: The Aging Trajectory. Curr. Res. Physiol. 2025, 8, 100138. [Google Scholar] [CrossRef]
  6. Deane, C.S.; Piasecki, M.; Atherton, P.J. Skeletal Muscle Immobilisation-Induced Atrophy: Mechanistic Insights from Human Studies. Clin. Sci. 2024, 138, 741–756. [Google Scholar] [CrossRef] [PubMed]
  7. Huang, L.; Li, M.; Deng, C.; Qiu, J.; Wang, K.; Chang, M.; Zhou, S.; Gu, Y.; Shen, Y.; Wang, W.; et al. Potential Therapeutic Strategies for Skeletal Muscle Atrophy. Antioxidants 2023, 12, 44. [Google Scholar] [CrossRef]
  8. Damluji, A.A.; Alfaraidhy, M.; AlHajri, N.; Rohant, N.N.; Kumar, M.; Al Malouf, C.; Bahrainy, S.; Ji Kwak, M.; Batchelor, W.B.; Forman, D.E.; et al. Sarcopenia and Cardiovascular Diseases. Circulation 2023, 147, 1534–1553. [Google Scholar] [CrossRef]
  9. Lavalle, S.; Valerio, M.R.; Masiello, E.; Gebbia, V.; Scandurra, G. Unveiling the Intricate Dance: How Cancer Orchestrates Muscle Wasting and Sarcopenia. In Vivo 2024, 38, 1520–1529. [Google Scholar] [CrossRef]
  10. Setiawan, T.; Sari, I.N.; Wijaya, Y.T.; Julianto, N.M.; Muhammad, J.A.; Lee, H.; Chae, J.H.; Kwon, H.Y. Cancer Cachexia: Molecular Mechanisms and Treatment Strategies. J. Hematol. Oncol. 2023, 16, 54. [Google Scholar] [CrossRef]
  11. VanderVeen, B.N.; Murphy, E.A.; Carson, J.A. The Impact of Immune Cells on the Skeletal Muscle Microenvironment during Cancer Cachexia. Front. Physiol. 2020, 11, 1037. [Google Scholar] [CrossRef]
  12. Armstrong, V.S.; Fitzgerald, L.W.; Bathe, O.F. Cancer-Associated Muscle Wasting—Candidate Mechanisms and Molecular Pathways. Int. J. Mol. Sci. 2020, 21, 9268. [Google Scholar] [CrossRef]
  13. Zhong, P.; Li, X.; Li, J. Mechanisms, Assessment, and Exercise Interventions for Skeletal Muscle Dysfunction Post-Chemotherapy in Breast Cancer: From Inflammation Factors to Clinical Practice. Front. Oncol. 2025, 15, 1551561. [Google Scholar] [CrossRef]
  14. Gilmore, L.A.; Parry, T.L.; Thomas, G.A.; Khamoui, A.V. Skeletal Muscle Omics Signatures in Cancer Cachexia: Perspectives and Opportunities. J. Natl. Cancer Inst. Monogr. 2023, 2023, 30–42. [Google Scholar] [CrossRef] [PubMed]
  15. Lyu, J.; Mantz, L.; Katz-Agranov, N.S.; Ouyang, T.; Hanna, P.E.; Mejia, S.M.; Moreno, D.; Lee, M.; Cao, A.; Dinulos, J.E.; et al. Sarcopenia, Elevated Body Mass Index, and Platinum-Associated Adverse Events in Patients with Lung Cancer. J. Natl. Compr. Cancer Netw. 2025, 23, 393–400. [Google Scholar] [CrossRef]
  16. Huang, K.; Chiang, Y.; Ali, M.; Hsia, S. Cisplatin-induced Muscle Wasting and Atrophy: Molecular Mechanism and Potential Therapeutic Interventions. J. Cachexia Sarcopenia Muscle 2025, 16, e13817. [Google Scholar] [CrossRef] [PubMed]
  17. Huang, K.; Chiang, Y.; Huang, T.; Chen, H.; Lin, P.; Ali, M.; Hsia, S. Capsaicin Alleviates Cisplatin-induced Muscle Loss and Atrophy in Vitro and in Vivo. J. Cachexia Sarcopenia Muscle 2022, 14, 182–197. [Google Scholar] [CrossRef] [PubMed]
  18. Liu, X.; Xu, M.; Yu, Y.; Chen, Y.; Weng, X.; Zhu, L. PD-1 Alleviates Cisplatin-Induced Muscle Atrophy by Regulating Inflammation and Oxidative Stress. Antioxidants 2022, 11, 1839. [Google Scholar] [CrossRef]
  19. Huot, J.R.; Pin, F.; Chatterjee, R.; Bonetto, A. PGC1α Overexpression Preserves Muscle Mass and Function in Cisplatin-induced Cachexia. J. Cachexia Sarcopenia Muscle 2022, 13, 2480–2491. [Google Scholar] [CrossRef]
  20. Conte, E.; Bresciani, E.; Rizzi, L.; Cappellari, O.; De Luca, A.; Torsello, A.; Liantonio, A. Cisplatin-Induced Skeletal Muscle Dysfunction: Mechanisms and Counteracting Therapeutic Strategies. Int. J. Mol. Sci. 2020, 21, 1242. [Google Scholar] [CrossRef]
  21. Lee, C.; Jeong, H.; Lee, H.; Hong, M.; Park, S.; Bae, H. Magnolol Attenuates Cisplatin-Induced Muscle Wasting by M2c Macrophage Activation. Front. Immunol. 2020, 11, 77. [Google Scholar] [CrossRef]
  22. Lønbro, S.; Gam, S.; Hermann, A.P.; Hansen, C.R.; Johansen, J. Accelerated Loss of Lean Body Mass in Head and Neck Cancer Patients during Cisplatin-Based Chemoradiation. Acta Oncol. 2023, 62, 1403–1411. [Google Scholar] [CrossRef] [PubMed]
  23. Park, S.E.; Choi, J.H.; Park, J.Y.; Kim, B.J.; Kim, J.G.; Kim, J.W.; Park, J.-M.; Chi, K.-C.; Hwang, I.G. Loss of Skeletal Muscle Mass during Palliative Chemotherapy Is a Poor Prognostic Factor in Patients with Advanced Gastric Cancer. Sci. Rep. 2020, 10, 17683. [Google Scholar] [CrossRef] [PubMed]
  24. You, L. Dihydromyricetin Inhibits Ferroptosis to Attenuate Cisplatin-Induced Muscle Atrophy. Physiol. Res. 2024, 73, 405–413. [Google Scholar] [CrossRef]
  25. Yeh, S.-L.; Chen, P.-Y.; Liao, J.-W.; Huang, R.-L.; Yu, S.-H.; Chen, L.-N.; Lee, M.-H.; Chen, L.-W.; Chen, H.-W.; Yang, Y.-C.; et al. The Protective Effects of Perch Essence against Muscle Atrophy in Cancer Cachexia and Cisplatin Treatment. Curr. Issues Mol. Biol. 2025, 47, 152. [Google Scholar] [CrossRef]
  26. Xu, M.; Liu, X.; Bao, P.; Wang, Y.J.; Lu, J.; Liu, Y.J. H2S Protects against Immobilization-Induced Muscle Atrophy via Reducing Oxidative Stress and Inflammation. Front. Physiol. 2022, 13, 844539. [Google Scholar] [CrossRef]
  27. Xu, M.; Liu, X.; Bao, P.; Wang, Y.; Zhu, X.; Liu, Y.; Ni, X.; Lu, J. Skeletal Muscle CSE Deficiency Leads to Insulin Resistance in Mice. Antioxidants 2022, 11, 2216. [Google Scholar] [CrossRef]
  28. Chen, L.-K. Sarcopenia in the Era of Precision Health: Toward Personalized Interventions for Healthy Longevity. J. Chin. Med. Assoc. 2024, 87, 980. [Google Scholar] [CrossRef]
  29. Pedrosa, M.B.; Barbosa, S.; Vitorino, R.; Ferreira, R.; Moreira-Gonçalves, D.; Santos, L.L. Chemotherapy-Induced Molecular Changes in Skeletal Muscle. Biomedicines 2023, 11, 905. [Google Scholar] [CrossRef] [PubMed]
  30. Nakagawa, N.; Yamamoto, S.; Hanai, A.; Oiwa, A.; Arao, H. Exercise Intervention for the Management of Chemotherapy-Induced Peripheral Neuropathy: A Systematic Review and Network Meta-Analysis. Front. Neurol. 2024, 15, 1346099. [Google Scholar] [CrossRef] [PubMed]
  31. Halle, J.L.; Counts, B.R.; Carson, J.A. Exercise as a Therapy for Cancer-Induced Muscle Wasting. Sports Med. Health Sci. 2020, 2, 186–194. [Google Scholar] [CrossRef]
  32. Cortiula, F.; Hendriks, L.E.L.; van de Worp, W.R.P.H.; Schols, A.M.W.J.; Vaes, R.D.W.; Langen, R.C.J.; De Ruysscher, D. Physical Exercise at the Crossroad between Muscle Wasting and the Immune System: Implications for Lung Cancer Cachexia. J. Cachexia Sarcopenia Muscle 2022, 13, 55–67. [Google Scholar] [CrossRef]
  33. Hardee, J.P.; Carson, J.A. Muscular Contraction’s Therapeutic Potential for Cancer-Induced Wasting. Am. J. Physiol. Cell Physiol. 2022, 323, C378–C384. [Google Scholar] [CrossRef] [PubMed]
  34. Mavropalias, G.; Sim, M.; Taaffe, D.R.; Galvão, D.A.; Spry, N.; Kraemer, W.J.; Häkkinen, K.; Newton, R.U. Exercise Medicine for Cancer Cachexia: Targeted Exercise to Counteract Mechanisms and Treatment Side Effects. J. Cancer Res. Clin. Oncol. 2022, 148, 1389–1406. [Google Scholar] [CrossRef]
  35. Aires, I.; Duarte, J.A.; Vitorino, R.; Moreira-Gonçalves, D.; Oliveira, P.; Ferreira, R. Restoring Skeletal Muscle Health through Exercise in Breast Cancer Patients and after Receiving Chemotherapy. Int. J. Mol. Sci. 2024, 25, 7533. [Google Scholar] [CrossRef]
  36. Wang, Y.-H.; Tan, J.; Zhou, H.-H.; Cao, M.; Zou, Y. Long-Term Exercise Training and Inflammatory Biomarkers in Healthy Subjects: A Meta-Analysis of Randomized Controlled Trials. Front. Psychol. 2023, 14, 1253329. [Google Scholar] [CrossRef]
  37. Feng, Y.; Rao, Z.; Tian, X.; Hu, Y.; Yue, L.; Meng, Y.; Zhong, Q.; Chen, W.; Xu, W.; Li, H.; et al. Endurance Training Enhances Skeletal Muscle Mitochondrial Respiration by Promoting MOTS-c Secretion. Free Radic. Biol. Med. 2025, 227, 619–628. [Google Scholar] [CrossRef] [PubMed]
  38. Pham, T.C.P.; Raun, S.H.; Havula, E.; Henriquez-Olguín, C.; Rubalcava-Gracia, D.; Frank, E.; Fritzen, A.M.; Jannig, P.R.; Andersen, N.R.; Kruse, R.; et al. The Mitochondrial mRNA-Stabilizing Protein SLIRP Regulates Skeletal Muscle Mitochondrial Structure and Respiration by Exercise-Recoverable Mechanisms. Nat. Commun. 2024, 15, 9826. [Google Scholar] [CrossRef] [PubMed]
  39. Dong, H.; Tsai, S.-Y. Mitochondrial Properties in Skeletal Muscle Fiber. Cells 2023, 12, 2183. [Google Scholar] [CrossRef]
  40. Fu, P.; Zhu, R.; Gao, W.; Gong, L. Effects of Resistance Training on Alleviating Hypoxia-induced Muscle Atrophy: Focus on Acetylation of FoxO1. J. Cell. Mol. Med. 2023, 28, e18096. [Google Scholar] [CrossRef]
  41. Pang, X.; Zhang, P.; Chen, X.; Liu, W. Ubiquitin-Proteasome Pathway in Skeletal Muscle Atrophy. Front. Physiol. 2023, 14, 1289537. [Google Scholar] [CrossRef]
  42. Bodine, S.C. The Role of mTORC1 in the Regulation of Skeletal Muscle Mass. Fac. Rev. 2022, 11, 32. [Google Scholar] [CrossRef] [PubMed]
  43. Lu, Z.; Wang, Z.; Zhang, X.-A.; Ning, K. Myokines May Be the Answer to the Beneficial Immunomodulation of Tailored Exercise—A Narrative Review. Biomolecules 2024, 14, 1205. [Google Scholar] [CrossRef]
  44. Bettariga, F.; Taaffe, D.R.; Galvão, D.A.; Lopez, P.; Bishop, C.; Markarian, A.M.; Natalucci, V.; Kim, J.-S.; Newton, R.U. Exercise Training Mode Effects on Myokine Expression in Healthy Adults: A Systematic Review with Meta-Analysis. J. Sport Health Sci. 2024, 13, 764–779. [Google Scholar] [CrossRef]
  45. Trettel, C.d.S.; Pelozin, B.R.d.A.; Barros, M.P.; Bachi, A.L.L.; Braga, P.G.S.; Momesso, C.M.; Furtado, G.E.; Valente, P.A.; Oliveira, E.M.; Hogervorst, E.; et al. Irisin: An Anti-Inflammatory Exerkine in Aging and Redox-Mediated Comorbidities. Front. Endocrinol. 2023, 14, 1106529. [Google Scholar] [CrossRef]
  46. Zhao, C.; Wu, Y.; Zhu, S.; Liu, H.; Xu, S. Irisin Protects Musculoskeletal Homeostasis via a Mitochondrial Quality Control Mechanism. Int. J. Mol. Sci. 2024, 25, 10116. [Google Scholar] [CrossRef] [PubMed]
  47. Han, F.; Ding, Z.; Shi, X.; Zhu, Q.; Shen, Q.; Xu, X.; Zhang, J.; Gong, W.; Xiao, W.; Wang, D.; et al. Irisin Inhibits Neutrophil Extracellular Traps Formation and Protects against Acute Pancreatitis in Mice. Redox Biol. 2023, 64, 102787. [Google Scholar] [CrossRef]
  48. Duan, Z.; Yang, Y.; Qin, M.; Yi, X. Interleukin 15: A New Intermediary in the Effects of Exercise and Training on Skeletal Muscle and Bone Function. J. Cell. Mol. Med. 2024, 28, e70136. [Google Scholar] [CrossRef]
  49. Khalafi, M.; Maleki, A.H.; Symonds, M.E.; Sakhaei, M.H.; Rosenkranz, S.K.; Ehsanifar, M.; Korivi, M.; Liu, Y. Interleukin-15 Responses to Acute and Chronic Exercise in Adults: A Systematic Review and Meta-Analysis. Front. Immunol. 2024, 14, 1288537. [Google Scholar] [CrossRef] [PubMed]
  50. Balanyà-Segura, M.; Polishchuk, A.; Just-Borràs, L.; Cilleros-Mañé, V.; Silvera, C.; Ardévol, A.; Tomàs, M.; Lanuza, M.A.; Hurtado, E.; Tomàs, J. Molecular Adaptations of BDNF/NT-4 Neurotrophic and Muscarinic Pathways in Ageing Neuromuscular Synapses. Int. J. Mol. Sci. 2024, 25, 8018. [Google Scholar] [CrossRef]
  51. Chan, W.S.; Ng, C.F.; Pang, B.P.S.; Hang, M.; Tse, M.C.L.; Iu, E.C.Y.; Ooi, X.C.; Yang, X.; Kim, J.K.; Lee, C.W.; et al. Exercise-Induced BDNF Promotes PPARδ-Dependent Reprogramming of Lipid Metabolism in Skeletal Muscle during Exercise Recovery. Sci. Signal. 2024, 17, eadh2783. [Google Scholar] [CrossRef]
  52. Thakir, T.M.; Wang, A.R.; Decker-Farrell, A.R.; Ferrer, M.; Guin, R.N.; Kleeman, S.; Levett, L.; Zhao, X.; Janowitz, T. Cancer Therapy and Cachexia. J. Clin. Investig. 2025, 135, e191934. [Google Scholar] [CrossRef]
  53. Hsu, W.-H.; Ko, A.-T.; Weng, C.-S.; Chang, C.-L.; Jan, Y.-T.; Lin, J.-B.; Chien, H.-J.; Lin, W.-C.; Sun, F.-J.; Wu, K.-P.; et al. Explainable Machine Learning Model for Predicting Skeletal Muscle Loss during Surgery and Adjuvant Chemotherapy in Ovarian Cancer. J. Cachexia Sarcopenia Muscle 2023, 14, 2044–2053. [Google Scholar] [CrossRef]
  54. Weng, C.; Huang, W.; Chang, C.; Jan, Y.; Chen, T.; Lee, J. Association of Malignant Ascites with Systemic Inflammation and Muscle Loss after Treatment in Advanced-stage Ovarian Cancer. J. Cachexia Sarcopenia Muscle 2023, 14, 2114–2125. [Google Scholar] [CrossRef]
  55. Amitani, M.; Oba, T.; Kiyosawa, N.; Morikawa, H.; Chino, T.; Soma, A.; Shimizu, T.; Ohno, K.; Ono, M.; Ito, T.; et al. Skeletal Muscle Loss during Neoadjuvant Chemotherapy Predicts Poor Prognosis in Patients with Breast Cancer. BMC Cancer 2022, 22, 327. [Google Scholar] [CrossRef]
  56. Becker, J.-N.; Hermann, R.; Wichmann, J.; Sonnhoff, M.; Christiansen, H.; Bruns, F. Low Skeletal Muscle Mass Is Predictive of Dose-Limiting Toxicities in Head and Neck Cancer Patients Undergoing Low-Dose Weekly Cisplatin Chemoradiotherapy. PLoS ONE 2023, 18, e0282015. [Google Scholar] [CrossRef]
  57. Suzuki, S.; Yokota, T.; Notsu, A.; Hamauchi, S.; Onozawa, Y.; Fushiki, K.; Oshima, K.; Kawakami, T.; Tsushima, T.; Yasui, H.; et al. Impact of Relative Cisplatin Dose to Skeletal Muscle Mass on Adverse Events in Patients with Head and Neck Cancer Undergoing Chemoradiotherapy. Oncologist 2024, 29, e1315–e1323. [Google Scholar] [CrossRef]
  58. Jang, M.K.; Park, S.; Park, C.; Doorenbos, A.Z.; Go, J.; Kim, S. Does Neoadjuvant Chemotherapy Regimen Affect Sarcopenia Status in Patients with Breast Cancer? Breast 2022, 66, 1–7. [Google Scholar] [CrossRef]
  59. Van der Zanden, V.; van Soolingen, N.J.; Viddeleer, A.R.; Trum, J.W.; Amant, F.; Mourits, M.J.E.; Portielje, J.E.A.; Baalbergen, A.; Souwer, E.T.D.; van Munster, B.C. Loss of Skeletal Muscle Density during Neoadjuvant Chemotherapy in Older Women with Advanced Stage Ovarian Cancer Is Associated with Postoperative Complications. Eur. J. Surg. Oncol. 2022, 48, 896–902. [Google Scholar] [CrossRef]
  60. Schaeffers, A.W.M.A.; Scholten, H.A.; van Beers, M.A.; Meussen, B.W.; Smid, E.J.; van Gils, C.H.; Devriese, L.A.; de Bree, R. The Effect of Skeletal Muscle Mass on Dose-Limiting Toxicities during (Chemo)Radiotherapy in Patients with Head and Neck Cancer: A Systematic Review and Meta-Analysis. Oral. Oncol. 2024, 157, 106978. [Google Scholar] [CrossRef]
  61. De Jong, C.; Chargi, N.; Herder, G.J.M.; van Haarlem, S.W.A.; van der Meer, F.; van Lindert, A.S.R.; ten Heuvel, A.; Brouwer, J.; de Jong, P.A.; Devriese, L.A.; et al. The Association between Skeletal Muscle Measures and Chemotherapy-induced Toxicity in Non-small Cell Lung Cancer Patients. J. Cachexia Sarcopenia Muscle 2022, 13, 1554–1564. [Google Scholar] [CrossRef]
  62. Purcell, S.A.; Kok, D.E.; Ketterl, T.; Garcia, M.B.; Joffe, L.; Brown, J.C.; Dieli-Conwright, C.M.; Williams, G.R. Pharmacokinetics of Cancer Therapeutics and Energy Balance: The Role of Diet Intake, Energy Expenditure, and Body Composition. J. Natl. Cancer Inst. Monogr. 2023, 2023, 3–11. [Google Scholar] [CrossRef]
  63. Schmulenson, E.; Zimmermann, N.; Müller, L.; Kapsa, S.; Sihinevich, I.; Jaehde, U. Influence of the Skeletal Muscle Index on Pharmacokinetics and Toxicity of Fluorouracil. Cancer Med. 2022, 12, 2580–2589. [Google Scholar] [CrossRef]
  64. Mallard, J.; Hucteau, E.; Schott, R.; Trensz, P.; Pflumio, C.; Kalish-Weindling, M.; Favret, F.; Pivot, X.; Hureau, T.J.; Pagano, A.F. Early Skeletal Muscle Deconditioning and Reduced Exercise Capacity during (Neo)Adjuvant Chemotherapy in Patients with Breast Cancer. Cancer 2023, 129, 215–225. [Google Scholar] [CrossRef]
  65. Phan, T.T.; Scott, K.S.; Chelette, B.; West, A.P.; Dantzer, R. The Fatigue-Inducing Effects of Cancer and Its Therapy Are Characterized by Decreased Physical Activity in the Absence of Any Motivational Deficit. Brain Behav. Immun. 2024, 117, 205–214. [Google Scholar] [CrossRef]
  66. Jackson, K.M.; Cole, C.L.; Dunne, R.F. From Bench to Bedside: Updates in Basic Science, Translational and Clinical Research on Muscle Fatigue in Cancer Cachexia. Curr. Opin. Clin. Nutr. Metab. Care 2021, 24, 216–222. [Google Scholar] [CrossRef]
  67. Anabtawi, N.M.; Pasala, M.S.; Grimshaw, A.A.; Kharel, P.; Bal, S.; Godby, K.; Siwakoti, A.; Buford, T.W.; Bhatia, S.; Costa, L.J.; et al. Low Skeletal Muscle Mass and Treatment Outcomes among Adults with Haematologic Malignancies: A Systematic Review and Meta-analysis. J. Cachexia Sarcopenia Muscle 2024, 15, 1084–1093. [Google Scholar] [CrossRef]
  68. Luo, K.; Chen, K.; Li, Y.; Ji, Y. Association between Sarcopenia and Outcomes of Surgically Treated Oral Squamous Cell Carcinoma: A Systematic Review and Meta-analysis. Front. Oncol. 2024, 14, 1445956. [Google Scholar] [CrossRef]
  69. Huiskamp, L.F.J.; Chargi, N.; Devriese, L.A.; de Jong, P.A.; de Bree, R. The Predictive and Prognostic Value of Low Skeletal Muscle Mass for Dose-Limiting Toxicity and Survival in Head and Neck Cancer Patients Receiving Concomitant Cetuximab and Radiotherapy. Eur. Arch. Otorhinolaryngol. 2020, 277, 2847–2858. [Google Scholar] [CrossRef]
  70. Hatt, J.; Smart, T.F.F.; Hardy, E.J.; Doleman, B.; Lund, J.N.; Philips, B.E. The Impact of Low Muscle Mass on Prognosis Following Neoadjuvant Chemotherapy for Resectable Locally Advanced Rectal Cancer: A Systematic Review and Meta-Analysis. JCSM Clin. Rep. 2023, 8, 27–35. [Google Scholar] [CrossRef]
  71. Rinninella, E.; Fagotti, A.; Cintoni, M.; Raoul, P.; Scaletta, G.; Scambia, G.; Gasbarrini, A.; Mele, M.C. Skeletal Muscle Mass as a Prognostic Indicator of Outcomes in Ovarian Cancer: A Systematic Review and Meta-Analysis. Int. J. Gynecol. Cancer 2020, 30, 654–663. [Google Scholar] [CrossRef]
  72. Cheung, C.-L.; Lee, G.K.-Y.; Au, P.C.-M.; Li, G.H.-Y.; Chan, M.; Li, H.-L.; Cheung, B.M.-Y.; Wong, I.C.-K.; Lee, V.H.-F.; Mok, J.; et al. Systematic Review and Meta-Analysis of Lean Mass and Mortality: Rationale and Study Description. Osteoporos. Sarcopenia 2021, 7, S3–S12. [Google Scholar] [CrossRef]
  73. Chargi, N.; Wegner, I.; Markazi, N.; Smid, E.; de Jong, P.; Devriese, L.; de Bree, R. Patterns, Predictors, and Prognostic Value of Skeletal Muscle Mass Loss in Patients with Locally Advanced Head and Neck Cancer Undergoing Cisplatin-Based Chemoradiotherapy. J. Clin. Med. 2021, 10, 1762. [Google Scholar] [CrossRef]
  74. Leão, I.; Garcia, C.; Antunes, P.; Campolargo, A.; Dias, I.; Coimbra, E.; Oliveira, P.; Zenha, H.; Costa, H.; Capela, A.; et al. Acute Impact of Cancer Treatment on Head and Neck Cancer Patients: FIT4TREATMENT. Cancers 2022, 14, 2698. [Google Scholar] [CrossRef]
  75. Sekine, H.; Matsumoto, C.; Fujitsuka, N.; Mogami, S.; Ohnishi, S.; Takeda, H. Hochuekkito Accelerates Recovery from Cisplatin Induced-Muscle Atrophy Accompanied by Slow-Twitch Fiber-Specific microRNA Upregulation in Mice. Front. Pharmacol. 2025, 16, 1502563. [Google Scholar] [CrossRef]
  76. Zhang, H.; Chi, M.; Wang, Y.; Chen, L.; Sun, X.; Wan, L.; Yang, Q.; Guo, C. Naringenin Alleviates Cisplatin Induced Muscle Atrophy by Regulating RIPK1/AMPK/NF-κB Pathway. J. Funct. Foods 2021, 86, 104714. [Google Scholar] [CrossRef]
  77. Seo, D.Y.; Bae, J.H.; Zhang, D.; Song, W.; Kwak, H.-B.; Heo, J.-W.; Jung, S.-J.; Yun, H.R.; Kim, T.N.; Lee, S.H.; et al. Effects of Cisplatin on Mitochondrial Function and Autophagy-Related Proteins in Skeletal Muscle of Rats. BMB Rep. 2021, 54, 575–580. [Google Scholar] [CrossRef]
  78. Katanić Stanković, J.S.; Selaković, D.; Rosić, G. Oxidative Damage as a Fundament of Systemic Toxicities Induced by Cisplatin—The Crucial Limitation or Potential Therapeutic Target? Int. J. Mol. Sci. 2023, 24, 14574. [Google Scholar] [CrossRef]
  79. Matsumoto, C.; Sekine, H.; Nahata, M.; Mogami, S.; Ohbuchi, K.; Fujitsuka, N.; Takeda, H. Role of Mitochondrial Dysfunction in the Pathogenesis of Cisplatin-Induced Myotube Atrophy. Biol. Pharm. Bull. 2022, 45, 780–792. [Google Scholar] [CrossRef]
  80. Hsing, C.-H.; Tsai, C.-C.; Chen, C.-L.; Lin, Y.-H.; Tseng, P.-C.; Satria, R.D.; Lin, C.-F. Pharmacologically Inhibiting Glycogen Synthase Kinase-3β Ameliorates Renal Inflammation and Nephrotoxicity in an Animal Model of Cisplatin-Induced Acute Kidney Injury. Biomedicines 2021, 9, 887. [Google Scholar] [CrossRef]
  81. Rozhkov, S.V.; Sharlo, K.A.; Shenkman, B.S.; Mirzoev, T.M. The Role of Glycogen Synthase Kinase-3 in the Regulation of Ribosome Biogenesis in Rat Soleus Muscle under Disuse Conditions. Int. J. Mol. Sci. 2022, 23, 2751. [Google Scholar] [CrossRef]
  82. Qiu, L.; Sun, Y.; Ning, H.; Chen, G.; Zhao, W.; Gao, Y. The Scaffold Protein AXIN1: Gene Ontology, Signal Network, and Physiological Function. Cell Commun. Signaling 2024, 22, 77. [Google Scholar] [CrossRef]
  83. Sharma, T.; Olea-Flores, M.; Imbalzano, A.N. Regulation of the Wnt Signaling Pathway during Myogenesis by the Mammalian SWI/SNF ATPase BRG1. Front. Cell Dev. Biol. 2023, 11, 1160227. [Google Scholar] [CrossRef]
  84. Marcella, B.M.; Hockey, B.L.; Braun, J.L.; Whitley, K.C.; Geromella, M.S.; Baranowski, R.W.; Watson, C.J.F.; Silvera, S.; Hamstra, S.I.; Wasilewicz, L.J.; et al. GSK3 Inhibition Improves Skeletal Muscle Function and Whole-Body Metabolism in Male Mouse Models of Duchenne Muscular Dystrophy. Nat. Commun. 2024, 15, 10210. [Google Scholar] [CrossRef]
  85. Hockey, B.L.; Braun, J.L.; Vandenboom, R.; LeBlanc, P.J.; MacPherson, R.E.K.; Fajardo, V.A. Can the Inhibition of Glycogen Synthase Kinase 3 Mimic the Physiological Effects of Exercise on Muscle and Other Organs? Adv. Exerc. Health Sci. 2025, 2, 108–120. [Google Scholar] [CrossRef]
  86. Sato, K.; Satoshi, Y.; Miyauchi, Y.; Sato, F.; Kon, R.; Ikarashi, N.; Chiba, Y.; Hosoe, T.; Sakai, H. Downregulation of PGC-1α during Cisplatin-Induced Muscle Atrophy in Murine Skeletal Muscle. Biochim. Biophys. Acta Mol. Basis Dis. 2024, 1870, 166877. [Google Scholar] [CrossRef]
  87. Bhatt, B.J.; Ghosh, S.; Mazurak, V.; Brun, A.Q.; Bathe, O.; Baracos, V.E.; Damaraju, S. Molecular Subtypes of Human Skeletal Muscle in Cancer Cachexia. Nature 2025, 646, 973–982. [Google Scholar] [CrossRef]
  88. Liang, S.-M.; Kuo, C.-L.; Lu, Y.-J.; Chang, T.-C.; Liou, J.-Y. Antrodia Cinnamomea Extract Attenuates Cisplatin-Induced Muscle Atrophy, Apoptosis, and Cell Growth Suppression. J. Food Biochem. 2023, 2023, 5593854. [Google Scholar] [CrossRef]
  89. Park, S.-Y.; Hwang, B.-O.; Song, N.-Y. The Role of Myokines in Cancer: Crosstalk between Skeletal Muscle and Tumor. BMB Rep. 2023, 56, 365–373. [Google Scholar] [CrossRef]
  90. De Castro, G.S.; Correia-Lima, J.; Simoes, E.; Orsso, C.E.; Xiao, J.; Gama, L.R.; Gomes, S.P.; Gonçalves, D.C.; Costa, R.G.F.; Radloff, K.; et al. Myokines in Treatment-Naïve Patients with Cancer-Associated Cachexia. Clin. Nutr. 2021, 40, 2443–2455. [Google Scholar] [CrossRef]
  91. Law, M.L. Cancer Cachexia: Pathophysiology and Association with Cancer-Related Pain. Front. Pain Res. 2022, 3, 971295. [Google Scholar] [CrossRef]
  92. Ikeno, Y.; Inomata, M.; Tsukimura, Y.; Suzuki, Y.; Takeuchi, H.; Harada, Y.; Kon, R.; Ikarashi, N.; Chiba, Y.; Yamada, T.; et al. Eicosapentaenoic Acid Suppresses Cisplatin-Induced Muscle Atrophy by Attenuating the up-Regulated Gene Expression of Ubiquitin. J. Nutr. Biochem. 2022, 103, 108953. [Google Scholar] [CrossRef]
  93. Zhang, H.; Chi, M.; Chen, L.; Sun, X.; Wan, L.; Yang, Q.; Guo, C. Daidzein Alleviates Cisplatin-Induced Muscle Atrophy by Regulating Glut4/AMPK/FoxO Pathway. Phytother. Res. 2021, 35, 4363–4376. [Google Scholar] [CrossRef]
  94. Matsumoto, C.; Sekine, H.; Zhang, N.; Mogami, S.; Fujitsuka, N.; Takeda, H. Role of P53 in Cisplatin-Induced Myotube Atrophy. Int. J. Mol. Sci. 2023, 24, 9176. [Google Scholar] [CrossRef]
  95. Jang, J.; Lee, H.; Song, J.; Bae, T.; Park, M.; Kwon, Y.V.; Lee, D.; Yoon, Y. Paeonia Lactiflora Extract Suppresses Cisplatin-Induced Muscle Wasting via Downregulation of Muscle-Specific Ubiquitin E3 Ligases, NF-κB Signaling, and Cytokine Levels. J. Ethnopharmacol. 2021, 266, 113403. [Google Scholar] [CrossRef]
  96. Ji, L.L.; Yeo, D.; Kang, C. Muscle Disuse Atrophy Caused by Discord of Intracellular Signaling. Antioxid. Redox Signal. 2020, 33, 727–744. [Google Scholar] [CrossRef]
  97. Chen, X.; Ji, Y.; Liu, R.; Zhu, X.; Wang, K.; Yang, X.; Liu, B.; Gao, Z.; Huang, Y.; Shen, Y.; et al. Mitochondrial Dysfunction: Roles in Skeletal Muscle Atrophy. J. Transl. Med. 2023, 21, 503. [Google Scholar] [CrossRef]
  98. Mirzoev, T.M. Skeletal Muscle Recovery from Disuse Atrophy: Protein Turnover Signaling and Strategies for Accelerating Muscle Regrowth. Int. J. Mol. Sci. 2020, 21, 7940. [Google Scholar] [CrossRef]
  99. Misiąg, W.; Piszczyk, A.; Szymańska-Chabowska, A.; Chabowski, M. Physical Activity and Cancer Care—A Review. Cancers 2022, 14, 4154. [Google Scholar] [CrossRef]
  100. Brook, M.S. Investigating Muscle Protein Synthesis Using Deuterium Oxide: The Impact of Dietary Protein Interventions across the Lifespan. Exp. Physiol. 2025, 110, 949–960. [Google Scholar] [CrossRef]
  101. Lim, C.; McKendry, J.; Lees, M.; Atherton, P.J.; Burd, N.A.; Holwerda, A.M.; van Loon, L.J.C.; McGlory, C.; Mitchell, C.J.; Smith, K.; et al. Turning over New Ideas in Human Skeletal Muscle Proteostasis: What Do We Know and Where to from Here? Exp. Physiol. 2025. [Google Scholar] [CrossRef] [PubMed]
  102. Lei, Y.; Gan, M.; Qiu, Y.; Chen, Q.; Wang, X.; Liao, T.; Zhao, M.; Chen, L.; Zhang, S.; Zhao, Y.; et al. The Role of Mitochondrial Dynamics and Mitophagy in Skeletal Muscle Atrophy: From Molecular Mechanisms to Therapeutic Insights. Cell. Mol. Biol. Lett. 2024, 29, 59. [Google Scholar] [CrossRef]
  103. Sakai, H.; Ikeno, Y.; Tsukimura, Y.; Inomata, M.; Suzuki, Y.; Kon, R.; Ikarashi, N.; Chiba, Y.; Yamada, T.; Kamei, J. Upregulation of Ubiquitinated Proteins and Their Degradation Pathway in Muscle Atrophy Induced by Cisplatin in Mice. Toxicol. Appl. Pharmacol. 2020, 403, 115165. [Google Scholar] [CrossRef]
  104. Sakai, H.; Zhou, Y.; Miyauchi, Y.; Suzuki, Y.; Ikeno, Y.; Kon, R.; Ikarashi, N.; Chiba, Y.; Hosoe, T.; Kamei, J. Increased 20S Proteasome Expression and the Effect of Bortezomib during Cisplatin-Induced Muscle Atrophy. Biol. Pharm. Bull. 2022, 45, 910–918. [Google Scholar] [CrossRef]
  105. Bae, J.H.; Seo, D.Y.; Lee, S.H.; Shin, C.; Jamrasi, P.; Han, J.; Song, W. Effects of Exercise on AKT/PGC1-α/FOXO3a Pathway and Muscle Atrophy in Cisplatin-Administered Rat Skeletal Muscle. Korean J. Physiol. Pharmacol. 2021, 25, 585–592. [Google Scholar] [CrossRef]
  106. Yin, L.; Li, N.; Jia, W.; Wang, N.; Liang, M.; Yang, X.; Du, G. Skeletal Muscle Atrophy: From Mechanisms to Treatments. Pharmacol. Res. 2021, 172, 105807. [Google Scholar] [CrossRef]
  107. Plotkin, D.L.; Mattingly, M.L.; Anglin, D.A.; Michel, J.M.; Godwin, J.S.; McIntosh, M.C.; Kontos, N.J.; Bergamasco, J.G.A.; Scarpelli, M.C.; Angleri, V.; et al. Skeletal Muscle Myosin Heavy Chain Fragmentation as a Potential Marker of Protein Degradation in Response to Resistance Training and Disuse Atrophy. Exp. Physiol. 2024, 109, 1739–1754. [Google Scholar] [CrossRef]
  108. Gao, S.; Zhang, G.; Zhang, Z.; Zhu, J.Z.; Li, L.; Zhou, Y.; Rodney, G.G.; Abo-Zahrah, R.S.; Anderson, L.; Garcia, J.M.; et al. UBR2 Targets Myosin Heavy Chain IIb and IIx for Degradation: Molecular Mechanism Essential for Cancer-Induced Muscle Wasting. Proc. Natl. Acad. Sci. USA 2022, 119, e2200215119. [Google Scholar] [CrossRef]
  109. Yang, Y.; Liu, L.; Tian, Y.; Gu, M.; Wang, Y.; Ashrafizadeh, M.; Reza Aref, A.; Cañadas, I.; Klionsky, D.J.; Goel, A.; et al. Autophagy-Driven Regulation of Cisplatin Response in Human Cancers: Exploring Molecular and Cell Death Dynamics. Cancer Lett. 2024, 587, 216659. [Google Scholar] [CrossRef]
  110. Cao, Z.; Tian, K.; Ran, Y.; Zhou, H.; Zhou, L.; Ding, Y.; Tang, X. Beclin-1: A Therapeutic Target at the Intersection of Autophagy, Immunotherapy, and Cancer Treatment. Front. Immunol. 2024, 15, 1506426. [Google Scholar] [CrossRef] [PubMed]
  111. Xia, Q.; Huang, X.; Huang, J.; Zheng, Y.; March, M.E.; Li, J.; Wei, Y. The Role of Autophagy in Skeletal Muscle Diseases. Front. Physiol. 2021, 12, 638983. [Google Scholar] [CrossRef]
  112. Sebastián, D.; Zorzano, A. Self-Eating for Muscle Fitness: Autophagy in the Control of Energy Metabolism. Dev. Cell 2020, 54, 268–281. [Google Scholar] [CrossRef] [PubMed]
  113. Sakai, H.; Asami, M.; Naito, H.; Kitora, S.; Suzuki, Y.; Miyauchi, Y.; Tachinooka, R.; Yoshida, S.; Kon, R.; Ikarashi, N.; et al. Exogenous Insulin-like Growth Factor 1 Attenuates Cisplatin-Induced Muscle Atrophy in Mice. J. Cachexia Sarcopenia Muscle 2021, 12, 1570–1581. [Google Scholar] [CrossRef] [PubMed]
  114. Pan, Z.; Zhang, H.; Dokudovskaya, S. The Role of mTORC1 Pathway and Autophagy in Resistance to Platinum-Based Chemotherapeutics. Int. J. Mol. Sci. 2023, 24, 10651. [Google Scholar] [CrossRef]
  115. Cormerais, Y.; Lapp, S.C.; Kalafut, K.C.; Cissé, M.Y.; Shin, J.; Stefadu, B.; Personnaz, J.; Schrötter, S.; Freed, J.; D’Amore, A.; et al. AKT-Mediated Phosphorylation of TSC2 Controls Stimulus- and Tissue-Specific mTORC1 Signaling and Organ Growth. Dev. Cell 2025, 60, 2544–2557. [Google Scholar] [CrossRef]
  116. Panwar, V.; Singh, A.; Bhatt, M.; Tonk, R.K.; Azizov, S.; Raza, A.S.; Sengupta, S.; Kumar, D.; Garg, M. Multifaceted Role of mTOR (Mammalian Target of Rapamycin) Signaling Pathway in Human Health and Disease. Sig. Transduct. Target. Ther. 2023, 8, 375. [Google Scholar] [CrossRef]
  117. Maharati, A.; Rajabloo, Y.; Moghbeli, M. Molecular Mechanisms of mTOR-Mediated Cisplatin Response in Tumor Cells. Heliyon 2024, 11, e41483. [Google Scholar] [CrossRef]
  118. Lu, W.; Ni, K.; Li, Z.; Xiao, L.; Li, Y.; Jiang, Y.; Zhang, J.; Shi, H. Salubrinal Protects against Cisplatin-Induced Cochlear Hair Cell Endoplasmic Reticulum Stress by Regulating Eukaryotic Translation Initiation Factor 2α Signalling. Front. Mol. Neurosci. 2022, 15, 916458. [Google Scholar] [CrossRef]
  119. Zhang, Q.; Huang, Z.; Rui, X.; Wang, Y.; Wang, Y.; Zhou, Y.; Chen, R.; Chen, Y.; Wang, Y.; Li, S.; et al. GSDMD Enhances Cisplatin-Induced Apoptosis by Promoting the Phosphorylation of eIF2α and Activating the ER-Stress Response. Cell Death Discov. 2022, 8, 114. [Google Scholar] [CrossRef]
  120. Shu, S.; Wang, H.; Zhu, J.; Fu, Y.; Cai, J.; Chen, A.; Tang, C.; Dong, Z. Endoplasmic Reticulum Stress Contributes to Cisplatin-Induced Chronic Kidney Disease via the PERK–PKCδ Pathway. Cell. Mol. Life Sci. 2022, 79, 452. [Google Scholar] [CrossRef] [PubMed]
  121. Mu, W.; Zhi, Y.; Zhou, J.; Wang, C.; Chai, K.; Fan, Z.; Lv, G. Endoplasmic Reticulum Stress and Quality Control in Relation to Cisplatin Resistance in Tumor Cells. Front. Pharmacol. 2024, 15, 1419468. [Google Scholar] [CrossRef]
  122. Kim, M.-C.; Hwang, S.-H.; Yang, Y.; Kim, N.-Y.; Kim, Y. Reduction in Mitochondrial Oxidative Stress Mediates Hypoxia-Induced Resistance to Cisplatin in Human Transitional Cell Carcinoma Cells. Neoplasia 2021, 23, 653–662. [Google Scholar] [CrossRef]
  123. Lin, Y.; Yang, B.; Huang, Y.; Zhang, Y.; Jiang, Y.; Ma, L.; Shen, Y.-Q. Mitochondrial DNA-Targeted Therapy: A Novel Approach to Combat Cancer. Cell Insight 2023, 2, 100113. [Google Scholar] [CrossRef]
  124. Elmorsy, E.A.; Saber, S.; Hamad, R.S.; Abdel-Reheim, M.A.; El-Kott, A.F.; AlShehri, M.A.; Morsy, K.; Salama, S.A.; Youssef, M.E. Advances in Understanding Cisplatin-Induced Toxicity: Molecular Mechanisms and Protective Strategies. Eur. J. Pharm. Sci. 2024, 203, 106939. [Google Scholar] [CrossRef]
  125. Li, F.; Zhu, F.; Wang, S.; Hu, H.; Zhang, D.; He, Z.; Chen, J.; Li, X.; Cheng, L.; Zhong, F. Icariin Alleviates Cisplatin-Induced Premature Ovarian Failure by Inhibiting Ferroptosis through Activation of the Nrf2/ARE Pathway. Sci. Rep. 2024, 14, 17318. [Google Scholar] [CrossRef]
  126. Chi, M.; Zhang, H.; Wang, Y.; Sun, X.; Yang, Q.; Guo, C. Silibinin Alleviates Muscle Atrophy Caused by Oxidative Stress Induced by Cisplatin through ERK/FoxO and JNK/FoxO Pathways. Oxid. Med. Cell. Longev. 2022, 2022, 5694223. [Google Scholar] [CrossRef] [PubMed]
  127. Zhang, H.; Chi, M.; Chen, L.; Sun, X.; Wan, L.; Yang, Q.; Guo, C. Linalool Prevents Cisplatin Induced Muscle Atrophy by Regulating IGF-1/Akt/FoxO Pathway. Front. Pharmacol. 2020, 11, 598166. [Google Scholar] [CrossRef] [PubMed]
  128. Rahman, F.A.; Graham, M.Q.; Adam, A.M.; Juracic, E.S.; Tupling, A.R.; Quadrilatero, J. Mitophagy Is Required to Protect against Excessive Skeletal Muscle Atrophy Following Hindlimb Immobilization. J. Biomed. Sci. 2025, 32, 29. [Google Scholar] [CrossRef]
  129. Domingo, I.K.; Latif, A.; Bhavsar, A.P. Pro-Inflammatory Signalling PRRopels Cisplatin-Induced Toxicity. Int. J. Mol. Sci. 2022, 23, 7227. [Google Scholar] [CrossRef] [PubMed]
  130. Ji, Y.; Li, M.; Chang, M.; Liu, R.; Qiu, J.; Wang, K.; Deng, C.; Shen, Y.; Zhu, J.; Wang, W.; et al. Inflammation: Roles in Skeletal Muscle Atrophy. Antioxidants 2022, 11, 1686. [Google Scholar] [CrossRef]
  131. Huang, Z.; Zhong, L.; Zhu, J.; Xu, H.; Ma, W.; Zhang, L.; Shen, Y.; Law, B.Y.-K.; Ding, F.; Gu, X.; et al. Inhibition of IL-6/JAK/STAT3 Pathway Rescues Denervation-Induced Skeletal Muscle Atrophy. Ann. Transl. Med. 2020, 8, 1681. [Google Scholar] [CrossRef]
  132. Xie, T.; Lv, T.; Zhang, T.; Feng, D.; Zhu, F.; Xu, Y.; Zhang, L.; Gu, L.; Guo, Z.; Ding, C.; et al. Interleukin-6 Promotes Skeletal Muscle Catabolism by Activating Tryptophan–Indoleamine 2,3-Dioxygenase 1–Kynurenine Pathway during Intra-Abdominal Sepsis. J. Cachexia Sarcopenia Muscle 2023, 14, 1046–1059. [Google Scholar] [CrossRef] [PubMed]
  133. Lv, X.; Ding, S. Unraveling the Role of STAT3 in Cancer Cachexia: Pathogenic Mechanisms and Therapeutic Opportunities. Front. Endocrinol. 2025, 16, 1608612. [Google Scholar] [CrossRef]
  134. Chen, Z.; Lan, H.; Liao, Z.; Huang, J.; Jian, X.; Hu, J.; Liao, H. Regulatory T Cells-Centered Regulatory Networks of Skeletal Muscle Inflammation and Regeneration. Cell Biosci. 2022, 12, 112. [Google Scholar] [CrossRef] [PubMed]
  135. Giovarelli, M.; Mocciaro, E.; Carnovale, C.; Cervia, D.; Perrotta, C.; Clementi, E. Immunosenescence in Skeletal Muscle: The Role-Play in Cancer Cachexia Chessboard. Semin. Cancer Biol. 2025, 111, 48–59. [Google Scholar] [CrossRef] [PubMed]
  136. Negrón-Piñeiro, L.J.; Wu, Y.; Popsuj, S.; José-Edwards, D.S.; Stolfi, A.; Di Gregorio, A. Cis-Regulatory Interfaces Reveal the Molecular Mechanisms Underlying the Notochord Gene Regulatory Network of Ciona. Nat. Commun. 2024, 15, 3025. [Google Scholar] [CrossRef]
  137. Ali, S.I.M.; Alrashid, S.Z. A Review of Methods for Gene Regulatory Networks Reconstruction and Analysis. Artif. Intell. Rev. 2025, 58, 256. [Google Scholar] [CrossRef]
  138. Lian, D.; Chen, M.-M.; Wu, H.; Deng, S.; Hu, X. The Role of Oxidative Stress in Skeletal Muscle Myogenesis and Muscle Disease. Antioxidants 2022, 11, 755. [Google Scholar] [CrossRef]
  139. Xu, X.; Pang, Y.; Fan, X. Mitochondria in Oxidative Stress, Inflammation and Aging: From Mechanisms to Therapeutic Advances. Sig. Transduct. Target. Ther. 2025, 10, 190. [Google Scholar] [CrossRef]
  140. Shi, S.; Chen, Y.; Luo, Z.; Nie, G.; Dai, Y. Role of Oxidative Stress and Inflammation-Related Signaling Pathways in Doxorubicin-Induced Cardiomyopathy. Cell Commun. Signal. 2023, 21, 61. [Google Scholar] [CrossRef]
  141. Chen, P.; Jia, F.; Wang, M.; Yang, S. Analysis of the Mechanism of Skeletal Muscle Atrophy from the Pathway of Decreased Protein Synthesis. Front. Physiol. 2025, 16, 1533394. [Google Scholar] [CrossRef]
  142. Chen, X.; Shi, C.; He, M.; Xiong, S.; Xia, X. Endoplasmic Reticulum Stress: Molecular Mechanism and Therapeutic Targets. Sig. Transduct. Target. Ther. 2023, 8, 352. [Google Scholar] [CrossRef]
  143. Vietri, M.; Miranda, M.R.; Amodio, G.; Ciaglia, T.; Bertamino, A.; Campiglia, P.; Remondelli, P.; Vestuto, V.; Moltedo, O. The Link between Endoplasmic Reticulum Stress and Lysosomal Dysfunction under Oxidative Stress in Cancer Cells. Biomolecules 2025, 15, 930. [Google Scholar] [CrossRef] [PubMed]
  144. Pigg, Q.W.; Harris, D.R.; Inoue, D.S.; Janini Gomes, M. Mitigating Doxorubicin-Induced Skeletal Muscle Toxicity: A Review of Oxidative Stress Mechanisms and the Therapeutic Role of Exercise. Antioxidants 2025, 14, 870. [Google Scholar] [CrossRef]
  145. Sanft, T.; Harrigan, M.; McGowan, C.; Cartmel, B.; Zupa, M.; Li, F.-Y.; Ferrucci, L.M.; Puklin, L.; Cao, A.; Nguyen, T.H.; et al. Randomized Trial of Exercise and Nutrition on Chemotherapy Completion and Pathologic Complete Response in Women with Breast Cancer: The Lifestyle, Exercise, and Nutrition Early after Diagnosis Study. J. Clin. Oncol. 2023, 41, 5285–5295. [Google Scholar] [CrossRef] [PubMed]
  146. Bilski, J.; Szlachcic, A.; Ptak-Belowska, A.; Brzozowski, T. Physical Activity, Exerkines, and Their Role in Cancer Cachexia. Int. J. Mol. Sci. 2025, 26, 8011. [Google Scholar] [CrossRef]
  147. Pradhan, R.; Dieterich, W.; Natarajan, A.; Schwappacher, R.; Reljic, D.; Herrmann, H.J.; Neurath, M.F.; Zopf, Y. Influence of Amino Acids and Exercise on Muscle Protein Turnover, Particularly in Cancer Cachexia. Cancers 2024, 16, 1921. [Google Scholar] [CrossRef] [PubMed]
  148. Ji, F.; Lee, H.S.; Kim, J.-H. Resistance Exercise and Skeletal Muscle: Protein Synthesis, Degradation, and Controversies. Eur. J. Appl. Physiol. 2025, 125, 2353–2382. [Google Scholar] [CrossRef]
  149. Thomas, A.C.Q.; Stead, C.A.; Burniston, J.G.; Phillips, S.M. Exercise-Specific Adaptations in Human Skeletal Muscle: Molecular Mechanisms of Making Muscles Fit and Mighty. Free Radic. Biol. Med. 2024, 223, 341–356. [Google Scholar] [CrossRef]
  150. Stec, M.J.; Graham, Z.A.; Su, Q.; Adler, C.; Ni, M.; Rouzic, V.L.; Golann, D.R.; Ferrara, P.J.; Halasz, G.; Sleeman, M.W.; et al. Combined Endurance and Resistance Exercise Training Alters the Spatial Transcriptome of Skeletal Muscle in Young Adults. iScience 2025, 28, 113301. [Google Scholar] [CrossRef]
  151. Furrer, R.; Hawley, J.A.; Handschin, C. The Molecular Athlete: Exercise Physiology from Mechanisms to Medals. Physiol. Rev. 2023, 103, 1693–1787. [Google Scholar] [CrossRef]
  152. Wang, J.; Jia, D.; Zhang, Z.; Wang, D. Exerkines and Sarcopenia: Unveiling the Mechanism behind Exercise-Induced Mitochondrial Homeostasis. Metabolites 2025, 15, 59. [Google Scholar] [CrossRef]
  153. Letukienė, A.; Hendrixson, V.; Ginevičienė, V. Current Knowledge and Scientific Trends in Myokines and Exercise Research in the Context of Obesity. Front. Med. 2024, 11, 1421962. [Google Scholar] [CrossRef]
  154. Khemraj, P.; Kuznyetsova, A.; Hood, D.A. Adaptations in Mitochondrial Quality Control and Interactions with Innate Immune Signaling within Skeletal Muscle: A Narrative Review. J. Sport Health Sci. 2025, 101049. [Google Scholar] [CrossRef] [PubMed]
  155. Zhang, H.; Zhang, Y.; Zhang, J.; Jia, D. Exercise Alleviates Cardiovascular Diseases by Improving Mitochondrial Homeostasis. J. Am. Heart Assoc. 2024, 13, e036555. [Google Scholar] [CrossRef] [PubMed]
  156. Dominiak, K.; Galganski, L.; Budzinska, A.; Woyda-Ploszczyca, A.; Zoladz, J.A.; Jarmuszkiewicz, W. Effects of Endurance Training on the Coenzyme Q Redox State in Rat Heart, Liver, and Brain at the Tissue and Mitochondrial Levels: Implications for Reactive Oxygen Species Formation and Respiratory Chain Remodeling. Int. J. Mol. Sci. 2022, 23, 896. [Google Scholar] [CrossRef]
  157. Li, J.; Zhang, S.; Li, C.; Zhang, X.; Shan, Y.; Zhang, Z.; Bo, H.; Zhang, Y. Endurance Exercise-Induced Histone Methylation Modification Involved in Skeletal Muscle Fiber Type Transition and Mitochondrial Biogenesis. Sci. Rep. 2024, 14, 21154. [Google Scholar] [CrossRef]
  158. Gao, T.; Hu, Y.; Zhang, H.; Shi, R.; Song, Y.; Ding, M.; Gao, F. Aerobic Capacity beyond Cardiorespiratory Fitness Linking Mitochondrial Function, Disease Resilience and Healthy Aging. FASEB J. 2025, 39, e70655. [Google Scholar] [CrossRef] [PubMed]
  159. Yuan, L.; Yuan, Y.; Liu, F.; Li, L.; Liu, J.; Chen, Y.; Cheng, J.; Lu, Y. PGC-1α Alleviates Mitochondrial Dysfunction via TFEB-Mediated Autophagy in Cisplatin-Induced Acute Kidney Injury. Aging 2021, 13, 8421–8439. [Google Scholar] [CrossRef]
  160. Craige, S.M.; Mammel, R.K.; Amiri, N.; Willoughby, O.S.; Drake, J.C. Interplay of ROS, Mitochondrial Quality, and Exercise in Aging: Potential Role of Spatially Discrete Signaling. Redox Biol. 2024, 77, 103371. [Google Scholar] [CrossRef]
  161. Memme, J.M.; Slavin, M.; Moradi, N.; Hood, D.A. Mitochondrial Bioenergetics and Turnover during Chronic Muscle Disuse. Int. J. Mol. Sci. 2021, 22, 5179. [Google Scholar] [CrossRef]
  162. Tayebi, S.M.; Poorhabibi, H.; Heidary, D.; Amini, M.A.; Sadeghi, A. Impact of Aerobic Exercise on Chronic Inflammation in Older Adults: A Systematic Review and Meta-Analysis. BMC Sports Sci. Med. Rehabil. 2025, 17, 229. [Google Scholar] [CrossRef] [PubMed]
  163. Magni, O.; Arnaoutis, G.; Panagiotakos, D. The Impact of Exercise on Chronic Systemic Inflammation: A Systematic Review and Meta–Meta-Analysis. Sport. Sci. Health 2025, 21, 1405–1417. [Google Scholar] [CrossRef]
  164. Wang, J.; He, Y.; Wang, Z.; Wang, Z.; Miao, Y.; Choi, J.-Y. Effects of Different Exercise Prescription Parameters on Metabolic and Inflammatory Biomarkers in Cancer Patients: A Systematic Review, Meta-Analysis, and Meta-Regression. Front. Immunol. 2025, 16, 1663560. [Google Scholar] [CrossRef] [PubMed]
  165. YAO, B.; LI, L.; GUAN, X.; ZHU, J.; LIU, Q.; QU, B.; DING, H. Endurance Training Inhibits the JAK2/STAT3 Pathway to Alleviate Sarcopenia. Physiol. Res. 2024, 73, 295–304. [Google Scholar] [CrossRef]
  166. Ducharme, J.B.; Specht, J.W.; Bailly, A.R.; Deyhle, M.R. Serum Cytokines and Their Soluble Receptors Are Differently Regulated between Trained and Untrained Men after Vigorous Endurance Exercise. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2025, 328, R581–R587. [Google Scholar] [CrossRef]
  167. Currier, B.S.; Mcleod, J.C.; Banfield, L.; Beyene, J.; Welton, N.J.; D’Souza, A.C.; Keogh, J.A.J.; Lin, L.; Coletta, G.; Yang, A.; et al. Resistance Training Prescription for Muscle Strength and Hypertrophy in Healthy Adults: A Systematic Review and Bayesian Network Meta-Analysis. Br. J. Sports Med. 2023, 57, 1211–1220. [Google Scholar] [CrossRef]
  168. Ugurlu, D.; Gülü, M.; Yapici, H.; Yagin, F.H.; Comertpay, E.; Eroglu, O.; Afonso, J.; Aldhahi, M.I. Dose-Response Effects of 8-Week Resistance Training on Body Composition and Muscular Performance in Untrained Young Women: A Quasi-Experimental Design. Medicine 2024, 103, e40322. [Google Scholar] [CrossRef]
  169. Mcleod, J.C.; Currier, B.S.; Lowisz, C.V.; Phillips, S.M. The Influence of Resistance Exercise Training Prescription Variables on Skeletal Muscle Mass, Strength, and Physical Function in Healthy Adults: An Umbrella Review. J. Sport. Health Sci. 2024, 13, 47–60. [Google Scholar] [CrossRef]
  170. Abou Sawan, S.; Nunes, E.A.; Lim, C.; McKendry, J.; Phillips, S.M. The Health Benefits of Resistance Exercise: Beyond Hypertrophy and Big Weights. Exerc. Sport Mov. 2023, 1, e00001. [Google Scholar] [CrossRef]
  171. D’Hulst, G.; Masschelein, E.; De Bock, K. Resistance Exercise Enhances Long-Term mTORC1 Sensitivity to Leucine. Mol. Metab. 2022, 66, 101615. [Google Scholar] [CrossRef]
  172. Pietras, P.; Aulas, A.; Fay, M.M.; Leśniczak-Staszak, M.; Sowiński, M.; Lyons, S.M.; Szaflarski, W.; Ivanov, P. Translation Inhibition and Suppression of Stress Granules Formation by Cisplatin. Biomed. Pharmacother. 2022, 145, 112382. [Google Scholar] [CrossRef]
  173. Jiao, L.; Liu, Y.; Yu, X.-Y.; Pan, X.; Zhang, Y.; Tu, J.; Song, Y.-H.; Li, Y. Ribosome Biogenesis in Disease: New Players and Therapeutic Targets. Sig. Transduct. Target. Ther. 2023, 8, 15. [Google Scholar] [CrossRef]
  174. Scarpelli, M.C.; Bergamasco, J.G.A.; Godwin, J.S.; Mesquita, P.H.C.; Chaves, T.S.; Silva, D.G.; Bittencourt, D.; Dias, N.F.; Medalha Junior, R.A.; Carello Filho, P.C.; et al. Resistance Training-Induced Changes in Muscle Proteolysis and Extracellular Matrix Remodeling Biomarkers in the Untrained and Trained States. Eur. J. Appl. Physiol. 2024, 124, 2749–2762. [Google Scholar] [CrossRef] [PubMed]
  175. Michel, J.M.; Godwin, J.S.; Plotkin, D.L.; Mesquita, P.H.C.; McIntosh, M.C.; Ruple, B.A.; Libardi, C.A.; Mobley, C.B.; Kavazis, A.N.; Roberts, M.D. Proteolytic Markers Associated with a Gain and Loss of Leg Muscle Mass with Resistance Training Followed by High-Intensity Interval Training. Exp. Physiol. 2023, 108, 1268–1281. [Google Scholar] [CrossRef]
  176. Moradi, Y.; Zehsaz, F.; Nourazar, M.A. Concurrent Exercise Training and Murf-l and Atrogin-1 Gene Expression in the Vastus Lateralis Muscle of Male Wistar Rats. Apunt. Med. Esport. 2020, 55, 21–27. [Google Scholar] [CrossRef]
  177. Cardiel-Gutiérrez, S.; Villicaña-Gómez, E.A.; Márquez-Gamiño, S.; Vera-Delgado, K.S.; Sotelo-Barroso, F.; Caudillo-Cisneros, C.; Sánchez-Duarte, E. Moderate-Intensity Training Reduces the Expression of Atrogin-1 and Murf-1 in Gastrocnemius Muscle of Diabetic Rats: 1428. Med. Sci. Sports Exerc. 2021, 53, 467. [Google Scholar] [CrossRef]
  178. Chu, Y.; Yuan, X.; Tao, Y.; Yang, B.; Luo, J. Autophagy in Muscle Regeneration: Mechanisms, Targets, and Therapeutic Perspective. Int. J. Mol. Sci. 2024, 25, 11901. [Google Scholar] [CrossRef] [PubMed]
  179. Cao, Y.; Zhou, J.; Quan, H.; Li, W.; Li, T.; Wang, L. Resistance Training Alleviates Muscle Atrophy and Muscle Dysfunction by Reducing Inflammation and Regulating Compromised Autophagy in Aged Skeletal Muscle. Front. Immunol. 2025, 16, 1597222. [Google Scholar] [CrossRef]
  180. Schwendinger, F.; Pocecco, E. Counteracting Physical Inactivity during the COVID-19 Pandemic: Evidence-Based Recommendations for Home-Based Exercise. Int. J. Environ. Res. Public Health 2020, 17, 3909. [Google Scholar] [CrossRef]
  181. Correale, L.; Buzzachera, C.F.; Liberali, G.; Codrons, E.; Mallucci, G.; Vandoni, M.; Montomoli, C.; Bergamaschi, R. Effects of Combined Endurance and Resistance Training in Women with Multiple Sclerosis: A Randomized Controlled Study. Front. Neurol. 2021, 12, 698460. [Google Scholar] [CrossRef] [PubMed]
  182. Li, Z.; Qi, Y.; Chen, X.; Li, J.; Zhang, J.; Li, P.; Zhou, Z. Synergistic Effects of Concurrent Aerobic and Strength Training on Fitness in Children and Adolescents: A Multivariate and Network Meta-Analysis. Scand. J. Med. Sci. Sports 2024, 34, e14764. [Google Scholar] [CrossRef]
  183. Al-Mhanna, S.B.; Batrakoulis, A.; Norhayati, M.N.; Mohamed, M.; Drenowatz, C.; Irekeola, A.A.; Afolabi, H.A.; Gülü, M.; Alkhamees, N.H.; Wan Ghazali, W.S. Combined Aerobic and Resistance Training Improves Body Composition, Alters Cardiometabolic Risk, and Ameliorates Cancer-Related Indicators in Breast Cancer Patients and Survivors with Overweight/Obesity: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J. Sports Sci. Med. 2024, 23, 366–395. [Google Scholar] [CrossRef]
  184. Zhu, C.; Ma, H.; He, A.; Li, Y.; He, C.; Xia, Y. Exercise in Cancer Prevention and Anticancer Therapy: Efficacy, Molecular Mechanisms and Clinical Information. Cancer Lett. 2022, 544, 215814. [Google Scholar] [CrossRef]
  185. Wen, H.; Deng, H.; Li, B.; Chen, J.; Zhu, J.; Zhang, X.; Yoshida, S.; Zhou, Y. Mitochondrial Diseases: From Molecular Mechanisms to Therapeutic Advances. Sig. Transduct. Target. Ther. 2025, 10, 9. [Google Scholar] [CrossRef] [PubMed]
  186. Burtscher, J.; Romani, M.; Bernardo, G.; Popa, T.; Ziviani, E.; Hummel, F.C.; Sorrentino, V.; Millet, G.P. Boosting Mitochondrial Health to Counteract Neurodegeneration. Prog. Neurobiol. 2022, 215, 102289. [Google Scholar] [CrossRef]
  187. Liu, R.; Gao, X.; Wang, L. Network Meta-Analysis of the Intervention Effects of Different Exercise Measures on Sarcopenia in Cancer Patients. BMC Public Health 2024, 24, 1281. [Google Scholar] [CrossRef]
  188. Yen, C.-J.; Hung, C.-H.; Tsai, W.-M.; Cheng, H.-C.; Yang, H.-L.; Lu, Y.-J.; Tsai, K.-L. Effect of Exercise Training on Exercise Tolerance and Level of Oxidative Stress for Head and Neck Cancer Patients Following Chemotherapy. Front. Oncol. 2020, 10, 1536. [Google Scholar] [CrossRef]
  189. Lin, K.-Y.; Cheng, H.-C.; Yen, C.-J.; Hung, C.-H.; Huang, Y.-T.; Yang, H.-L.; Cheng, W.-T.; Tsai, K.-L. Effects of Exercise in Patients Undergoing Chemotherapy for Head and Neck Cancer: A Pilot Randomized Controlled Trial. Int. J. Environ. Res. Public. Health 2021, 18, 1291. [Google Scholar] [CrossRef]
  190. Iglesias, P. Muscle in Endocrinology: From Skeletal Muscle Hormone Regulation to Myokine Secretion and Its Implications in Endocrine–Metabolic Diseases. J. Clin. Med. 2025, 14, 4490. [Google Scholar] [CrossRef] [PubMed]
  191. Wang, H.; He, W.; Chen, P.; Wang, H.; Wang, H.; Zhu, L.; Liu, X. Exerkines and Myokines in Aging Sarcopenia. Front. Endocrinol. 2025, 16, 1592491. [Google Scholar] [CrossRef]
  192. Chow, L.S.; Gerszten, R.E.; Taylor, J.M.; Pedersen, B.K.; van Praag, H.; Trappe, S.; Febbraio, M.A.; Galis, Z.S.; Gao, Y.; Haus, J.M.; et al. Exerkines in Health, Resilience and Disease. Nat. Rev. Endocrinol. 2022, 18, 273–289. [Google Scholar] [CrossRef] [PubMed]
  193. Severinsen, M.C.K.; Pedersen, B.K. Muscle–Organ Crosstalk: The Emerging Roles of Myokines. Endocr. Rev. 2020, 41, 594–609. [Google Scholar] [CrossRef]
  194. Wang, Z.; Li, L.; Yang, M.; Li, B.; Hu, S. From Skeletal Muscle to Myocardium: Molecular Mechanisms of Exercise-Induced Irisin Regulation of Cardiac Fibrosis. Int. J. Mol. Sci. 2025, 26, 3550. [Google Scholar] [CrossRef]
  195. Brown, A.D.; Marko, A.D.; Marko, D.M.; Baranowski, B.J.; Silvera, S.; Finch, M.S.; Yang, A.J.; Dhaliwal, R.; Ryan, C.R.; Roy, B.D.; et al. Brain-Derived Neurotrophic Factor Drives Muscle Adaptation Similar to Aerobic Training in Mice. FASEB J. 2025, 39, e70321. [Google Scholar] [CrossRef] [PubMed]
  196. Ahmadi Hekmatikar, A.; Nelson, A.; Petersen, A. Highlighting the Idea of Exerkines in the Management of Cancer Patients with Cachexia: Novel Insights and a Critical Review. BMC Cancer 2023, 23, 889. [Google Scholar] [CrossRef]
  197. Zhang, B.; Chang, J.Y.; Lee, M.H.; Ju, S.-H.; Yi, H.-S.; Shong, M. Mitochondrial Stress and Mitokines: Therapeutic Perspectives for the Treatment of Metabolic Diseases. Diabetes Metab. J. 2024, 48, 1–18. [Google Scholar] [CrossRef] [PubMed]
  198. Arias-Calderón, M.; Casas, M.; Balanta-Melo, J.; Morales-Jiménez, C.; Hernández, N.; Llanos, P.; Jaimovich, E.; Buvinic, S. Fibroblast Growth Factor 21 Is Expressed and Secreted from Skeletal Muscle Following Electrical Stimulation via Extracellular ATP Activation of the PI3K/Akt/mTOR Signaling Pathway. Front. Endocrinol. 2023, 14, 1059020. [Google Scholar] [CrossRef]
  199. Kilpiö, T.; Skarp, S.; Perjés, Á.; Swan, J.; Kaikkonen, L.; Saarimäki, S.; Szokodi, I.; Penninger, J.M.; Szabó, Z.; Magga, J.; et al. Apelin Regulates Skeletal Muscle Adaptation to Exercise in a High-Intensity Interval Training Model. Am. J. Physiol. Cell Physiol. 2024, 326, C1437–C1450. [Google Scholar] [CrossRef]
  200. Le Moal, E.; Liu, Y.; Collerette-Tremblay, J.; Dumontier, S.; Fabre, P.; Molina, T.; Dort, J.; Orfi, Z.; Denault, N.; Boutin, J.; et al. Apelin Stimulation of the Vascular Skeletal Muscle Stem Cell Niche Enhances Endogenous Repair in Dystrophic Mice. Sci. Transl. Med. 2024, 16, eabn8529. [Google Scholar] [CrossRef]
  201. Walzik, D.; Wences Chirino, T.Y.; Zimmer, P.; Joisten, N. Molecular Insights of Exercise Therapy in Disease Prevention and Treatment. Sig. Transduct. Target. Ther. 2024, 9, 138. [Google Scholar] [CrossRef]
  202. Broniec, M.N.; Norland, K.; Thomas, J.; Wang, X.; Harris, R.A. The Decorin and Myostatin Response to Acute Whole Body Vibration: Impact of Adiposity, Sex, and Race. Int. J. Obes. 2024, 48, 1803–1808. [Google Scholar] [CrossRef] [PubMed]
  203. Baghy, K.; Szakadáti, H.; Kovalszky, I. Decorin the Antifibrotic Proteoglycan and Its Progression in Therapy. Am. J. Physiol.-Cell Physiol. 2025, 328, C1853–C1865. [Google Scholar] [CrossRef]
  204. Lu, X.; Chen, Y.; Shi, Y.; Shi, Y.; Su, X.; Chen, P.; Wu, D.; Shi, H. Exercise and Exerkines: Mechanisms and Roles in Anti-Aging and Disease Prevention. Exp. Gerontol. 2025, 200, 112685. [Google Scholar] [CrossRef]
  205. Petro, J.L.; Gallo-Villegas, J.; Calderón, J.C. Myonectin and Metabolic Health: A Systematic Review. Front. Endocrinol. 2025, 16, 1557142. [Google Scholar] [CrossRef]
  206. Saw, E.L.; Werner, L.D.; Cooper, H.L.; Pimental, D.R.; Zamani, P.; Chirinos, J.A.; Valero-Muñoz, M.; Sam, F. Musclin Counteracts Skeletal Muscle Dysfunction and Exercise Intolerance in Heart Failure with Preserved Ejection Fraction. Circ. Heart Fail. 2025, 18, e012350. [Google Scholar] [CrossRef] [PubMed]
  207. Qi, C.; Song, X.; Wang, H.; Yan, Y.; Liu, B. The Role of Exercise-Induced Myokines in Promoting Angiogenesis. Front. Physiol. 2022, 13, 981577. [Google Scholar] [CrossRef] [PubMed]
  208. Bucarey, J.L.; Trujillo-González, I.; Paules, E.M.; Espinosa, A. Myokines and Their Potential Protective Role against Oxidative Stress in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Antioxidants 2024, 13, 1363. [Google Scholar] [CrossRef]
  209. Rathor, R.; Suryakumar, G. Myokines: A Central Point in Managing Redox Homeostasis and Quality of Life. BioFactors 2024, 50, 885–909. [Google Scholar] [CrossRef]
  210. Uurasmaa, T.-M.; Ricardo, C.; Autio, A.; Heinonen, I.H.A.; Rundqvist, H.; Anttila, K. Voluntary Wheel Running Reduces Tumor Growth and Increases Capillarity in the Heart during Doxorubicin Chemotherapy in a Murine Model of Breast Cancer. Front. Physiol. 2024, 15, 1347347. [Google Scholar] [CrossRef]
  211. Halle, J.L.; Counts, B.R.; Zhang, Q.; James, K.M.; Puppa, M.J.; Alway, S.E.; Carson, J.A. Mouse Skeletal Muscle Adaptations to Different Durations of Treadmill Exercise after the Cessation of FOLFOX Chemotherapy. Front. Physiol. 2023, 14, 1283674. [Google Scholar] [CrossRef]
  212. Nguyen, B.L.; Baumfalk, D.R.; Lapierre-Nguyen, S.S.; Zhong, R.; Doerr, V.; Montalvo, R.N.; Wei-LaPierre, L.; Smuder, A.J. Effects of Exercise and Doxorubicin on Acute Diaphragm Neuromuscular Transmission Failure. Exp. Neurol. 2024, 378, 114818. [Google Scholar] [CrossRef]
  213. Tamayo-Torres, E.; Garrido, A.; de Cabo, R.; Carretero, J.; Gómez-Cabrera, M.C. Molecular Mechanisms of Cancer Cachexia. Role of Exercise Training. Mol. Asp. Med. 2024, 99, 101293. [Google Scholar] [CrossRef] [PubMed]
  214. Frühwein, H.; Paul, N.W. “Lost in Translation?” Animal Research in the Era of Precision Medicine. J. Transl. Med. 2025, 23, 152. [Google Scholar] [CrossRef] [PubMed]
  215. Ineichen, B.V.; Furrer, E.; Grüninger, S.L.; Zürrer, W.E.; Macleod, M.R. Analysis of Animal-to-Human Translation Shows That Only 5% of Animal-Tested Therapeutic Interventions Obtain Regulatory Approval for Human Applications. PLoS Biol. 2024, 22, e3002667. [Google Scholar] [CrossRef]
  216. Berg, I.; Härvelid, P.; Zürrer, W.E.; Rosso, M.; Reich, D.S.; Ineichen, B.V. Which Experimental Factors Govern Successful Animal-to-Human Translation in Multiple Sclerosis Drug Development? A Systematic Review and Meta-Analysis. eBioMedicine 2024, 110, 105434. [Google Scholar] [CrossRef]
  217. Bishop, D.J.; Lee, M.J.-C.; Picard, M. Exercise as Mitochondrial Medicine: How Does the Exercise Prescription Affect Mitochondrial Adaptations to Training? Annu. Rev. Physiol. 2025, 87, 107–129. [Google Scholar] [CrossRef]
  218. Fairman, C.M.; Lønbro, S.; Cardaci, T.D.; VanderVeen, B.N.; Nilsen, T.S.; Murphy, A.E. Muscle Wasting in Cancer: Opportunities and Challenges for Exercise in Clinical Cancer Trials. JCSM Rapid Commun. 2022, 5, 52–67. [Google Scholar] [CrossRef] [PubMed]
  219. Tsitkanou, S.; Murach, K.A.; Washington, T.A.; Greene, N.P. Exercise Counteracts the Deleterious Effects of Cancer Cachexia. Cancers 2022, 14, 2512. [Google Scholar] [CrossRef]
  220. Kearney, N.; Connolly, D.; Begic, S.; Mockler, D.; Guinan, E. Feasibility Metrics of Exercise Interventions during Chemotherapy: A Systematic Review. Crit. Rev. Oncol. Hematol. 2024, 195, 104272. [Google Scholar] [CrossRef]
  221. Perše, M. Cisplatin Mouse Models: Treatment, Toxicity and Translatability. Biomedicines 2021, 9, 1406. [Google Scholar] [CrossRef]
  222. Amarelo, A.; Mota, M.; Amarelo, B.; Ferreira, M.C.; Fernandes, C.S. Technological Resources for Physical Rehabilitation in Cancer Patients Undergoing Chemotherapy: A Scoping Review. Cancers 2024, 16, 3949. [Google Scholar] [CrossRef]
  223. Underwood, W.P.; Michalski, M.G.; Lee, C.P.; Fickera, G.A.; Chun, S.S.; Eng, S.E.; Liu, L.Y.; Tsai, B.L.; Moskowitz, C.S.; Lavery, J.A.; et al. A Digital, Decentralized Trial of Exercise Therapy in Patients with Cancer. npj Digit. Med. 2024, 7, 304. [Google Scholar] [CrossRef] [PubMed]
  224. Shaffer, K.M.; Turner, K.L.; Siwik, C.; Gonzalez, B.D.; Upasani, R.; Glazer, J.V.; Ferguson, R.J.; Joshua, C.; Low, C.A. Digital Health and Telehealth in Cancer Care: A Scoping Review of Reviews. Lancet Digit. Health 2023, 5, e316–e327. [Google Scholar] [CrossRef] [PubMed]
  225. De Vleeschauwer, S.I.; van de Ven, M.; Oudin, A.; Debusschere, K.; Connor, K.; Byrne, A.T.; Ram, D.; Rhebergen, A.M.; Raeves, Y.D.; Dahlhoff, M.; et al. OBSERVE: Guidelines for the Refinement of Rodent Cancer Models. Nat. Protoc. 2024, 19, 2571–2596. [Google Scholar] [CrossRef]
  226. Delphan, M.; Delfan, N.; West, D.; Delfan, M. Exercise Protocols: The Gap between Preclinical and Clinical Exercise Oncology Studies. Metab. Open 2022, 13, 100165. [Google Scholar] [CrossRef]
  227. Avancini, A.; Borsati, A.; Toniolo, L.; Ciurnelli, C.; Belluomini, L.; Budolfsen, T.; Lillelund, C.; Milella, M.; Quist, M.; Pilotto, S. Physical Activity Guidelines in Oncology: A Systematic Review of the Current Recommendations. Crit. Rev. Oncol./Hematol. 2025, 210, 104718. [Google Scholar] [CrossRef]
  228. Saleh, A.Y.; Shareef, A.; Bishoyi, A.K.; Jyothi, S.R.; Panigrahi, R.; Pargaien, A.; Garg, G.; Hafizova, M.; Sameer, H.N.; Yaseen, A.; et al. Personalized Exercise Programs in Oncology. Oncol. Rev. 2025, 19, 1645505. [Google Scholar] [CrossRef] [PubMed]
  229. Novinger, L.J.; Weinzierl, N.M.; Bonetto, A. Diversity in Chemotherapy-Induced Cachexia. Am. J. Physiol. Cell Physiol. 2025, 328, C139–C147. [Google Scholar] [CrossRef]
  230. Park, S.H.; Ko, J.R.; Han, J. Exercise Alleviates Cisplatin-Induced Toxicity in the Hippocampus of Mice by Inhibiting Neuroinflammation and Improving Synaptic Plasticity. Korean J. Physiol. Pharmacol. 2024, 28, 145–152. [Google Scholar] [CrossRef]
  231. Collao, N.; Sanders, O.; Caminiti, T.; Messeiller, L.; De Lisio, M. Resistance and Endurance Exercise Training Improves Muscle Mass and the Inflammatory/Fibrotic Transcriptome in a Rhabdomyosarcoma Model. J. Cachexia Sarcopenia Muscle 2023, 14, 781–793. [Google Scholar] [CrossRef]
  232. Tuğral, A.; Arıbaş, Z.; Kaya Uçar, G.; Arslan, F.D.; Bakar, Y.; Karakoyun, I.; Akyol, M. The Effect of Supervised Aerobic Exercise on Adipokine and Myokine Biomarkers in Patients with Cancer during Systemic Chemotherapy: A Single-Blinded Prospective Controlled Trial. Support. Care Cancer 2025, 33, 741. [Google Scholar] [CrossRef] [PubMed]
  233. Wilkinson, D.J.; Crossland, H. Metabolomic and Proteomic Applications to Exercise Biomedicine. Transl. Exerc. Biomed. 2024, 1, 9–22. [Google Scholar] [CrossRef]
  234. Wang, Y.; Li, X.; Li, N.; Du, J.; Qin, X.; Sun, X.; Wang, Y.; Li, C. Integrated Proteomic and Metabolomic Analysis of Muscle Atrophy Induced by Hindlimb Unloading. Biomolecules 2024, 15, 14. [Google Scholar] [CrossRef]
  235. Murach, K.A.; Peterson, C.A. A Muscle Exercise Research Revolution Powered by -Omics at Single Cell and Nucleus Resolution. BMC Biol. 2023, 21, 298. [Google Scholar] [CrossRef]
  236. Yin, L.; Wu, S.; Bai, P.; Wang, X. Combination of Transcriptomics and Proteomics for Analyzing Potential Biomarker and Molecular Mechanism Underlying Skeletal Muscle Atrophy. J. Proteom. 2024, 309, 105283. [Google Scholar] [CrossRef]
  237. Li, X.; Fang, L.; Zhou, R.; Yao, L.; Clayton, S.W.; Muscat, S.; Kamm, D.R.; Wang, C.; Liu, C.-J.; Qin, L.; et al. Current Cutting-Edge Omics Techniques on Musculoskeletal Tissues and Diseases. Bone Res. 2025, 13, 59. [Google Scholar] [CrossRef]
  238. Moreno-Justicia, R.; Van der Stede, T.; Stocks, B.; Laitila, J.; Seaborne, R.A.; Van de Loock, A.; Lievens, E.; Samodova, D.; Marín-Arraiza, L.; Dmytriyeva, O.; et al. Human Skeletal Muscle Fiber Heterogeneity beyond Myosin Heavy Chains. Nat. Commun. 2025, 16, 1764. [Google Scholar] [CrossRef]
  239. Aquila, G.; Re Cecconi, A.D.; Brault, J.J.; Corli, O.; Piccirillo, R. Nutraceuticals and Exercise against Muscle Wasting during Cancer Cachexia. Cells 2020, 9, 2536. [Google Scholar] [CrossRef] [PubMed]
  240. Beaudry, A.G.; Law, M.L. Leucine Supplementation in Cancer Cachexia: Mechanisms and a Review of the Pre-Clinical Literature. Nutrients 2022, 14, 2824. [Google Scholar] [CrossRef] [PubMed]
  241. Woldeselassie, M.; Tamene, A. Therapeutic Controversies over Use of Antioxidant Supplements during Cancer Treatment: A Scoping Review. Front. Nutr. 2024, 11, 1480780. [Google Scholar] [CrossRef]
  242. Pfister, C.; Schoenemann, J. Selenium in Cancer Rehabilitation—A Retrospective Study from a Specialized Clinic. Nutrients 2023, 15, 3827. [Google Scholar] [CrossRef]
  243. Sugimoto, R.; Lee, L.; Tanaka, Y.; Morita, Y.; Hijioka, M.; Hisano, T.; Furukawa, M. Zinc Deficiency as a General Feature of Cancer: A Review of the Literature. Biol. Trace Elem. Res. 2024, 202, 1937–1947. [Google Scholar] [CrossRef] [PubMed]
  244. Zhou, J.; Nie, R.; Yin, Y.; Cai, X.; Xie, D.; Cai, M. Protective Effect of Natural Antioxidants on Reducing Cisplatin-Induced Nephrotoxicity. Dis. Markers 2022, 2022, 1612348. [Google Scholar] [CrossRef] [PubMed]
  245. Arends, J.; Strasser, F.; Gonella, S.; Solheim, T.S.; Madeddu, C.; Ravasco, P.; Buonaccorso, L.; de van der Schueren, M.A.E.; Baldwin, C.; Chasen, M.; et al. Cancer Cachexia in Adult Patients: ESMO Clinical Practice Guidelines. Esmo Open 2021, 6, 100092. [Google Scholar] [CrossRef] [PubMed]
  246. Feng, Y.; Feng, X.; Wan, R.; Luo, Z.; Qu, L.; Wang, Q. Impact of Exercise on Cancer: Mechanistic Perspectives and New Insights. Front. Immunol. 2024, 15, 1474770. [Google Scholar] [CrossRef]
Figure 1. Literature selection flow diagram.
Figure 1. Literature selection flow diagram.
Biomolecules 15 01495 g001
Figure 2. Overview of molecular mechanisms underlying cisplatin-induced muscle atrophy. Cisplatin induces mitochondrial dysfunction, endoplasmic reticulum stress, and inflammatory responses, which activate the ubiquitin–proteasome system and autophagy, thereby enhancing protein degradation. At the same time, cisplatin suppresses the Akt/mTOR signaling pathway, leading to reduced protein synthesis. The imbalance between protein degradation and synthesis ultimately results in muscle atrophy. In the figure, “↑” denotes upregulation or activation, “↓” indicates downregulation or suppression, and “⊥” represents inhibitory regulation. ROS, reactive oxygen species; ER, endoplasmic reticulum; UPS, ubiquitin–proteasome system; LC3-II, microtubule-associated protein 1 light chain 3-II; mTOR, mechanistic target of rapamycin; Akt, protein kinase B; NF-κB, nuclear factor kappa B; STAT3, signal transducer and activator of transcription 3; FoxO, forkhead box O; TNF-α, tumor necrosis factor alpha; IL-6, interleukin-6; IFN-γ, interferon gamma.
Figure 2. Overview of molecular mechanisms underlying cisplatin-induced muscle atrophy. Cisplatin induces mitochondrial dysfunction, endoplasmic reticulum stress, and inflammatory responses, which activate the ubiquitin–proteasome system and autophagy, thereby enhancing protein degradation. At the same time, cisplatin suppresses the Akt/mTOR signaling pathway, leading to reduced protein synthesis. The imbalance between protein degradation and synthesis ultimately results in muscle atrophy. In the figure, “↑” denotes upregulation or activation, “↓” indicates downregulation or suppression, and “⊥” represents inhibitory regulation. ROS, reactive oxygen species; ER, endoplasmic reticulum; UPS, ubiquitin–proteasome system; LC3-II, microtubule-associated protein 1 light chain 3-II; mTOR, mechanistic target of rapamycin; Akt, protein kinase B; NF-κB, nuclear factor kappa B; STAT3, signal transducer and activator of transcription 3; FoxO, forkhead box O; TNF-α, tumor necrosis factor alpha; IL-6, interleukin-6; IFN-γ, interferon gamma.
Biomolecules 15 01495 g002
Figure 3. Protective effects of exercise intervention against cisplatin-induced muscle atrophy. Endurance exercise enhances mitochondrial biogenesis via PGC-1α, reduces ROS and inflammation, and restores metabolic balance. Resistance exercise activates Akt/mTOR signaling, promotes protein synthesis, inhibits degradation and excessive autophagy, leading to anabolic dominance. Combined exercise exerts synergistic benefits by integrating metabolic restoration and anabolic signaling. Together, these effects preserve muscle mass and attenuate cisplatin-induced atrophy. Arrows indicate regulation: ↑ upregulation/activation; ↓ downregulation/inhibition. Blue lines represent endurance pathways, green lines resistance pathways, and purple dashed lines synergistic interactions. PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1-alpha; ROS, reactive oxygen species; Akt, protein kinase B; mTOR, mechanistic target of rapamycin.
Figure 3. Protective effects of exercise intervention against cisplatin-induced muscle atrophy. Endurance exercise enhances mitochondrial biogenesis via PGC-1α, reduces ROS and inflammation, and restores metabolic balance. Resistance exercise activates Akt/mTOR signaling, promotes protein synthesis, inhibits degradation and excessive autophagy, leading to anabolic dominance. Combined exercise exerts synergistic benefits by integrating metabolic restoration and anabolic signaling. Together, these effects preserve muscle mass and attenuate cisplatin-induced atrophy. Arrows indicate regulation: ↑ upregulation/activation; ↓ downregulation/inhibition. Blue lines represent endurance pathways, green lines resistance pathways, and purple dashed lines synergistic interactions. PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1-alpha; ROS, reactive oxygen species; Akt, protein kinase B; mTOR, mechanistic target of rapamycin.
Biomolecules 15 01495 g003
Figure 4. Role of exercise-induced myokines in counteracting cisplatin-induced muscle atrophy. The schematic illustrates how exercise promotes the release of multiple myokines from skeletal muscle, including Irisin, IL-15, BDNF, FGF21, Apelin, Decorin, CTRP15 (Myonectin), and Musclin. These myokines exert distinct biological effects by regulating mitochondrial biogenesis, protein turnover, metabolic flexibility, angiogenesis, and regenerative processes. Downstream, they converge on six protective mechanisms: inflammation and oxidative stress, protein synthesis, protein degradation, regeneration, angiogenesis, and metabolic flexibility, ultimately leading to reduced cisplatin-induced muscle atrophy and preserved muscle mass and function. ↑ indicate stimulation or increase; ↓ indicate reduction or suppression; ⊣ represent inhibition of signaling pathways. IL-15, interleukin-15; BDNF, brain-derived neurotrophic factor; FGF21, fibroblast growth factor 21; CTRP15, C1q/TNF-related protein 15 (Myonectin); AMPK, AMP-activated protein kinase; PGC-1α, peroxisome proliferator-activated receptor gamma coactivator-1α; TGF-β, transforming growth factor-beta.
Figure 4. Role of exercise-induced myokines in counteracting cisplatin-induced muscle atrophy. The schematic illustrates how exercise promotes the release of multiple myokines from skeletal muscle, including Irisin, IL-15, BDNF, FGF21, Apelin, Decorin, CTRP15 (Myonectin), and Musclin. These myokines exert distinct biological effects by regulating mitochondrial biogenesis, protein turnover, metabolic flexibility, angiogenesis, and regenerative processes. Downstream, they converge on six protective mechanisms: inflammation and oxidative stress, protein synthesis, protein degradation, regeneration, angiogenesis, and metabolic flexibility, ultimately leading to reduced cisplatin-induced muscle atrophy and preserved muscle mass and function. ↑ indicate stimulation or increase; ↓ indicate reduction or suppression; ⊣ represent inhibition of signaling pathways. IL-15, interleukin-15; BDNF, brain-derived neurotrophic factor; FGF21, fibroblast growth factor 21; CTRP15, C1q/TNF-related protein 15 (Myonectin); AMPK, AMP-activated protein kinase; PGC-1α, peroxisome proliferator-activated receptor gamma coactivator-1α; TGF-β, transforming growth factor-beta.
Biomolecules 15 01495 g004
Table 1. Comparative features of cisplatin-induced versus other types of skeletal muscle atrophy.
Table 1. Comparative features of cisplatin-induced versus other types of skeletal muscle atrophy.
Feature/MechanismCisplatin-Induced AtrophyDisuse/Denervation AtrophyCancer Cachexia/Systemic AtrophyReferences
Primary causeChemotherapy toxicity and systemic stressMechanical unloading or loss of neural inputChronic inflammation and tumor–host interaction[16,20]
Mitochondrial effectDirect mtDNA damage and oxidative stress via cisplatin–DNA adductsReduced mitochondrial activity due to inactivityImpaired mitochondrial biogenesis secondary to inflammation[1,77,86]
Inflammatory cytokinesMarked systemic IL-6 and TNF-α elevation following chemotherapyMild local inflammationSevere systemic inflammation (IL-1β, IL-6, TNF-α)[16,18]
Myokine regulationDecreased secretion of IL-15 and irisin; imbalance in myostatin signalingSlightly reduced myokine production due to inactivityAltered myokine profile driven by tumor–host crosstalk[89,90]
PGC-1α and mitochondrial biogenesisMarkedly suppressed, leading to energy deficit and impaired regenerationReduced but recoverable with reloadingSuppressed by chronic inflammatory signaling[1,86]
Dominant mechanismDual mechanism: direct cytotoxic injury and systemic metabolic dysregulationLocal disuse-induced proteolysisSystemic inflammation–driven catabolism[1,20]
ReversibilityPartially reversible with exercise or nutritional interventionsFully reversible with reloadingPoorly reversible without treating the underlying disease[75,88,91]
Note: IL, interleukin; TNF-α, tumor necrosis factor alpha; mtDNA, mitochondrial DNA; ROS, reactive oxygen species; PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1-alpha.
Table 2. Clinical and preclinical evidence of cisplatin-induced muscle atrophy.
Table 2. Clinical and preclinical evidence of cisplatin-induced muscle atrophy.
Study TypeSubjectsKey FindingsReferences
ClinicalHead and Neck Cancer PatientsCisplatin chemotherapy results in significant muscle mass loss and muscle atrophy symptoms.[1]
ClinicalLung Cancer PatientsCisplatin treatment leads to substantial muscle mass and strength decline, impacting quality of life.[6]
ClinicalOvarian Cancer PatientsCisplatin treatment decreases muscle index, resulting in impaired physical function.[53]
PreclinicalMouse ModelCisplatin induces muscle mass reduction, with a significant decrease in muscle fiber cross-sectional area.[7]
PreclinicalMouse ModelCisplatin causes a decrease in muscle protein synthesis, accompanied by activation of the protein degradation system.[16]
PreclinicalMouse ModelCisplatin induces muscle atrophy with increased oxidative stress and inflammation.[18]
Table 3. Challenges and future directions for clinical implementation of exercise interventions.
Table 3. Challenges and future directions for clinical implementation of exercise interventions.
ChallengesFuture DirectionsReferences
Patient heterogeneityDevelop personalized exercise prescriptions tailored to cancer type, treatment regimen, baseline physical status, and genetic background.[220,221,222]
Treatment side effectsFocus on understanding how different exercise modalities can reduce treatment-related muscle wasting and improve overall health.[9,10,34]
Adherence to exercise programsIntegrate digital health tools, such as wearables and tele-rehabilitation, to enhance feasibility and patient compliance.[220,223,224]
Lack of standardized exercise protocolsStandardize exercise protocols and establish reproducible interventions in preclinical and clinical research.[220,225,226]
Limited clinical trials on exercise interventionsConduct large-scale clinical trials to validate the efficacy of exercise interventions during cisplatin-based chemotherapy.[189,219,220]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Xu, M.; Liu, X. Cisplatin-Induced Skeletal Muscle Atrophy: Biomolecular Mechanisms and the Protective Role of Exercise-Induced Myokines. Biomolecules 2025, 15, 1495. https://doi.org/10.3390/biom15111495

AMA Style

Xu M, Liu X. Cisplatin-Induced Skeletal Muscle Atrophy: Biomolecular Mechanisms and the Protective Role of Exercise-Induced Myokines. Biomolecules. 2025; 15(11):1495. https://doi.org/10.3390/biom15111495

Chicago/Turabian Style

Xu, Miaomiao, and Xiaoguang Liu. 2025. "Cisplatin-Induced Skeletal Muscle Atrophy: Biomolecular Mechanisms and the Protective Role of Exercise-Induced Myokines" Biomolecules 15, no. 11: 1495. https://doi.org/10.3390/biom15111495

APA Style

Xu, M., & Liu, X. (2025). Cisplatin-Induced Skeletal Muscle Atrophy: Biomolecular Mechanisms and the Protective Role of Exercise-Induced Myokines. Biomolecules, 15(11), 1495. https://doi.org/10.3390/biom15111495

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop