Editorial Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. May 28, 2025; 31(20): 105346
Published online May 28, 2025. doi: 10.3748/wjg.v31.i20.105346
Metabolic dysfunction-associated steatotic liver disease: A story of muscle and mass
Vasily Isakov, Department of Gastroenterology and Hepatology, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow 115446, Russia
ORCID number: Vasily Isakov (0000-0002-4417-8076).
Author contributions: Isakov V wrote and edited the manuscript and reviewed the literature, read and approved the final version of the manuscript to be published.
Supported by Russian Science Foundation, No. 19-76-30014.
Conflict-of-interest statement: Isakov V has nothing to disclose.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Vasily Isakov, MD, PhD, Professor, Department of Gastroenterology and Hepatology, Federal Research Center of Nutrition, Biotechnology and Food Safety, 21 Kashirskoye Shosse, Moscow 115446, Russia. vasily.isakov@gmail.com
Received: January 20, 2025
Revised: April 13, 2025
Accepted: May 9, 2025
Published online: May 28, 2025
Processing time: 129 Days and 14.3 Hours

Abstract

Skeletal muscle alterations (SMA) are increasingly recognized as both contributors and consequences of metabolic dysfunction-associated steatotic liver disease (MASLD), affecting disease progression and outcomes. Sarcopenia is common in patients with MASLD, with a prevalence ranging from 20% to 40% depending on the population and diagnostic criteria used. In advanced stages, such as metabolic dysfunction-associated steatohepatitis and fibrosis, its prevalence is even higher. Sarcopenia exacerbates insulin resistance, systemic inflammation, and oxidative stress, all of which worsen MASLD. It is an independent risk factor for fibrosis progression and poor outcomes including mortality. Myosteatosis refers to the abnormal accumulation of fat within muscle tissue, leading to decreased muscle quality. Myosteatosis is prevalent (> 30%) in patients with MASLD, especially those with obesity or type 2 diabetes, although this can vary with the imaging techniques used. It reduces muscle strength and metabolic efficiency, further contributing to insulin resistance and is usually associated with advanced liver disease, cardiovascular complications, and lower levels of physical activity. Altered muscle metabolism, which includes mitochondrial dysfunction and impaired amino acid metabolism, has been reported in metabolic syndromes, including MASLD, although its actual prevalence is unknown. Altered muscle metabolism limits glucose uptake and oxidation, worsening hyperglycemia and lipotoxicity. Reduced muscle perfusion and oxygenation due to endothelial dysfunction and systemic metabolic alterations are common in MASLD associated with comorbidities, such as obesity, hypertension, and atherosclerosis. It decreases the muscle capacity for aerobic metabolism, leading to fatigue and reduced physical activity in patients with MASLD, aggravating metabolic dysfunction. Various SMA in MASLD worsen insulin resistance and hepatic fat accumulation, may accelerate progression to fibrosis and cirrhosis, and increase the risk of cardiovascular disease and mortality. Management strategies for SMA include resistance training, aerobic exercise, and nutritional support (e.g., high-protein diets, vitamin D, and omega-3 fatty acids), which are essential for mitigating skeletal muscle loss and improving outcomes. However, pharmacological agents that target the muscle and liver (such as glucagon-like peptide-1 receptor agonists) show promise but have not yet been approved for the treatment of MASLD.

Key Words: Metabolic dysfunction-associated steatotic liver disease; Skeletal muscle alterations; Sarcopenia; Skeletal muscle mass index; Myosteatosis; Dietary patterns; High-protein diet; Physical activity; Glucagon-like peptide-1 receptor agonists

Core Tip: Skeletal muscle alterations, such as sarcopenia, myosteatosis, and altered muscle metabolism, are highly prevalent in metabolic dysfunction-associated steatotic liver disease (MASLD) and are increasingly recognized as both contributors and consequences of MASLD, affecting disease progression and outcomes. Common treatment approaches for both conditions include nutritional interventions and physical activity/exercise aimed at increasing insulin sensitivity and reducing fat mass but maintaining muscle mass and function. Pharmacological agents that target the muscle and liver (such as glucagon-like peptide-1 receptor agonists) show promise, but have not yet been approved for the treatment of MASLD.



INTRODUCTION

Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease, is characterized by the accumulation of fat in the liver (hepatic steatosis) in individuals with metabolic risk factors such as obesity, type 2 diabetes, metabolic syndrome, and minimal or no alcohol consumption[1,2]. According to the new nomenclature the term MASLD comprises different conditions, including isolated liver steatosis [metabolic dysfunction-associated steatotic liver (MASL)], metabolic dysfunction-associated steatohepatitis (MASH), as well as fibrosis and cirrhosis. Prevalence of MASL is increasing worldwide as well as of progressive form of the disease-MASH, which involves liver inflammation leading to fibrosis, cirrhosis, or hepatocellular carcinoma (HCC)[3,4]. Skeletal muscle alterations (SMA) are increasingly recognized as both contributors and consequences of MASLD, affecting disease progression and outcomes.

SARCOPENIA

Sarcopenia, characterized by the loss of skeletal muscle mass and function, is considered a common comorbidity in patients with MASLD, affecting both the progression of liver disease and overall health outcomes. Sarcopenia was described in 1988 by Rosenberg[5] as a loss of muscle mass related to ageing. However, recent guidelines separate primary sarcopenia, which is defined as age-related muscle decline in which no causes other than aging itself can be indicated, and secondary sarcopenia as the condition due to other detectable causes[6]. The latter is associated with MASLD. The prevalence of sarcopenia in patients with MASLD can be substantial and influenced by various factors, including age, sex, the severity of liver disease[7], and estimates suggest that between 25% and 70% of individuals with chronic liver disease exhibit signs of sarcopenia[8]. This prevalence is particularly pronounced in non-obese patients who may experience "hidden obesity", where muscle loss occurs despite normal body weight[9]. Recent meta-analyses showed that the prevalence of sarcopenia was higher in the MASLD group, with odds ratios (ORs) varied from 1.02 to 2.08, which is more prevalent in Asian patients and in males[7,10,11]. Results of the recent study evaluating the prevalence of SMA in 62 Spanish MASLD patients showed no significant sarcopenia and only a trend of decline in physical performance in patients with advanced stages of liver disease[12]. Despite the low number of patients, low prevalence of advanced hepatic fibrosis in the cohort, and the use of less sensitive bioelectrical impedance to measure appendicular skeletal muscle mass, this study stresses the ethnical/geographical differences in the prevalence of sarcopenia in MASLD patients.

According to one meta-analysis, sarcopenia was strongly associated with MASLD progression and markedly correlated with MASLD-associated mortality[11]. A follow-up study over 23 years showed that sarcopenia was associated with increased all-cause mortality only in individuals with MASLD, whereas this association was absent in those without MASLD. Individuals with both sarcopenia and MASLD had a higher risk of all-cause mortality [hazard ratio (HR) = 1.28, 95%CI: 1.06-1.55] compared with those without sarcopenia and MASLD[13]. The association between sarcopenia and MASLD is complex and multifactorial, involving shared pathophysiological mechanisms, including insulin resistance, inflammation, and metabolic dysregulation[14-16]. It is well established that insulin resistance not only contributes to the development of MASLD but also exacerbates muscle loss by reducing protein synthesis, promoting catabolism[17] and also changes in myokines, which are critical for muscle metabolism[18,19]. Furthermore, elevated levels of pro-inflammatory cytokines such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) are frequently observed in MASLD and are known to promote muscle degradation[20,21]. This inflammatory milieu may lead to a vicious cycle in which sarcopenia worsens liver function, further enhancing insulin resistance and inflammation[22]. Vitamin D deficiency has also been implicated in the association between sarcopenia and MASLD. Low vitamin D levels are common in patients with MASLD and are associated with accelerated loss of muscle mass[23]. This deficiency may exacerbate the risk of sarcopenia, highlighting the importance of nutritional factors in managing MASLD and its complications[16]. It was shown that, the presence of sarcopenia in patients with MASLD has been linked to increased severity of liver fibrosis, which is a critical determinant of long-term outcomes in these patients[22,24-26]. Studies have shown that sarcopenia is associated with increased liver fibrosis and a higher risk of adverse outcomes including mortality[25].

Sarcopenia is not only a consequence of MASLD but may also contribute to its risk and progression. Several meta-analyses have estimated the risk of MASLD among patients with sarcopenia and showed that individuals with sarcopenia are at a higher risk of developing MASLD[27,28]. This relationship is underscored by findings suggesting that lower skeletal muscle mass is an independent predictor of MASLD, regardless of other metabolic risk factors[28,29]. The contribution of relative muscle mass in the development of MASLD was demonstrated during the 7-year longitudinal cohort study in which 1864 (14.8%) of the 12624 subjects without the disease at baseline developed MASLD. Cox proportional hazard analysis of skeletal muscle mass index (SMI) estimated by bioelectrical impedance analysis during the follow-up showed that the highest SMI tertile was inversely associated with incident MASLD [adjusted HR (AHR) = 0.44, 95%CI: 0.38-0.51] and positively associated with the resolution of baseline MASLD (AHR = 2.09, 95%CI: 1.02-4.28) in comparison with the lowest sex-specific SMI tertile at baseline. Furthermore, compared with the lowest tertile of change in SMI over a year, the highest tertile exhibited a significant beneficial association with incident MASLD and resolution of baseline MASLD even after adjustment for many covariates, including age, sex, waist circumference, diabetes status, hypertension, smoking, regular exercise, and baseline SMI[30]. The results of this study demonstrated that the increase in relative skeletal muscle mass over time may have a protective effect against MASLD and support the concept that physical activity, which increases muscle mass and prevents sarcopenia, is one of the pillars in the treatment of the disease.

MYOSTEATOSIS

Myosteatosis, characterized by the excessive accumulation of fat within skeletal muscle, has emerged as a significant pathological condition linked to various metabolic disorders, including MASLD. The diagnostic criteria for myosteatosis remain underdeveloped, but indicators such as skeletal muscle density and intramuscular adipose tissue are commonly used[31]. The lack of standardized diagnostic criteria complicates the assessment of myosteatosis in clinical practice, particularly in the context of MASLD. Research indicates that myosteatosis is frequently observed in patients with MASLD (from 30% to 50%), highlighting its potential role as a biomarker for liver disease progression and overall metabolic health[32-34]. In a large cohort study of 18154 participants the multivariable-adjusted OR for myosteatosis was elevated in the MASLD group (OR = 1.75, 95%CI: 1.52–2.02), while it was slightly decreased in subjects with cryptogenic steatotic liver disease (OR = 0.52, 95%CI: 0.29–0.95), in whom steatosis was found by abdominal ultrasound, no other causes of steatosis, like alcohol, metabolic factor, viruses or drug use were revealed[35]. These data emphasize that myosteatosis shares a common pathogenesis with MASLD, but not with liver steatosis itself. The prevalence of myosteatosis in advanced liver disease such as liver cirrhosis is even higher (up to 76%-82%)[34,36,37], however it may be related to liver insufficiency independent of the aetiology of liver disease itself and due to excessive amounts of ammonia delivered to the muscle, which led to the loss of muscle quality and quantity, as in the majority of the studies, mixed groups of patients with cirrhosis were studied. However, it was shown that myosteatosis is more common in patients with MASLD-associated HCC compared than in those with viral-associated HCC, suggesting that the underlying liver disease significantly influences the presence of myosteatosis even in end-stage liver disease[38].

Myosteatosis is prevalent even in non-cirrhotic stages of MASLD and has been linked to poor clinical outcomes, including increased all-cause mortality and cardiovascular events[33]. Myosteatosis is particularly concerning as it can exacerbate the metabolic dysfunction associated with MASLD, leading to a higher risk of developing MASH and liver fibrosis than the loss of muscle mass itself[39,40]. The accumulation of fat in the skeletal muscle, as indicated by lower muscle radiodensity on computed tomography, serves as a significant prognostic indicator for the progression of simple liver steatosis to MASH[41]. However, there are interesting sexual differences in the roles of sarcopenia and miosteatosis in the progression of MASLD. Thus, lower skeletal muscle mass combined with abdominal obesity was strongly associated with the presence of MASH only in men[42], whereas myosteatosis was strongly associated with non-obese MASLD in both sexes after adjusting for various known risk factors of MASLD[43]. Moreover, the relationship between myosteatosis and liver health is underscored by findings suggesting that muscle fat content, rather than muscle mass alone, is a determinant of liver fibrosis in patients with severe obesity[39]. These findings indicate that interventions aimed at reducing myosteatosis even before muscle mass loss could have beneficial effects on MASLD progression. For example, glucagon-like peptide-1 receptor agonists have been shown to improve liver steatosis in patients with MASLD and type 2 diabetes mellitus, suggesting a potential therapeutic avenue for addressing both muscle and liver fat accumulation[44]. Therefore, addressing myosteatosis may be crucial in managing MASLD and preventing its progression to more severe forms such as MASH and cirrhosis[45]. In conclusion, myosteatosis represents a critical area of research in the context of MASLD and in broader metabolic health. Its association with insulin resistance, liver disease progression, and poor clinical outcomes underscores the necessity for further investigation of its pathophysiological mechanisms and potential therapeutic interventions. As our understanding of myosteatosis evolves, it may serve as a valuable biomarker for assessing the severity of MASLD and guiding treatment strategies aimed at improving patient outcomes.

ALTERED MUSCLE METABOLISM

Altered muscle metabolism in MASLD is a complex issue that is intertwined with insulin resistance, muscle mass, and systemic inflammation. Insulin resistance is one of the primary mechanisms linking muscle metabolism to MASLD development. In patients with MASLD, muscle insulin resistance is often observed, which can precede hepatic insulin resistance, suggesting a sequential relationship in which muscle dysfunction aggravates liver pathology[46,47]. This relationship is further complicated by the presence of increased free fatty acids in the bloodstream, which can lead to increased lipolysis and the subsequent accumulation of lipids in both muscle and liver tissues[48]. High-fat diets trigger mammalian target of rapamycin complex (mTORC) hyperactivation in skeletal muscles[49,50]. In these studies, high-fat challenges increased mTORС1 (and, in some instances, mTORС2) activity, which in turn enhanced S6 kinases 1 phosphorylation and induced inhibitory serine phosphorylation of insulin receptor substrate-1. This cascade diminishes protein kinase B activation and compromises insulin signaling, as observed in both skeletal muscle and liver tissues, thereby altering insulin-dependent pathways and promoting lipid accumulation. Exercise training results in the reduction of mTORC activation with the reversal of lipid-induced skeletal muscle insulin resistance[51,52] and identifies mTORC as a potential target for the treatment of SMA in patients with MASLD. Additionally, systemic inflammation through different cytokines plays a crucial role in the altered muscle metabolism observed in MASLD patients. TNF-α levels were elevated in patients with MASH. TNF-α contributes to insulin resistance by interfering with insulin signaling pathways, which can lead to impaired glucose uptake in muscle tissues[53,54]. Additionally, IL-6 is a key player in the inflammatory response associated with MASLD. Elevated IL-6 levels are linked to increased insulin resistance and can affect muscle metabolism by promoting a catabolic state, which may hinder muscle growth and repair[55,56]. The role of cytokines extends beyond direct effects on insulin signaling; they also influence the secretion of myokines from skeletal muscles. Myokines such as IL-15 and irisin are involved in muscle metabolism and can modulate the inflammatory response. Dysregulation of these myokines can exacerbate muscle wasting and metabolic dysfunction[57]. Notably, in the study of Spanish cohort of patients with MASLD, published in the World Journal of Gastroenterology, no significant SMA but a marked increase in serum levels of irisin and fibroblast growth factor 21 (FGF-21) in advanced disease stages were found[12]. Whether these changes in myokine levels precede the development of SMA in the future in patients with further progression of MASLD or reflect a lower predisposition for the development of SMA in Caucasian populations than in Asian cohorts is not clear. Interestingly, irisin and FGF-21 may play a protective role for MASLD[58]; at least in animal models, exercise-induced increase in irisin levels was associated with decreased inflammation and improved MASLD[59].

Evidence suggests that muscle-derived FGF-21 improves hepatic insulin sensitivity through several mechanisms. FGF-21 improves hepatic insulin sensitivity by inhibiting mTORC1, which enhances insulin signaling and glycogen synthesis in the liver[60]. Administration of FGF-21 decreases the hepatocellular diacylglycerol content and reduces protein kinase C activation, which is associated with improved insulin sensitivity in the liver[61]. It also enhances the expression and secretion of adiponectin, an insulin-sensitizing adipokine that mediates its effects on liver and muscle insulin sensitivity[62]. Studies using animal models have demonstrated that FGF-21 administration can lead to significant improvements in metabolic parameters, including reductions in body weight, plasma insulin levels, and liver steatosis[63,64]. However, its effects may vary between sexes, with more pronounced benefits observed in males in certain models[65]. FGF-21 plays a crucial role in enhancing hepatic insulin sensitivity through its regulatory effects on lipid and glucose metabolism. Its potential as a therapeutic agent for metabolic diseases has been supported by various studies highlighting its ability to improve insulin sensitivity and reduce liver fat accumulation[66].

Altered amino acid metabolism, which is observed in many liver diseases including MASLD, may affect muscle metabolism and alter muscle function and structure. Metabolomic analysis revealed that several major amino acid pathways are dysregulated in MASLD, with tyrosine metabolism being the most affected[67]. Tyrosine metabolism plays a crucial role in regulating skeletal muscle function and adaptation. Tyrosine phosphorylation, a post-translational modification, is integral to various signaling pathways that influence muscle metabolism, insulin sensitivity, and overall muscle health. Research has shown that tyrosine phosphorylation is involved in the regulation of key metabolic enzymes in skeletal muscle, such as lactate dehydrogenase A and pyruvate dehydrogenase kinase 1, which are critical for glycolytic processes[68]. Furthermore, the phosphorylation state of insulin receptor substrate proteins, particularly insulin receptor substrate-1, is significantly affected by tyrosine modifications, which can lead to insulin resistance—a common issue in metabolic disorders[69,70]. The role of tyrosine phosphorylation extends beyond metabolic regulation and influences the structural aspects of muscle cells. For example, proteins such as paxillin, which are involved in focal adhesion and cytoskeletal dynamics, undergo tyrosine phosphorylation that affects their function in muscle cells[71,72]. This suggests that tyrosine metabolism not only regulates metabolic pathways, but also contributes to the structural integrity and adaptability of skeletal muscles, which are important for the alteration of muscle functions in MASLD. Thus, altered muscle metabolism includes muscle insulin resistance, which may precede hepatic insulin resistance and development of MASLD. The increase in proinflammatory cytokines and disturbances in tyrosine metabolism that occur during the course of MASLD may further alter muscle metabolism, leading to the development of myosteatosis and/or sarcopenia.

TREATMENT OF SMA IN MASLD PATIENTS

No specific pharmacological treatment for SMA in MASLD patients has been approved yet. However, common factors preceding both conditions, such as insulin resistance, may be potential targets for non-pharmacological and pharmacological interventions to correct them. Diet and nutritional interventions in combination with physical activity are still the cornerstones of the treatment of MASLD[1]. Specific dietary patterns are related to MASLD and sarcopenic obesity[73] and to MASLD in lean subjects[74]; therefore, addressing them through dietary counseling, such as reducing sugar-sweetened beverage intake[75], may help manage both sarcopenia and MASLD by mitigating visceral fat accumulation and improving insulin sensitivity. Caloric restriction is a key factor for effective weight reduction, which is associated with the resolution of MASLD and improvement of MASH/fibrosis[76,77]. However, muscles are major consumers of energy; therefore, specific nutrition should be prescribed to reduce fat tissue and preserve muscle mass. Usually, an isocaloric diet or diet with high protein content, especially enriched with leucin, can preserve muscle mass and provide adequate weight control if patients with MASLD are compliant with long-term dietary interventions[78-80]. Controlled trials showed that the Mediterranean dietary pattern was at least as effective as using diets with caloric restriction but provided better compliance in patients with MASLD[81,82]. Adherence to a Mediterranean diet is generally associated with positive effects on muscle mass and function. However, evidence regarding its impact on muscle strength is less clear and no definitive positive effect on sarcopenia has been established[83,84]. Greater adherence to the Mediterranean diet was associated with a lower risk of probable sarcopenia. This association is particularly evident in observational studies, suggesting that the diet may help maintain muscle function in older adults[85,86]. The Mediterranean diet is rich in anti-inflammatory and antioxidant nutrients, such as extra virgin olive oil, fruits, vegetables, and fish, which may contribute to its protective effects against muscle deterioration[87]. While observational studies suggest benefits, there is a notable lack of interventional studies to confirm the effectiveness of the Mediterranean diet in preventing or treating sarcopenia[85,88].

Functional foods rich in specific nutrients may enhance the muscle performance and mitigate the effects of sarcopenia. Nutraceuticals such as curcumin, resveratrol, catechin, soy protein, and ginseng have been reported to improve physical performance, muscle strength, and mitochondrial function in patients with sarcopenia. These products also increase muscle mass and inhibit muscle atrophy without significant side effects[89,90]. Interestingly, a recently published double-blind placebo-controlled trial of 52 patients with biopsy-proven MASH treated with curcumin for 72 weeks demonstrated significant regression of liver inflammation and fibrosis, regardless of changes in body mass index (BMI)[91]. Unfortunately, SMA were not evaluated in this study; however, profound decreases in blood insulin, glucose, cholesterol, truglycerides, and inflammatory marker levels allowed us to speculate that curcumin may have a positive effect on SMA, considering the common pathogenesis of MASLD. A novel food containing omega-3 fatty acids, leucine, and probiotic Lactobacillus paracasei PS23 significantly improved appendicular lean mass, muscle performance, and handgrip strength in elderly sarcopenic patients over a 2-month period[92]. Consumption of antioxidant-rich foods, including fruits and vegetables, along with supplements, such as magnesium, vitamin E, and vitamin D, has been associated with improved muscle strength and function. These interventions according to meta-analyses have shown to reduce the time to complete physical tasks and increase handgrip strength[93,94]. Although these findings are promising, further longitudinal and clinical studies are necessary to establish definitive treatment strategies.

Physical activity and different exercise protocols have been confirmed by numerous meta-analyses to be effective tools for decreasing insulin resistance and therefore for the prevention and treatment of MASLD[95-98]. Similarly, the most effective exercise protocols for improving muscle mass and function in individuals with sarcopenia involve a combination of resistance training, high-intensity interval training, and nutritional intervention. Conventional high-intensity resistance training is effective for improving muscle mass and strength. Low-load resistance training with blood flow restriction is an alternative that improves muscle strength and cardiovascular health, making it suitable for individuals who may not tolerate high-intensity exercise and has been shown to significantly enhance appendicular SMI and physical performance measures[99]. High-intensity interval training is a potent method for enhancing muscle function, strength, and hypertrophy in older adults. It improves body composition and cardiorespiratory capacity, which are crucial for sarcopenia management[100]. Combining resistance exercise with nutritional support provides better results than exercise alone, and usually includes a diet that is well balanced for protein consumption. Protein intake of ≥ 0.8 g/kg body weight/day has shown beneficial effects on muscle mass, particularly when combined with exercise[88], whereas a higher protein intake of 1.0–1.2 g/kg/day is suggested for healthy older adults[101]. Instead of increasing protein consumption, additional amino acid intake or their metabolites can enhance muscle protein synthesis and improve muscle mass and function[102]. Although protein intake is crucial for muscle health, the multifactorial nature of sarcopenia suggests that a holistic approach may yield better outcomes. Using vitamin D and omega-3 fatty acids together with a high-protein diet significantly improves muscle regeneration, strength, and lipid profiles[101]; however, a whole diet approach, including fruits, vegetables, and adherence to the Mediterranean diet, is associated with better patient compliance with similar physical performance and muscle function[88]. Therefore, for both MASLD and SMA, a combination of specific diet and physical activity/exercise is crucial for successful treatment; however, there are no results of the study published yet in which both conditions were taken into account, monitored, and specifically treated with different diets and exercise protocols to find the best one for long-term outcomes and good patient adherence. This is a key direction for future research.

Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) have been introduced for the treatment of diabetes mellitus; however, they are increasingly used for weight loss in obese individuals, who often have MASLD, and their effects on muscle mass during this process are complex and can vary depending on the context and specific GLP-1 agonist used. Elevated blood GLP-1 levels have been associated with reduced myogenic differentiation, which potentially negatively affects muscle health. Thus, patients with sarcopenia exhibit elevated plasma levels of GLP-1, in contrast to non-sarcopenic individuals. This can be explained by the effect of GLP-1 on both the membrane translocation of glucose transporter 4 and dispersion of mitochondria, which significantly hinders glucose uptake and the production of mitochondrial adenosine triphosphate necessary for the myogenic program, leading to a reduction in myogenic differentiation[103]. Despite these concerns, a large pharmacovigilance study found no significant association between GLP-1 RAs and sarcopenia, suggesting that these drugs do not inherently increase sarcopenia risk[104]. Moreover, dulaglutide, a GLP-1 RA, protects against muscle injury in diabetic sarcopenia by reducing inflammation and promoting myoblast differentiation[105]. This suggests its potential therapeutic role in the management of sarcopenia-related muscle damage.

While GLP-1 RAs are effective in reducing body weight primarily through fat loss, they also affect lean body mass, which includes muscle. Meta-analyses indicate that semaglutide and liraglutide can lead to reductions in lean body mass, although the percentage of lean mass loss relative to total weight loss is comparable to non-users. This suggests that while there is a loss, it may not be disproportionately large compared with the overall weight loss[106]. Similar to low-calorie diets, GLP-1 RAs can lead to muscle atrophy through caloric restriction, which is a common consequence of weight loss interventions. However, GLP-1 RAs have been shown to ameliorate muscle atrophy by modulating pathways such as the SIRT1 pathway, which is involved in muscle preservation[107]. Treatment with GLP-1 RAs has also been associated with improved muscle quality, as evidenced by enhanced insulin sensitivity and reduced muscle fat infiltration. These changes suggest an adaptive response that may help maintain muscle function despite reduction in muscle volume[108,109]. Even in elderly obese patients, GLP-1 RAs such as semaglutide have shown potential in preserving lean body mass during weight loss, with studies indicating a significant reduction in fat mass while maintaining lean mass[110]. In patients with sarcopenic obesity, which is often associated with MASLD, the combination of GLP-1 RAs with future drugs, such as myostatin and activin A inhibitors, is a promising strategy. Animal models have shown that such combinations can preserve or even increase muscle mass while enhancing fat loss[111,112].

A dozen clinical trials on GLP-1 RAs in patients with MASLD have reported inconsistent results. However, according to meta-analyses, they significantly improved hepatic function markers, hepatic steatosis, inflammation, and fibrosis as well as reduced BMI, subcutaneous fat mass, and waist circumference[113,114]. GLP-1 RAs have not yet been approved for the treatment of MASLD; they are actively used in at least 1/3 of patients with MASLD who have diabetes[115], which provides an excellent opportunity to evaluate their long-term safety and efficacy for MASLD and SMA. Tirzepatide, a dual agonist of glucose-dependent insulinotropic polypeptide and GLP-1 receptors, has shown promising therapeutic effects in MASH. Compared to other treatments, such as semaglutide, tirzepatide has shown superior effects in reducing hepatic fat deposition and improving liver inflammation in animal models[116]. In clinical settings, switching from other GLP-1 analogs to tirzepatide resulted in improved liver enzyme levels and a trend towards reduced fibrosis, indicating its potential for broader application in liver disease management[117]. In a phase 2 study involving 157 patients with MASH and moderate or severe fibrosis, treatment with tirzepatide for 52 weeks was more effective than placebo with respect to the resolution of MASH without worsening of fibrosis[118]. However, the small sample size did not provide adequate statistical power to evaluate the effects of turzepatide on liver fibrosis. While GLP-1 RAs offer significant benefits in terms of weight loss and metabolic improvements, as well as liver inflammation and steatosis, the potential for muscle mass loss remains a concern, particularly in MASLD populations with SMA or those at high risk for sarcopenia, such as elderly patients.

CONCLUSION

The development of new diagnostic approaches, new combination therapies, and exploration of mechanisms to preserve muscle mass are critical areas for future research. Additionally, more comprehensive assessments of muscle health, including function and strength, in different groups of patients with MASL/MASH are needed to fully understand the impact of existing and future treatment strategies of MASLD on SMA and vice versa.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: Russia

Peer-review report’s classification

Scientific Quality: Grade A, Grade B, Grade B

Novelty: Grade A, Grade A, Grade A

Creativity or Innovation: Grade A, Grade B, Grade B

Scientific Significance: Grade A, Grade A, Grade A

P-Reviewer: Huang AH; Li MY S-Editor: Luo ML L-Editor: A P-Editor: Yu HG

References
1.  European Association for the Study of the Liver (EASL); European Association for the Study of Diabetes (EASD);  European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines on the management of metabolic dysfunction-associated steatotic liver disease (MASLD). J Hepatol. 2024;81:492-542.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 70]  [Reference Citation Analysis (1)]
2.  Kanwal F, Neuschwander-Tetri BA, Loomba R, Rinella ME. Metabolic dysfunction-associated steatotic liver disease: Update and impact of new nomenclature on the American Association for the Study of Liver Diseases practice guidance on nonalcoholic fatty liver disease. Hepatology. 2024;79:1212-1219.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 92]  [Cited by in RCA: 15]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
3.  Golabi P, Isakov V, Younossi ZM. Nonalcoholic Fatty Liver Disease: Disease Burden and Disease Awareness. Clin Liver Dis. 2023;27:173-186.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 17]  [Cited by in RCA: 15]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
4.  Feng G, Targher G, Byrne CD, Yilmaz Y, Wai-Sun Wong V, Adithya Lesmana CR, Adams LA, Boursier J, Papatheodoridis G, El-Kassas M, Méndez-Sánchez N, Sookoian S, Castera L, Chan WK, Ye F, Treeprasertsuk S, Cortez-Pinto H, Yu HH, Kim W, Romero-Gómez M, Nakajima A, Win KM, Kim SU, Holleboom AG, Sebastiani G, Ocama P, Ryan JD, Lupșor-Platon M, Ghazinyan H, Al-Mahtab M, Hamid S, Perera N, Alswat KA, Pan Q, Long MT, Isakov V, Mi M, Arrese M, Sanyal AJ, Sarin SK, Leite NC, Valenti L, Newsome PN, Hagström H, Petta S, Yki-Järvinen H, Schattenberg JM, Castellanos Fernández MI, Leclercq IA, Aghayeva G, Elzouki AN, Tumi A, Sharara AI, Labidi A, Sanai FM, Matar K, Al-Mattooq M, Akroush MW, Benazzouz M, Debzi N, Alkhatry M, Barakat S, Al-Busafi SA, Rwegasha J, Yang W, Adwoa A, Opio CK, Sotoudeheian M, Wong YJ, George J, Zheng MH. Global burden of metabolic dysfunction-associated steatotic liver disease, 2010 to 2021. JHEP Rep. 2025;7:101271.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Reference Citation Analysis (0)]
5.  Rosenberg IH. Sarcopenia: origins and clinical relevance. J Nutr. 1997;127:990S-991S.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1263]  [Cited by in RCA: 1425]  [Article Influence: 50.9]  [Reference Citation Analysis (0)]
6.  Cruz-Jentoft AJ, Bahat G, Bauer J, Boirie Y, Bruyère O, Cederholm T, Cooper C, Landi F, Rolland Y, Sayer AA, Schneider SM, Sieber CC, Topinkova E, Vandewoude M, Visser M, Zamboni M; Writing Group for the European Working Group on Sarcopenia in Older People 2 (EWGSOP2), and the Extended Group for EWGSOP2. Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing. 2019;48:16-31.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6646]  [Cited by in RCA: 7481]  [Article Influence: 1246.8]  [Reference Citation Analysis (0)]
7.  Deng C, Ou Q, Ou X, Pan D. Association between non-alcoholic fatty liver disease and risk of sarcopenia: a systematic review and meta-analysis. BMJ Open. 2024;14:e078933.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
8.  Evuri P, Selvi C, E M, A A, Karunakaran P, K M, R M, A C, I S. Study of Prevalence of Sarcopenia in Chronic Liver Disease and its Impact on Outcomes. IJIRMS. 2022;7:335-339.  [PubMed]  [DOI]  [Full Text]
9.  Himoto T, Miyatake K, Maeba T, Masaki T. Verification of the Nutritional and Dietary Factors Associated with Skeletal Muscle Index in Japanese Patients with Nonalcoholic Fatty Liver Disease. Can J Gastroenterol Hepatol. 2020;2020:3576974.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
10.  Malik A, Javaid S, Malik MI, Qureshi S. Relationship between sarcopenia and metabolic dysfunction-associated steatotic liver disease (MASLD): A systematic review and meta-analysis. Ann Hepatol. 2024;29:101544.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
11.  Li X, He J, Sun Q. The prevalence and effects of sarcopenia in patients with metabolic dysfunction-associated steatotic liver disease (MASLD): A systematic review and meta-analysis. Clin Nutr. 2024;43:2005-2016.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
12.  Real Martinez Y, Fernandez-Garcia CE, Fuertes-Yebra E, Calvo Soto M, Berlana A, Barrios V, Caldas M, Gonzalez Moreno L, Garcia-Buey L, Molina Baena B, Sampedro-Nuñez M, Beceiro MJ, García-Monzón C, González-Rodríguez Á. Assessment of skeletal muscle alterations and circulating myokines in metabolic dysfunction-associated steatotic liver disease: A cross-sectional study. World J Gastroenterol. 2025;31:100039.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (4)]
13.  Kim D, Wijarnpreecha K, Sandhu KK, Cholankeril G, Ahmed A. Sarcopenia in nonalcoholic fatty liver disease and all-cause and cause-specific mortality in the United States. Liver Int. 2021;41:1832-1840.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 50]  [Cited by in RCA: 48]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
14.  Zhai Y, Xiao Q. The Common Mechanisms of Sarcopenia and NAFLD. Biomed Res Int. 2017;2017:6297651.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 23]  [Cited by in RCA: 40]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
15.  Chung GE, Oh S, Ahn DW, Kim SH, Jung YJ, Kim JW, Kim BG, Lee KL, Jeong JB. Effects of additive interactions among obesity, visceral adiposity, and sarcopenia on nonalcoholic fatty liver disease. Sci Rep. 2023;13:3628.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
16.  Mikolasevic I, Pavic T, Kanizaj TF, Bender DV, Domislovic V, Krznaric Z. Nonalcoholic Fatty Liver Disease and Sarcopenia: Where Do We Stand? Can J Gastroenterol Hepatol. 2020;2020:8859719.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 17]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
17.  Sinn DH, Kang D, Kang M, Guallar E, Hong YS, Lee KH, Park J, Cho J, Gwak GY. Nonalcoholic fatty liver disease and accelerated loss of skeletal muscle mass: A longitudinal cohort study. Hepatology. 2022;76:1746-1754.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 29]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
18.  Guo M, Xiang L, Yao J, Zhang J, Zhu S, Wang D, Liu C, Li G, Wang J, Gao Y, Xie C, Ma X, Xu L, Zhou J. Comprehensive Transcriptome Profiling of NAFLD- and NASH-Induced Skeletal Muscle Dysfunction. Front Endocrinol (Lausanne). 2022;13:851520.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
19.  Wang J, Du J, Ge X, Peng W, Guo X, Li W, Huang S. Circulating Ism1 Reduces the Risk of Type 2 Diabetes but not Diabetes-Associated NAFLD. Front Endocrinol (Lausanne). 2022;13:890332.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
20.  Lee YH, Kim SU, Song K, Park JY, Kim DY, Ahn SH, Lee BW, Kang ES, Cha BS, Han KH. Sarcopenia is associated with significant liver fibrosis independently of obesity and insulin resistance in nonalcoholic fatty liver disease: Nationwide surveys (KNHANES 2008-2011). Hepatology. 2016;63:776-786.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 281]  [Cited by in RCA: 269]  [Article Influence: 29.9]  [Reference Citation Analysis (0)]
21.  Bhanji RA, Narayanan P, Allen AM, Malhi H, Watt KD. Sarcopenia in hiding: The risk and consequence of underestimating muscle dysfunction in nonalcoholic steatohepatitis. Hepatology. 2017;66:2055-2065.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 142]  [Cited by in RCA: 184]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
22.  Petta S, Ciminnisi S, Di Marco V, Cabibi D, Cammà C, Licata A, Marchesini G, Craxì A. Sarcopenia is associated with severe liver fibrosis in patients with non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2017;45:510-518.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 126]  [Cited by in RCA: 163]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
23.  Okubo T, Atsukawa M, Tsubota A, Ono H, Kawano T, Yoshida Y, Arai T, Hayama K, Itokawa N, Kondo C, Iwakiri K. Low vitamin D levels accelerates muscle mass loss in patients with chronic liver disease. PLoS One. 2024;19:e0299313.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
24.  Yu R, Shi Q, Liu L, Chen L. Relationship of sarcopenia with steatohepatitis and advanced liver fibrosis in non-alcoholic fatty liver disease: a meta-analysis. BMC Gastroenterol. 2018;18:51.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 49]  [Cited by in RCA: 65]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
25.  Golabi P, Gerber L, Paik JM, Deshpande R, de Avila L, Younossi ZM. Contribution of sarcopenia and physical inactivity to mortality in people with non-alcoholic fatty liver disease. JHEP Rep. 2020;2:100171.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 25]  [Cited by in RCA: 75]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
26.  Harring M, Golabi P, Paik JM, Shah D, Racila A, Cable R, Srishord M, Younossi ZM. Sarcopenia Among Patients With Nonalcoholic Fatty Liver Disease (NAFLD) Is Associated With Advanced Fibrosis. Clin Gastroenterol Hepatol. 2023;21:2876-2888.e5.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 21]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
27.  Wijarnpreecha K, Panjawatanan P, Thongprayoon C, Jaruvongvanich V, Ungprasert P. Sarcopenia and risk of nonalcoholic fatty liver disease: A meta-analysis. Saudi J Gastroenterol. 2018;24:12-17.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 48]  [Cited by in RCA: 52]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
28.  Cai C, Song X, Chen Y, Chen X, Yu C. Relationship between relative skeletal muscle mass and nonalcoholic fatty liver disease: a systematic review and meta-analysis. Hepatol Int. 2020;14:115-126.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 80]  [Cited by in RCA: 79]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
29.  Tovo CV, Fernandes SA, Buss C, de Mattos AA. Sarcopenia and non-alcoholic fatty liver disease: Is there a relationship? A systematic review. World J Hepatol. 2017;9:326-332.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 33]  [Cited by in RCA: 28]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
30.  Kim G, Lee SE, Lee YB, Jun JE, Ahn J, Bae JC, Jin SM, Hur KY, Jee JH, Lee MK, Kim JH. Relationship Between Relative Skeletal Muscle Mass and Nonalcoholic Fatty Liver Disease: A 7-Year Longitudinal Study. Hepatology. 2018;68:1755-1768.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 100]  [Cited by in RCA: 147]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
31.  Chen X, Xiang H, Tan L, Zhou J, Tang J, Hu X, Yang M. Psoriasis Is Associated With Myosteatosis but Not Sarcopenia: A Case-Control Study. Front Med (Lausanne). 2021;8:754932.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
32.  Henin G, Loumaye A, Deldicque L, Leclercq IA, Lanthier N. Unlocking liver health: Can tackling myosteatosis spark remission in metabolic dysfunction-associated steatotic liver disease? Liver Int. 2024;44:1781-1796.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
33.  Henin G, Loumaye A, Leclercq IA, Lanthier N. Myosteatosis: Diagnosis, pathophysiology and consequences in metabolic dysfunction-associated steatotic liver disease. JHEP Rep. 2024;6:100963.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 17]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
34.  Kamiliou A, Lekakis V, Chrysavgis L, Cholongitas E. Prevalence and impact on the outcome of myosteatosis in patients with cirrhosis: a systematic review and meta-analysis. Hepatol Int. 2024;18:688-699.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Reference Citation Analysis (0)]
35.  Kim MJ, Cho YK, Kim EH, Lee MJ, Lee WJ, Kim HK, Jung CH. Association between metabolic dysfunction-associated steatotic liver disease and myosteatosis measured by computed tomography. J Cachexia Sarcopenia Muscle. 2024;15:1942-1952.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
36.  Ebadi M, Tsien C, Bhanji RA, Dunichand-Hoedl AR, Rider E, Motamedrad M, Mazurak VC, Baracos V, Montano-Loza AJ. Myosteatosis in Cirrhosis: A Review of Diagnosis, Pathophysiological Mechanisms and Potential Interventions. Cells. 2022;11:1216.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 36]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
37.  Geladari E, Alexopoulos T, Kontogianni MD, Vasilieva L, Mani I, Tenta R, Sevastianos V, Vlachogiannakos I, Alexopoulou A. The Presence of Myosteatosis Is Associated with Age, Severity of Liver Disease and Poor Outcome and May Represent a Prodromal Phase of Sarcopenia in Patients with Liver Cirrhosis. J Clin Med. 2023;12:3332.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
38.  Kamiliou A, Lekakis V, Xynos G, Cholongitas E. Prevalence of and Impact on the Outcome of Myosteatosis in Patients with Hepatocellular Carcinoma: A Systematic Review and Meta-Analysis. Cancers (Basel). 2024;16:952.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
39.  Han E, Kim MK, Lee HW, Ryu S, Kim HS, Jang BK, Suh Y. Muscle fat contents rather than muscle mass determines nonalcoholic steatohepatitis and liver fibrosis in patients with severe obesity. Obesity (Silver Spring). 2022;30:2440-2449.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
40.  Nachit M, De Rudder M, Thissen JP, Schakman O, Bouzin C, Horsmans Y, Vande Velde G, Leclercq IA. Myosteatosis rather than sarcopenia associates with non-alcoholic steatohepatitis in non-alcoholic fatty liver disease preclinical models. J Cachexia Sarcopenia Muscle. 2021;12:144-158.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 22]  [Cited by in RCA: 44]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
41.  Nachit M, Kwanten WJ, Thissen JP, Op De Beeck B, Van Gaal L, Vonghia L, Verrijken A, Driessen A, Horsmans Y, Francque S, Leclercq IA. Muscle fat content is strongly associated with NASH: A longitudinal study in patients with morbid obesity. J Hepatol. 2021;75:292-301.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 36]  [Cited by in RCA: 84]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
42.  Li G, Rios RS, Wang XX, Yu Y, Zheng KI, Huang OY, Tang LJ, Ma HL, Jin Y, Targher G, Byrne CD, Pan XY, Zheng MH. Sex influences the association between appendicular skeletal muscle mass to visceral fat area ratio and non-alcoholic steatohepatitis in patients with biopsy-proven non-alcoholic fatty liver disease. Br J Nutr. 2022;127:1613-1620.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
43.  Kim HK, Bae SJ, Lee MJ, Kim EH, Park H, Kim HS, Cho YK, Jung CH, Lee WJ, Choe J. Association of Visceral Fat Obesity, Sarcopenia, and Myosteatosis with Non-Alcoholic Fatty Liver Disease without Obesity. Clin Mol Hepatol. 2023;29:987-1001.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 19]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
44.  Kakegawa T, Sugimoto K, Saito K, Yunaiyama D, Araki Y, Wada T, Takahashi H, Yoshimasu Y, Takeuchi H, Itoi T. Favorable liver and skeletal muscle changes in patients with MASLD and T2DM receiving glucagon-like peptide-1 receptor agonist: A prospective cohort study. Medicine (Baltimore). 2024;103:e38444.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Reference Citation Analysis (0)]
45.  Chakravarthy MV, Siddiqui MS, Forsgren MF, Sanyal AJ. Harnessing Muscle-Liver Crosstalk to Treat Nonalcoholic Steatohepatitis. Front Endocrinol (Lausanne). 2020;11:592373.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 37]  [Cited by in RCA: 48]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
46.  Rabøl R, Petersen KF, Dufour S, Flannery C, Shulman GI. Reversal of muscle insulin resistance with exercise reduces postprandial hepatic de novo lipogenesis in insulin resistant individuals. Proc Natl Acad Sci U S A. 2011;108:13705-13709.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 127]  [Cited by in RCA: 130]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
47.  Han X, Liu C, Xue Y, Wang J, Xue C, Yanagita T, Gao X, Wang Y. Long-term fatty liver-induced insulin resistance in orotic acid-induced nonalcoholic fatty liver rats. Biosci Biotechnol Biochem. 2016;80:735-743.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 10]  [Cited by in RCA: 9]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
48.  Gad H. Study of Plasma Adiponectin and Insulin Resistance in Subjects with Non-Alcoholic Fatty Liver Disease. Arch Gen Intern Med. 2018;2.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
49.  Khamzina L, Veilleux A, Bergeron S, Marette A. Increased activation of the mammalian target of rapamycin pathway in liver and skeletal muscle of obese rats: possible involvement in obesity-linked insulin resistance. Endocrinology. 2005;146:1473-1481.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 410]  [Cited by in RCA: 435]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
50.  Kumar A, Lawrence JC Jr, Jung DY, Ko HJ, Keller SR, Kim JK, Magnuson MA, Harris TE. Fat cell-specific ablation of rictor in mice impairs insulin-regulated fat cell and whole-body glucose and lipid metabolism. Diabetes. 2010;59:1397-1406.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 213]  [Cited by in RCA: 205]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
51.  Liu X, Yuan H, Niu Y, Niu W, Fu L. The role of AMPK/mTOR/S6K1 signaling axis in mediating the physiological process of exercise-induced insulin sensitization in skeletal muscle of C57BL/6 mice. Biochim Biophys Acta. 2012;1822:1716-1726.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 34]  [Cited by in RCA: 34]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
52.  Rivas DA, Yaspelkis BB 3rd, Hawley JA, Lessard SJ. Lipid-induced mTOR activation in rat skeletal muscle reversed by exercise and 5'-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside. J Endocrinol. 2009;202:441-451.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 49]  [Cited by in RCA: 48]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
53.  Duan Y, Pan X, Luo J, Xiao X, Li J, Bestman PL, Luo M. Association of Inflammatory Cytokines With Non-Alcoholic Fatty Liver Disease. Front Immunol. 2022;13:880298.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7]  [Cited by in RCA: 171]  [Article Influence: 57.0]  [Reference Citation Analysis (0)]
54.  Vansaun MN, Mendonsa AM, Lee Gorden D. Hepatocellular proliferation correlates with inflammatory cell and cytokine changes in a murine model of nonalchoholic fatty liver disease. PLoS One. 2013;8:e73054.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 34]  [Cited by in RCA: 43]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
55.  Mohamed AA, Shousha WG, Shaker O, Mahdy ME, Ibrahim EMA, Mohmoud S, Khairalla A. Role of Serum Adiponectin, IL-6 and Hs CRP in Nonalcoholic Fatty Liver Egyptian Patients. IJBCRR. 2014;4:493-504.  [PubMed]  [DOI]  [Full Text]
56.  Kempinska-Podhorodecka A, Wunsch E, Milkiewicz P, Stachowska E, Milkiewicz M. The Association between SOCS1-1656G>A Polymorphism, Insulin Resistance and Obesity in Nonalcoholic Fatty Liver Disease (NAFLD) Patients. J Clin Med. 2019;8:1912.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 6]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
57.  Jiang LQ, Franck N, Egan B, Sjögren RJ, Katayama M, Duque-Guimaraes D, Arner P, Zierath JR, Krook A. Autocrine role of interleukin-13 on skeletal muscle glucose metabolism in type 2 diabetic patients involves microRNA let-7. Am J Physiol Endocrinol Metab. 2013;305:E1359-E1366.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 88]  [Cited by in RCA: 94]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
58.  Bucarey JL, Trujillo-González I, Paules EM, Espinosa A. Myokines and Their Potential Protective Role Against Oxidative Stress in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Antioxidants (Basel). 2024;13:1363.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
59.  Zhu W, Sahar NE, Javaid HMA, Pak ES, Liang G, Wang Y, Ha H, Huh JY. Exercise-Induced Irisin Decreases Inflammation and Improves NAFLD by Competitive Binding with MD2. Cells. 2021;10:3306.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 35]  [Cited by in RCA: 64]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
60.  Gong Q, Hu Z, Zhang F, Cui A, Chen X, Jiang H, Gao J, Chen X, Han Y, Liang Q, Ye D, Shi L, Chin YE, Wang Y, Xiao H, Guo F, Liu Y, Zang M, Xu A, Li Y. Fibroblast growth factor 21 improves hepatic insulin sensitivity by inhibiting mammalian target of rapamycin complex 1 in mice. Hepatology. 2016;64:425-438.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 100]  [Cited by in RCA: 141]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
61.  Camporez JP, Jornayvaz FR, Petersen MC, Pesta D, Guigni BA, Serr J, Zhang D, Kahn M, Samuel VT, Jurczak MJ, Shulman GI. Cellular mechanisms by which FGF21 improves insulin sensitivity in male mice. Endocrinology. 2013;154:3099-3109.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 151]  [Cited by in RCA: 172]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
62.  Lin Z, Tian H, Lam KS, Lin S, Hoo RC, Konishi M, Itoh N, Wang Y, Bornstein SR, Xu A, Li X. Adiponectin mediates the metabolic effects of FGF21 on glucose homeostasis and insulin sensitivity in mice. Cell Metab. 2013;17:779-789.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 465]  [Cited by in RCA: 544]  [Article Influence: 45.3]  [Reference Citation Analysis (0)]
63.  Bartesaghi S, Wallenius K, Hovdal D, Liljeblad M, Wallin S, Dekker N, Barlind L, Davies N, Seeliger F, Winzell MS, Patel S, Theisen M, Brito L, Bergenhem N, Andersson S, Peng XR. Subcutaneous delivery of FGF21 mRNA therapy reverses obesity, insulin resistance, and hepatic steatosis in diet-induced obese mice. Mol Ther Nucleic Acids. 2022;28:500-513.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
64.  Yano K, Yamaguchi K, Seko Y, Okishio S, Ishiba H, Tochiki N, Takahashi A, Kataoka S, Okuda K, Liu Y, Fujii H, Umemura A, Moriguchi M, Okanoue T, Itoh Y. Hepatocyte-specific fibroblast growth factor 21 overexpression ameliorates high-fat diet-induced obesity and liver steatosis in mice. Lab Invest. 2022;102:281-289.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 16]  [Cited by in RCA: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
65.  Makarova E, Kazantseva A, Dubinina A, Denisova E, Jakovleva T, Balybina N, Bgatova N, Baranov K, Bazhan N. Fibroblast Growth Factor 21 (FGF21) Administration Sex-Specifically Affects Blood Insulin Levels and Liver Steatosis in Obese A(y) Mice. Cells. 2021;10:3440.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
66.  Falamarzi K, Malekpour M, Tafti MF, Azarpira N, Behboodi M, Zarei M. The role of FGF21 and its analogs on liver associated diseases. Front Med (Lausanne). 2022;9:967375.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 21]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
67.  Jin R, Banton S, Tran VT, Konomi JV, Li S, Jones DP, Vos MB. Amino Acid Metabolism is Altered in Adolescents with Nonalcoholic Fatty Liver Disease-An Untargeted, High Resolution Metabolomics Study. J Pediatr. 2016;172:14-19.e5.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 59]  [Cited by in RCA: 70]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
68.  Lundby A, Secher A, Lage K, Nordsborg NB, Dmytriyev A, Lundby C, Olsen JV. Quantitative maps of protein phosphorylation sites across 14 different rat organs and tissues. Nat Commun. 2012;3:876.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 269]  [Cited by in RCA: 279]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
69.  Kim JK, Fillmore JJ, Sunshine MJ, Albrecht B, Higashimori T, Kim DW, Liu ZX, Soos TJ, Cline GW, O'Brien WR, Littman DR, Shulman GI. PKC-theta knockout mice are protected from fat-induced insulin resistance. J Clin Invest. 2004;114:823-827.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 172]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
70.  Pilon G, Charbonneau A, White PJ, Dallaire P, Perreault M, Kapur S, Marette A. Endotoxin mediated-iNOS induction causes insulin resistance via ONOO⁻ induced tyrosine nitration of IRS-1 in skeletal muscle. PLoS One. 2010;5:e15912.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 53]  [Cited by in RCA: 54]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
71.  Gordon SE, Flück M, Booth FW. Selected Contribution: Skeletal muscle focal adhesion kinase, paxillin, and serum response factor are loading dependent. J Appl Physiol (1985). 2001;90:1174-83; discussion 1165.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 100]  [Cited by in RCA: 98]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
72.  Tang DD, Turner CE, Gunst SJ. Expression of non-phosphorylatable paxillin mutants in canine tracheal smooth muscle inhibits tension development. J Physiol. 2003;553:21-35.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 54]  [Cited by in RCA: 57]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
73.  Hoteit M, Dagher M, Tzenios N, Al Kaaki N, Rkein G, Chahine AR, Sacre Y, Hotayt S, Matar R, Hallal M, Maitar M, Hotayt B. Influence of Sugar-Sweetened Beverages Intake on Sarcopenic Obesity, Visceral Obesity, and Sarcopenia in Lebanese Patients with MASLD: A Case-Control Study. Healthcare (Basel). 2024;12:591.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
74.  Long MT, Noureddin M, Lim JK. AGA Clinical Practice Update: Diagnosis and Management of Nonalcoholic Fatty Liver Disease in Lean Individuals: Expert Review. Gastroenterology. 2022;163:764-774.e1.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 170]  [Cited by in RCA: 161]  [Article Influence: 53.7]  [Reference Citation Analysis (0)]
75.  Schwimmer JB, Ugalde-Nicalo P, Welsh JA, Angeles JE, Cordero M, Harlow KE, Alazraki A, Durelle J, Knight-Scott J, Newton KP, Cleeton R, Knott C, Konomi J, Middleton MS, Travers C, Sirlin CB, Hernandez A, Sekkarie A, McCracken C, Vos MB. Effect of a Low Free Sugar Diet vs Usual Diet on Nonalcoholic Fatty Liver Disease in Adolescent Boys: A Randomized Clinical Trial. JAMA. 2019;321:256-265.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 121]  [Cited by in RCA: 182]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
76.  Koutoukidis DA, Koshiaris C, Henry JA, Noreik M, Morris E, Manoharan I, Tudor K, Bodenham E, Dunnigan A, Jebb SA, Aveyard P. The effect of the magnitude of weight loss on non-alcoholic fatty liver disease: A systematic review and meta-analysis. Metabolism. 2021;115:154455.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 38]  [Cited by in RCA: 73]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
77.  Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, Torres-Gonzalez A, Gra-Oramas B, Gonzalez-Fabian L, Friedman SL, Diago M, Romero-Gomez M. Weight Loss Through Lifestyle Modification Significantly Reduces Features of Nonalcoholic Steatohepatitis. Gastroenterology. 2015;149:367-78.e5; quiz e14.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1181]  [Cited by in RCA: 1583]  [Article Influence: 158.3]  [Reference Citation Analysis (1)]
78.  Selezneva KS, Isakov VA, Sentsova TB, Kirillova OO. [An analysis of the efficacy of low-calorie and isocaloric diets in obese patients with nonalcoholic steatohepatitis]. Vopr Pitan. 2014;83:72-78.  [PubMed]  [DOI]
79.  Rachakonda V, Wills R, DeLany JP, Kershaw EE, Behari J. Differential Impact of Weight Loss on Nonalcoholic Fatty Liver Resolution in a North American Cohort with Obesity. Obesity (Silver Spring). 2017;25:1360-1368.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 23]  [Cited by in RCA: 26]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
80.  Anand AC. Nutrition and Muscle in Cirrhosis. J Clin Exp Hepatol. 2017;7:340-357.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 107]  [Cited by in RCA: 118]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
81.  Yaskolka Meir A, Rinott E, Tsaban G, Zelicha H, Kaplan A, Rosen P, Shelef I, Youngster I, Shalev A, Blüher M, Ceglarek U, Stumvoll M, Tuohy K, Diotallevi C, Vrhovsek U, Hu F, Stampfer M, Shai I. Effect of green-Mediterranean diet on intrahepatic fat: the DIRECT PLUS randomised controlled trial. Gut. 2021;70:2085-2095.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 61]  [Cited by in RCA: 155]  [Article Influence: 38.8]  [Reference Citation Analysis (0)]
82.  Marin-Alejandre BA, Cantero I, Perez-Diaz-Del-Campo N, Monreal JI, Elorz M, Herrero JI, Benito-Boillos A, Quiroga J, Martinez-Echeverria A, Uriz-Otano JI, Huarte-Muniesa MP, Tur JA, Martinez JA, Abete I, Zulet MA. Effects of two personalized dietary strategies during a 2-year intervention in subjects with nonalcoholic fatty liver disease: A randomized trial. Liver Int. 2021;41:1532-1544.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 28]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
83.  Papadopoulou SK, Detopoulou P, Voulgaridou G, Tsoumana D, Spanoudaki M, Sadikou F, Papadopoulou VG, Zidrou C, Chatziprodromidou IP, Giaginis C, Nikolaidis P. Mediterranean Diet and Sarcopenia Features in Apparently Healthy Adults over 65 Years: A Systematic Review. Nutrients. 2023;15:1104.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 37]  [Reference Citation Analysis (0)]
84.  Andreo-López MC, Contreras-Bolívar V, García-Fontana B, García-Fontana C, Muñoz-Torres M. The Influence of the Mediterranean Dietary Pattern on Osteoporosis and Sarcopenia. Nutrients. 2023;15:3224.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 17]  [Reference Citation Analysis (0)]
85.  Dominguez LJ, Veronese N, Smith L, Ragusa FS, Schirò P, Di Bella G, Barbagallo M. Associations Between Adherence to the Mediterranean Diet and Incident Sarcopenia in Prospective Cohort Studies. Nutrients. 2025;17:313.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
86.  Cacciatore S, Calvani R, Marzetti E, Picca A, Coelho-Júnior HJ, Martone AM, Massaro C, Tosato M, Landi F. Low Adherence to Mediterranean Diet Is Associated with Probable Sarcopenia in Community-Dwelling Older Adults: Results from the Longevity Check-Up (Lookup) 7+ Project. Nutrients. 2023;15:1026.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 18]  [Reference Citation Analysis (0)]
87.  Nazri NSM, Vanoh D, Soo KL. Natural Food for Sarcopenia: A Narrative Review. Malays J Med Sci. 2022;29:28-42.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
88.  Granic A, Sayer AA, Cooper R, Robinson SM. Nutrition in the prevention and treatment of skeletal muscle ageing and sarcopenia: a single nutrient, a whole food and a whole diet approach. Proc Nutr Soc. 2024;1-16.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
89.  Bagherniya M, Mahdavi A, Shokri-Mashhadi N, Banach M, Von Haehling S, Johnston TP, Sahebkar A. The beneficial therapeutic effects of plant-derived natural products for the treatment of sarcopenia. J Cachexia Sarcopenia Muscle. 2022;13:2772-2790.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 54]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
90.  Mayakrishnan V, Kannappan P, Balakarthikeyan J, Kim CY. Rodent model intervention for prevention and optimal management of sarcopenia: A systematic review on the beneficial effects of nutrients & non-nutrients and exercise to improve skeletal muscle health. Ageing Res Rev. 2024;102:102543.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
91.  Musso G, Pinach S, Mariano F, Saba F, De Michieli F, Framarin L, Berrutti M, Paschetta E, Parente R, Lizet Castillo Y, Leone N, Castellino F, Cassader M, Gambino R. Effect of phospholipid curcumin Meriva on liver histology and kidney disease in nonalcoholic steatohepatitis: A randomized, double-blind, placebo-controlled trial. Hepatology. 2025;81:560-575.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Reference Citation Analysis (0)]
92.  Rondanelli M, Gasparri C, Barrile GC, Battaglia S, Cavioni A, Giusti R, Mansueto F, Moroni A, Nannipieri F, Patelli Z, Razza C, Tartara A, Perna S. Effectiveness of a Novel Food Composed of Leucine, Omega-3 Fatty Acids and Probiotic Lactobacillus paracasei PS23 for the Treatment of Sarcopenia in Elderly Subjects: A 2-Month Randomized Double-Blind Placebo-Controlled Trial. Nutrients. 2022;14:4566.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 24]  [Reference Citation Analysis (0)]
93.  Besora-Moreno M, Llauradó E, Valls RM, Tarro L, Pedret A, Solà R. Antioxidant-rich foods, antioxidant supplements, and sarcopenia in old-young adults ≥55 years old: A systematic review and meta-analysis of observational studies and randomized controlled trials. Clin Nutr. 2022;41:2308-2324.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
94.  Hong SH, Bae YJ. Association of Dietary Vegetable and Fruit Consumption with Sarcopenia: A Systematic Review and Meta-Analysis. Nutrients. 2024;16:1707.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
95.  Hong F, Liu Y, Lebaka VR, Mohammed A, Ye W, Chen B, Korivi M. Effect of Exercise Training on Serum Transaminases in Patients With Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis. Front Physiol. 2022;13:894044.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Reference Citation Analysis (0)]
96.  Xiong Y, Peng Q, Cao C, Xu Z, Zhang B. Effect of Different Exercise Methods on Non-Alcoholic Fatty Liver Disease: A Meta-Analysis and Meta-Regression. Int J Environ Res Public Health. 2021;18:3242.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 8]  [Cited by in RCA: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
97.  Fu L, Zhang W, Ao Y, Zheng Z, Hu H. Efficacy of aerobic and resistance exercises in improving visceral adipose in patients with non-alcoholic fatty liver: a meta-analysis of randomized controlled trials. Z Gastroenterol. 2022;60:1644-1658.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
98.  Słomko J, Zalewska M, Niemiro W, Kujawski S, Słupski M, Januszko-Giergielewicz B, Zawadka-Kunikowska M, Newton J, Hodges L, Kubica J, Zalewski P. Evidence-Based Aerobic Exercise Training in Metabolic-Associated Fatty Liver Disease: Systematic Review with Meta-Analysis. J Clin Med. 2021;10:1659.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 8]  [Cited by in RCA: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
99.  Zhang M, Song Y, Zhu J, Ding P, Chen N. Effectiveness of low-load resistance training with blood flow restriction vs. conventional high-intensity resistance training in older people diagnosed with sarcopenia: a randomized controlled trial. Sci Rep. 2024;14:28427.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
100.  Morcillo-Losa JA, Díaz-Martínez MDP, Ceylan Hİ, Moreno-Vecino B, Bragazzi NL, Párraga Montilla J. Effects of High-Intensity Interval Training on Muscle Strength for the Prevention and Treatment of Sarcopenia in Older Adults: A Systematic Review of the Literature. J Clin Med. 2024;13:1299.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
101.  Tessier AJ, Chevalier S. An Update on Protein, Leucine, Omega-3 Fatty Acids, and Vitamin D in the Prevention and Treatment of Sarcopenia and Functional Decline. Nutrients. 2018;10:1099.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 78]  [Cited by in RCA: 77]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
102.  He W, Connolly ED, Cross HR, Wu G. Dietary protein and amino acid intakes for mitigating sarcopenia in humans. Crit Rev Food Sci Nutr. 2025;65:2538-2561.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Reference Citation Analysis (0)]
103.  Huang HH, Wang YJ, Jiang HY, Yu HW, Chen YQ, Chiou A, Kuo JC. Sarcopenia-related changes in serum GLP-1 level affect myogenic differentiation. J Cachexia Sarcopenia Muscle. 2024;15:1708-1721.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
104.  Kong J, Park S, Kim TH, Lee JE, Cho H, Oh J, Lee S, Jo H, Lee H, Lee K, Park J, Jacob L, Pizzol D, Rhee SY, Kim S, Yon DK. Worldwide burden of antidiabetic drug-induced sarcopenia: An international pharmacovigilance study. Arch Gerontol Geriatr. 2025;129:105656.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
105.  Deng F, Wu W, Fan X, Zhong X, Wang N, Wang Y, Pan T, Du Y. Dulaglutide Protects Mice against Diabetic Sarcopenia-Mediated Muscle Injury by Inhibiting Inflammation and Regulating the Differentiation of Myoblasts. Int J Endocrinol. 2023;2023:9926462.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
106.  Jiao R, Lin C, Cai X, Wang J, Wang Y, Lv F, Yang W, Ji L. Characterizing body composition modifying effects of a glucagon-like peptide 1 receptor-based agonist: A meta-analysis. Diabetes Obes Metab. 2025;27:259-267.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
107.  Xiang J, Qin L, Zhong J, Xia N, Liang Y. GLP-1RA Liraglutide and Semaglutide Improves Obesity-Induced Muscle Atrophy via SIRT1 Pathway. Diabetes Metab Syndr Obes. 2023;16:2433-2446.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
108.  Linge J, Birkenfeld AL, Neeland IJ. Muscle Mass and Glucagon-Like Peptide-1 Receptor Agonists: Adaptive or Maladaptive Response to Weight Loss? Circulation. 2024;150:1288-1298.  [PubMed]  [DOI]  [Full Text]
109.  Neeland IJ, Linge J, Birkenfeld AL. Changes in lean body mass with glucagon-like peptide-1-based therapies and mitigation strategies. Diabetes Obes Metab. 2024;26 Suppl 4:16-27.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
110.  Ghanim H, Chaudhuri A, Hejna J, Dandona P. 12608 Preserving Lean Body Mass During Weight Loss In Elderly Obese Patients With Glp-1 Receptor Agonist Treatment. J Endocr Soc. 2024;8.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
111.  Nunn E, Jaiswal N, Gavin M, Uehara K, Stefkovich M, Drareni K, Calhoun R, Lee M, Holman CD, Baur JA, Seale P, Titchenell PM. Antibody blockade of activin type II receptors preserves skeletal muscle mass and enhances fat loss during GLP-1 receptor agonism. Mol Metab. 2024;80:101880.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
112.  Mastaitis J, Gomez D, Le Rouzic V, Stec M, Khan N, Na E, Mcwilliams T, Min S, Sleeman M. 207-OR: Myostatin Inhibition Synergizes with GLP-1R Agonism to Accelerate Weight Loss in Male, Obese Nonhuman Primates. Diabetes. 2023;72.  [PubMed]  [DOI]  [Full Text]
113.  Jianping W, Xuelian Z, Anjiang W, Haiying X. Efficacy and Safety of Glucagon-like Peptide-1 Receptor Agonists in the Treatment of Metabolic Associated Fatty Liver Disease: A Systematic Review and Meta-analysis. J Clin Gastroenterol. 2021;55:586-593.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
114.  Njei B, Al-Ajlouni Y, Lemos SY, Ugwendum D, Ameyaw P, Njei LP, Boateng S. Efficacy and Safety of GLP-1 Receptor Agonists in Patients With Metabolic Dysfunction-Associated Steatotic Liver Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Cureus. 2024;16:e71366.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
115.  Forlano R, Malik H, Mullish BH, Yee M, Izzi‐engbeaya C, Manousou P. The beneficial hepatic effects of glucagon‐like peptide 1 receptor agonists in patients with diabetes and metabolic dysfunction‐associated steatotic liver disease are independent of weight loss. Liver Int. 2024;5.  [PubMed]  [DOI]  [Full Text]
116.  Iwamoto Y, Kimura T, Dan K, Iwamoto H, Sanada J, Fushimi Y, Katakura Y, Shimoda M, Yamasaki Y, Nogami Y, Shirakiya Y, Nakanishi S, Mune T, Kaku K, Kaneto H. Tirzepatide, a dual glucose-dependent insulinotropic polypeptide/glucagon-like peptide 1 receptor agonist, exhibits favourable effects on pancreatic β-cells and hepatic steatosis in obese type 2 diabetic db/db mice. Diabetes Obes Metab. 2024;26:5982-5994.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
117.  Sawamura T, Mizoguchi R, Ohmori A, Kometani M, Yoneda T, Karashima S. Effects of the switch from dulaglutide to tirzepatide on glycemic control, body weight, and fatty liver: a retrospective study. J Diabetes Metab Disord. 2024;23:2105-2113.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
118.  Loomba R, Hartman ML, Lawitz EJ, Vuppalanchi R, Boursier J, Bugianesi E, Yoneda M, Behling C, Cummings OW, Tang Y, Brouwers B, Robins DA, Nikooie A, Bunck MC, Haupt A, Sanyal AJ; SYNERGY-NASH Investigators. Tirzepatide for Metabolic Dysfunction-Associated Steatohepatitis with Liver Fibrosis. N Engl J Med. 2024;391:299-310.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 193]  [Cited by in RCA: 90]  [Article Influence: 90.0]  [Reference Citation Analysis (0)]