Next Article in Journal
Macular Pigment Optical Density as a Measurable Modifiable Clinical Biomarker
Next Article in Special Issue
Body Composition in Cases with Normal Alanine Aminotransferase Values in Medical Health Checkups
Previous Article in Journal
Effectiveness of Prebiotics and Mediterranean and Plant-Based Diet on Gut Microbiota and Glycemic Control in Patients with Prediabetes or Type 2 Diabetes: A Systematic Review and Meta-Analysis
Previous Article in Special Issue
Comparative Efficacy of Different Protein Supplements on Muscle Mass, Strength, and Physical Indices of Sarcopenia among Community-Dwelling, Hospitalized or Institutionalized Older Adults Undergoing Resistance Training: A Network Meta-Analysis of Randomized Controlled Trials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Emerging Targets and Treatments for Sarcopenia: A Narrative Review

1
Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
2
Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
3
Department of Medicine and Surgery, LUM University, Strada Statale 100 Km 18, 70100 Casamassima, Italy
*
Authors to whom correspondence should be addressed.
Nutrients 2024, 16(19), 3271; https://doi.org/10.3390/nu16193271
Submission received: 2 September 2024 / Revised: 23 September 2024 / Accepted: 26 September 2024 / Published: 27 September 2024

Abstract

:
Background: Sarcopenia is characterized by the progressive loss of skeletal muscle mass, strength, and function, significantly impacting overall health and quality of life in older adults. This narrative review explores emerging targets and potential treatments for sarcopenia, aiming to provide a comprehensive overview of current and prospective interventions. Methods: The review synthesizes current literature on sarcopenia treatment, focusing on recent advancements in muscle regeneration, mitochondrial function, nutritional strategies, and the muscle–microbiome axis. Additionally, pharmacological and lifestyle interventions targeting anabolic resistance and neuromuscular junction integrity are discussed. Results: Resistance training and adequate protein intake remain the cornerstone of sarcopenia management. Emerging strategies include targeting muscle regeneration through myosatellite cell activation, signaling pathways, and chronic inflammation control. Gene editing, stem cell therapy, and microRNA modulation show promise in enhancing muscle repair. Addressing mitochondrial dysfunction through interventions aimed at improving biogenesis, ATP production, and reducing oxidative stress is also highlighted. Nutritional strategies such as leucine supplementation and anti-inflammatory nutrients, along with dietary modifications and probiotics targeting the muscle–microbiome interplay, are discussed as potential treatment options. Hydration and muscle–water balance are emphasized as critical in maintaining muscle health in older adults. Conclusions: A combination of resistance training, nutrition, and emerging therapeutic interventions holds potential to significantly improve muscle function and overall health in the aging population. This review provides a detailed exploration of both established and novel approaches for the prevention and management of sarcopenia, highlighting the need for further research to optimize these strategies.

1. Introduction

Sarcopenia is a progressive and generalized skeletal muscle disease characterized by accelerated loss of muscle mass, strength, and physical function [1]. While its essential components and operational definitions are still under debate [2], it has been clarified that sarcopenia is not part of normal aging, rather a distinct condition that requires clinical attention and intervention [3,4]. Sarcopenia increases the risk of physical disability and impacts the quality of life, independence, and overall survival of affected individuals [5,6,7]. To date, the global prevalence rates of sarcopenia and severe sarcopenia in adults over the age of 60 are 8–36% and 2–9%, respectively [8]. However, these percentages are expected to rise in the coming years, making it a major public health concern [9]. Currently, pharmacological interventions for sarcopenia remain limited [10], and therapeutic approaches are mainly based on nutritional interventions and physical exercise [11,12]. However, recent developments in the understanding of the pathophysiological pathways of sarcopenia have led to the discovery of specific biomarkers and possible targets for its prevention and treatment (Figure 1) [13,14]. The aim of this narrative review is to provide a comprehensive appraisal of emerging targets and potential treatments for sarcopenia. After discussing the current evidence across various aspects of sarcopenia treatment, each section provides a summary of findings along with current clinical practices and recommendations for managing sarcopenia.

2. Literature Search

This narrative review was conducted through a comprehensive search of peer-reviewed literature on sarcopenia and its emerging treatments. Major databases, including PubMed, Scopus, and Web of Science, were used to identify relevant studies published up to 31 August 2024. The search strategy focused on broad keywords such as “sarcopenia”, “muscle regeneration”, “mitochondrial dysfunction”, “muscle-water balance”, “muscle-microbiota interplay”, “nutrient flow”, “anabolic resistance”, and “neuromuscular junction”, or related keywords. As a narrative review, this article aimed to provide a broad overview rather than a systematic synthesis of the evidence, and priority was given to randomized controlled trials, meta-analyses, systematic reviews, and high-impact studies. Inclusion criteria were (1) articles published in English and (2) studies involving human subjects, particularly older adults. Exclusion criteria were (1) articles with unclear methodologies or insufficient clinical data, or (2) preclinical studies without clear translational potential. The review was further refined by cross-referencing citations from included articles to ensure comprehensive coverage of the most recent advancements in the field.

3. Muscle Regeneration in Sarcopenia

Muscle regeneration is a critical process in maintaining muscle mass and function, and it is often impaired in sarcopenia [15,16]. Muscle regeneration involves the activation, proliferation, and differentiation of myosatellite cells (MSCs), which are specialized stem cells of the muscle tissue. Upon injury or stress, MSCs are activated and proliferate to repair and regenerate damaged muscle fibers. Several signaling pathways are involved in regulating MSC activity and muscle regeneration. For instance, insulin-like growth factor-1 (IGF-1) has been shown to stimulate MSC proliferation and differentiation through the activation of the phosphoinositide 3-kinase (PI3K)/Akt pathway, which promotes protein synthesis and inhibits apoptosis [17]. Conversely, myostatin, a member of the transforming growth factor-beta (TGF-β) superfamily, acts as a negative regulator of muscle growth by inhibiting satellite cell activation and proliferation [17].
Research has focused on understanding the relationship between these signaling pathways and their possible therapeutic applications. Targeting myostatin signaling emerged as a promising strategy to enhance muscle regeneration [18]. Monoclonal antibodies and receptor antagonists targeting the myostatin signaling pathway have been shown to increase muscle mass and strength in preclinical models [19]. Such molecules may act as direct inhibitors of myostatin, inhibitors of myostatin activation, and blockers of the receptors involved in myostatin signaling [20]. Bimagrumab (Novartis/Versanis) acts as an agonist by binding to activin type 2 receptors and preventing ligands from binding and activating these receptors. By blocking the binding site of the activin type 2 receptor, it increases circulating levels of ligands (e.g., bone morphogenetic protein (BMP)-9 and BMP-10) that can bind to other receptors, such as BMP receptor 2, thereby further promoting muscle growth and counteracting the effects of myostatin/activin A signaling [20]. Clinical trials are ongoing to evaluate the efficacy and safety of myostatin inhibitors in older adults with sarcopenia; however, preliminary results indicate inconsistent benefits in improving muscle function and reducing the occurrence of physical frailty and disability [21,22,23,24,25,26].
Another area of research focuses on the role of chronic inflammation in the impairment of MSC activity and muscle regeneration [27]. The suppression of MSC proliferation and differentiation has been linked to the action of proinflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) [28,29]. Conversely, anti-inflammatory therapies, including the use of cytokine inhibitors and non-steroidal anti-inflammatory drugs (NSAIDs), have shown potential in enhancing muscle regeneration and reducing muscle loss associated with sarcopenia [30,31]. However, significant side-effects of NSAIDs in older adults, such as gastrointestinal and cardiovascular complications, pose a challenge for their widespread use in clinical practice [32].
Together with MSCs, the extracellular matrix (ECM) is an important component in muscle regeneration. The ECM provides structural support and regulates the availability of growth factors and signaling molecules necessary for MSC function. Remodeling of the ECM is essential for efficient muscle repair, and a dysregulation of ECM components has been linked to impaired regeneration in sarcopenia [33,34]. Preliminary results indicate that therapeutic strategies aimed at modulating ECM composition and function, such as the use of matrix metalloproteinase inhibitors, may be beneficial to enhance muscle regeneration [35,36]; however, further studies are required in this field.
Another area of active investigation involves the role of the MSC niche in muscle regeneration. The niche, comprising various cell types and extracellular components surrounding MSCs, provides critical signals that regulate MSC behavior. Alterations in the niche, such as changes in cell composition or ECM structure, can impact MSCs and muscle regeneration [37,38]. Strategies aimed at restoring the niche environment, such as cell-based therapies or bioengineered scaffolds, are being explored to improve muscle regeneration in the setting of sarcopenia [39,40,41]. However, they are still in early stages of research, and further studies are required to determine their efficacy and safety in clinical settings.
Advances in gene editing technologies, particularly CRISPR/Cas9, have opened new possibilities for muscle regeneration therapies. Gene editing can be used to correct genetic mutations that impair muscle function or to enhance the expression of genes involved in muscle growth and regeneration. For instance, CRISPR/Cas9-mediated knockout of myostatin has been demonstrated to increase muscle mass and strength in animal models [42]. Translating these findings to clinical applications holds promise for developing novel treatments for sarcopenia and other muscle-wasting conditions, with a possible role of RNA-based gene therapy in the coming decades [43]. While preclinical findings are promising, translating these technologies to clinical applications remains challenging and raises ethical considerations.
Stem cell therapy is another promising approach for enhancing muscle regeneration leveraging on stem cell populations other than MSCs [43]. Mesenchymal stem cells can differentiate into myogenic cells and secrete paracrine factors that promote MSC activity and muscle repair [44]. Induced pluripotent stem cells derived from reprogrammed somatic cells offer the advantage of generating patient-specific stem cells for autologous transplantation, reducing the risk of immune rejection [45]. Recent clinical trials have shown promising results [46,47]; however, further studies are needed to evaluate the safety and efficacy of leveraging stem cells for sarcopenia and physical frailty in humans.
Emerging research is also exploring the role of microRNAs (miRNAs) in regulating muscle regeneration. miRNAs are small non-coding RNAs that modulate gene expression by targeting messenger RNAs for degradation or translational repression. Several miRNAs have been identified as key regulators of MSC function and muscle regeneration. For instance, miR-1 and miR-206 promote myogenic differentiation, while miR-489 maintains satellite cell quiescence [48]. Therapeutic modulation of miRNA expression, using miRNA mimics or inhibitors, offers a novel approach to enhance muscle regeneration and treat sarcopenia [48]; yet, extensive research is needed to explore its potential clinical applications.
More conventional approaches that rely on diet (Table 1) and physical activity (Table 2) have been shown to promote muscle regeneration.
Adequate protein intake is essential for providing the building blocks necessary for muscle repair and growth [11,49], while a balanced diet rich in antioxidants and anti-inflammatory nutrients can help reduce inflammation and promote muscle rejuvenation [52,64]. Specific amino acids, such as leucine, have been shown to stimulate muscle protein synthesis (MPS) and enhance MSC activity [49,50,65]. β-hydroxy-β-methyl butyrate (HMB) is a metabolite of the amino acid leucine, recognized for its role in promoting muscle health. HMB modulates chronic inflammation by reducing the levels of pro-inflammatory cytokines and inhibiting the nuclear factor kappa-light-chain-enhancer of the activated B cells (NF-κB) signaling pathway, which is crucial in the inflammatory response [54]. Moreover, HMB enhances the proliferation and differentiation of MSCs [54]. The anti-inflammatory and pro-regeneration effects of HMB are particularly beneficial in conditions of muscle wasting and chronic diseases [55]. In addition, nutritional supplements, including whey protein, creatine and omega-3 fatty acids, have been studied for their potential to support muscle recovery and counteract sarcopenia [66]. Exercise induces mechanical stress on muscle fibers, which triggers the activation of various signaling pathways involved in muscle regeneration. Resistance training, in particular, has been shown to activate MSCs, increase MPS, and enhance muscle strength and mass [67]. Combining exercise with other therapeutic interventions, such as nutritional supplementation or pharmacological agents, may provide synergistic benefits in promoting muscle regeneration and improving functional outcomes in sarcopenia [68]. Adherence to exercise programs, however, can be challenging, especially for older individuals with limited mobility or other health conditions. The optimal combination of exercise with nutritional or pharmacological interventions also requires further investigation to identify the most effective approaches for different patient populations. Additionally, while exercise is a powerful tool for combating sarcopenia, it may not be sufficient for severe sarcopenia, which might require additional interventions.
Table 2. Exercise interventions and outcomes in sarcopenia.
Table 2. Exercise interventions and outcomes in sarcopenia.
Exercise
Intervention
Types of
Exercise
Effects on
Muscle Mass
Effects on
Muscle Strength
Effects on
Muscle
Function
Synergistic Effects with Other
Therapies
References
Resistance trainingWeightlifting, resistance bands, bodyweight exercisesIncreases muscle massIncreases muscle strength, especially in large muscle groupsEnhances functional performance in daily activities (e.g., walking, stair climbing)Synergistic with protein supplementation and HMB in promoting muscle growth and strength[67,68]
Aerobic exerciseWalking, cycling, swimmingMay slightly increase muscle massImproves muscle endurance, with minor strength gainsEnhances cardiovascular fitness and overall functional capacityMay complement resistance training by improving mitochondrial function[69,70]
Combined resistance and aerobic trainingAlternating or concurrent resistance and aerobic workoutsIncreases muscle mass, potentially more than aerobic exercise aloneImproves both strength and enduranceEnhances both muscular and cardiovascular function, leading to improved overall fitnessSynergistic with dietary interventions such as protein or creatine supplementation for comprehensive muscle health benefits[70,71]
High-intensity
interval training
Short bursts of intense exercise followed by restMay increase muscle mass, especially in untrained individualsIncreases strength and endurance, depending on the intensityEnhances functional capacity, especially in tasks requiring both power and enduranceSynergistic with creatine supplementation for enhancing both strength and recovery[72]
Flexibility and
balance training
Yoga, tai chi, stretchingMarginal direct effects on muscle massMay improve strength through enhanced muscle controlImproves balance, reducing the risk of falls in older adultsComplements other exercise modalities by improving mobility and reducing injury risk during resistance training[73]
Abbreviation: HMB, β-hydroxy β-methylbutyrate.

Summary of Evidence and Current Clinical Recommendations

Muscle regeneration relies on the activation and proper function of MSCs, which are key to muscle repair. The IGF-1/PI3K/Akt and myostatin pathways regulate MSC activity. Although targeting myostatin has shown promise in preclinical models, clinical results are inconsistent. Chronic inflammation also impairs MSC function and, while anti-inflammatory therapies have potential, their use is limited by side-effects. Emerging treatments, like gene editing and stem cell therapy, hold promise but are still experimental.
Resistance training and adequate protein intake remain the primary interventions for supporting muscle regeneration in older adults. Myostatin inhibitors and anti-inflammatory therapies are not yet recommended due to inconsistent efficacy and potential risks. Future therapies involving stem cells and gene editing require more research before being applied in clinical settings.

4. Mitochondrial Dysfunction and Energy Enhancers in Sarcopenia

Mitochondrial dysfunction is one of the hallmarks of aging [74] and one of the main drivers in the development of age-related sarcopenia [75]. In the setting of sarcopenia, mitochondrial failure, mostly involving the ability to replace defective mitochondria, decreased adenosine triphosphate (ATP) generation, and increased oxidative stress, is a significant contributor to muscle weakness and fatigue [76]. The aging process itself induces several changes in mitochondrial quality control, including a decrease in mitochondrial biogenesis, alterations in mitochondrial DNA, and an increase in the production of reactive oxygen species [77]. These changes impair the ability of muscle cells to generate and sustain energy, highlighting the critical need for interventions that enhance mitochondrial function and energy metabolism.
Potential targets for energy enhancement in the management of sarcopenia focus on improving mitochondrial biogenesis, ATP production, and overall mitochondrial quality control. The peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), a key regulator of mitochondrial biogenesis, enhances the production of new mitochondria, improves mitochondrial function, and increases oxidative capacity [78]. Similarly, sirtuins, NAD+-dependent deacetylases like SIRT1 and SIRT3, regulate mitochondrial biogenesis and function, and enhancing their activity through pharmacological agents or calorie restriction mimetics holds promise for improving muscle energy metabolism [79]. Pharmacological sirtuin activators, such as resveratrol and NAD+ precursors like nicotinamide riboside, have demonstrated potential in enhancing mitochondrial function and reducing oxidative stress [49]. However, robust clinical evidence supporting the efficacy of these interventions in older adults with sarcopenia remains lacking. Many of these compounds are still in early stages of research, and their long-term safety and efficacy need further validation before they can be recommended in clinical practice.
ATP production and utilization can be ameliorated by creatine monohydrate, an amino acid derivative that facilitates rapid ATP production by donating a phosphate group to adenosine diphosphate, particularly beneficial in short-duration, high-intensity activities [56]. Adenosine monophosphate-activated protein kinase (AMPK), a cellular energy sensor, maintains energy homeostasis by enhancing glucose uptake, fatty acid oxidation, and mitochondrial biogenesis, with pharmacological activators like metformin and 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) being explored for their potential to enhance muscle energy metabolism and combat sarcopenia [69,80,81]. Despite these benefits, creatine and AMPK activators are not effective for all older adults. The individual variability in response to these supplements, as well as potential side-effects (such as gastrointestinal discomfort from creatine or the risk of hypoglycemia with metformin), limits their widespread application. Additionally, further clinical trials are needed to confirm their long-term benefits specifically in older adults with sarcopenia.
Mitochondrial quality control can be improved through mitophagy, the selective autophagic degradation of damaged mitochondria, which may be promoted via pharmacological agents or lifestyle interventions (e.g., exercise) [82]. Additionally, antioxidants like coenzyme Q10 (CoQ10), a key component of the mitochondrial electron transport chain, and N-acetylcysteine (NAC) mitigate oxidative damage, preserve mitochondrial integrity, and improve energy production [61,83]. However, clinical data supporting the use of antioxidants in sarcopenia are not robust, and more rigorous trials are necessary to establish their long-term efficacy. Additionally, while mitophagy is a theoretical target for enhancing mitochondrial quality, practical interventions that effectively enhance mitophagy in humans are still in the experimental phase.
l-carnitine, essential for transporting long-chain fatty acids into mitochondria for β-oxidation, improves muscle function, reduces fatigue, and boosts exercise performance [84]. Branched-chain amino acids (BCAAs), particularly leucine, stimulate MPS, improve muscle mass, and reduce muscle breakdown, while also serving as an energy source during prolonged exercise [51]. l-carnitine and BCAAs have demonstrated beneficial effects in terms of reducing muscle fatigue and enhancing muscle recovery, particularly when used in combination with exercise interventions. Notwithstanding these findings, more studies are needed to better understand the optimal dosages and long-term impacts of these supplements.
Regular exercise, especially resistance and endurance training, promotes mitochondrial biogenesis, improves oxidative capacity, and enhances insulin sensitivity, contributing to better muscle function and overall health [70]. Hormonal therapies, including testosterone, growth hormone, and IGF-1, have shown potential in improving muscle mass, strength, and function [85]. Yet, the use of hormonal therapies requires careful consideration of risks and benefits. Hormone replacement can lead to serious side-effects, including cardiovascular events and increased cancer risk.

Summary of Evidence and Current Clinical Recommendations

Mitochondrial dysfunction, characterized by reduced ATP production and increased oxidative stress, is considered to be a key factor in muscle weakness in sarcopenia. Enhancing mitochondrial biogenesis and improving mitochondrial quality through pathways like PGC-1α and sirtuins can potentially improve muscle energy metabolism. Pharmacological activators of sirtuins and AMPK, along with supplements such as creatine and CoQ10, have shown promise in early research but lack robust clinical data.
Physical exercise, particularly resistance and aerobic training, remains the most effective intervention for addressing mitochondrial dysfunction in older adults. Supplements like creatine and CoQ10 may offer additional benefits but require further validation. Pharmacological agents targeting mitochondrial function are still under development and not recommended in clinical practice.

5. Hydration and Muscle–Water Balance in Sarcopenia

Hydration is essential in cellular processes, nutrient transport, and biochemical reactions within muscle tissue, and both intracellular (ICW) and extracellular water (ECW) content play significant roles in muscle function and health [86]. ICW, which constitutes a large portion of muscle mass, is essential for maintaining cellular homeostasis, protein synthesis, and metabolic functions. Conversely, ECW, which is primarily found in the interstitial spaces and blood, supports nutrient transport and waste removal. The balance between these two compartments is finely regulated by various molecular mechanisms involving aquaporins, ion channels, and transporters [87]. Dysregulation of this balance can lead to muscle atrophy, impaired muscle contractility, and overall muscle weakness, all of which are hallmark features of sarcopenia [86,88,89,90].
ICW content in lean mass has been proposed as a marker of muscle quality and cellular hydration. A growing body of evidence suggest that ICW is associated with strength, physical function, and frailty [86]. During aging, a relative expansion of ECW compared to ICW accompanies the reduction in muscle mass [91]. The loss of ICW is associated with activation of catabolic processes and concomitant inhibition of the mechanistic target of rapamycin complex 1 (mTORC1) and insulin-stimulated anabolic pathways [92]. In a cohort of community-dwelling individuals aged 65–90 years, the ratio of ECW to ICW in the thigh was found to be negatively associated with knee extension strength and walking speed [93]. This finding suggests that cellular hydration may play a protective role against weakness and functional decline. However, it is possible that the observed reduction in ICW with aging is primarily due to a decrease in muscle mass, raising the question of whether the beneficial effects of higher ICW should be attributed to a greater muscle quantity or enhanced muscle quality through improved cellular hydration. The same researchers proposed a new indicator of muscle quality, defined as ICW content per unit of lean mass (mL/kg). This ratio was observed to decrease with age and to be positively correlation with muscle strength and function, and negatively related with frailty risk, irrespective of age, sex, and the number of diseases [86,89].
ICW content is closely linked to muscle cell volume and function. Aquaporins, a family of water channel proteins, facilitate the transport of water across cell membranes and are crucial for maintaining intracellular hydration. Among them, aquaporin-4 (AQP4) is predominantly expressed in skeletal muscle and plays a significant role in muscle physiology. Changes in AQP4 expression or function can disrupt ICW homeostasis, leading to muscle fiber swelling or shrinkage, which subsequently affects muscle strength and endurance [94]. Ion channels, such as sodium-potassium pumps (Na+/K+ ATPase) and chloride channels, also contribute to water movement by creating osmotic gradients that drive water into or out muscle cells [95]. Dysfunctions in these ion channels can alter intracellular osmolality, thereby impacting muscle cell hydration and function [96].
ECW balance is equally vital for muscle health. It ensures the proper delivery of oxygen and nutrients to muscle fibers and the removal of metabolic byproducts. The ECM, composed of collagen, elastin, and proteoglycans, is essential for regulating ECW content and preserving muscle tissue structure. Proteoglycans, in particular, have a high affinity for water and contribute to the viscoelastic properties of the ECM. In the setting of sarcopenia, changes in ECM composition and function can lead to increased ECW retention, edema, and inflammation, exacerbating muscle degeneration [34]. However, changes in ECM composition that contribute to ECW retention and inflammation are difficult to reverse, particularly in older adults with advanced sarcopenia. While hydration strategies may be useful, they may not fully counteract the detrimental effects of ECM dysregulation.
Several molecular mechanisms underpin the regulation of muscle–water balance. The mTORC signaling pathway is a key regulator of muscle cell growth and protein synthesis, and it also influences cellular hydration status. mTORC activity is sensitive to changes in cellular hydration, with hypo-osmotic stress (cell swelling) activating mTORC signaling, while hyper-osmotic stress (cell shrinkage) inhibits it. This dynamic response helps coordinate protein synthesis with cellular hydration state, ensuring optimal muscle function [82].
Maintaining proper hydration is essential for muscle function and overall health, especially in older adults at risk for sarcopenia. The European guidelines recommend older persons to ingest a daily water intake of 1.6 L for women and 2.0 L for men to maintain optimal hydration [60]. These recommendations aim to counteract the decreased thirst sensation and reduced renal function often observed in older adults, which can lead to insufficient fluid intake and dehydration. To achieve the necessary hydration levels, an integrated approach is required, including lifestyle modifications, dietary modifications, and the use of nutritional supplements to improve water intake and retention.
Supplementation with electrolytes and nutrients that support cellular hydration can also help maintain muscle–water balance and improve muscle performance. Beyond its role in energy metabolism, creatine helps retain ICW, supporting proper muscle hydration [57]. BCAAs, especially leucine, support MPS and hydration. They can also serve as an energy source during prolonged exercise, reducing muscle fatigue and supporting hydration [97]. Resistance training has been shown to increase muscle mass and strength, partly by enhancing ICW content and promoting favorable changes in muscle cell volume [71,72].
Medical interventions, including treatments that modulate aldosterone and antidiuretic hormone (ADH) pathways, may also help manage hydration levels in older adults who struggle to maintain adequate fluid balance [98,99].

Summary of Evidence and Current Clinical Recommendations

Proper hydration is crucial for muscle function. Aging leads to a reduction in ICW and an increase in ECW, contributing to muscle weakness and functional decline. Dysregulation of muscle–water balance impairs muscle metabolism, and factors such as aquaporin function and mTOR signaling are involved in maintaining hydration status.
Older adults are recommended to maintain adequate hydration, with daily water intake guidelines set at 1.6 L for women and 2.0 L for men. Resistance training and supplements like creatine may help support muscle hydration. Electrolyte supplementation and interventions targeting fluid regulation, such as those affecting aldosterone and ADH pathways, may benefit individuals who have difficulties with maintaining fluid balance.

6. Muscle–Microbiota Interplay in Sarcopenia

The human gut harbors a complex community of microorganisms known as the gut microbiota, which maintains a symbiotic and commensal relationship with the host [100]. The gut microbiota plays essential roles in the health of the host. It aids in nutrient absorption, synthesizes essential vitamins, regulates the immune system, and protects against pathogens. Additionally, it contributes to the host’s metabolism, assists in mineral absorption, regulates fat storage, activates bioactive compounds, and serves as an endocrine and neuronal organ [101,102,103].
The human gut microbiota undergoes distinct transformations across different life stages, characterized by rapid changes in infancy, stability in adulthood, and shifts in old age [104,105]. Despite advancements, understanding the timing and effects of these age-related changes on host physiology remains challenging. Recent research suggests that alterations in the gut microbiome of centenarians and older individuals may be adaptations to aging rather than deviations from a healthy microbial profile [106], holding clinical significance for diagnostics and personalized microbiota modulation.
Inflammaging, immune senescence, and dietary modifications contribute to changes in gut microbiota composition and function in older adults, especially in frailer individuals [107]. Studies consistently show a decline in microbial stability and diversity, accompanied by an increase in pro-inflammatory microbes [106]. In the ELDERMET study, researchers identified a predominant presence of the genera Bacteroides, Alistipes, and Parabacteroides in individuals aged 65 years and older, in comparison to healthy younger controls [108]. Furthermore, significant differences were observed between community-dwelling older adults and long-term care residents [109]. Also, frail individuals in the ELDERMET study displayed elevated levels of lithocholic acid and deoxycholic acid compared with non-frail counterparts [110]. Ghosh et al. [110] found that individuals over 60 years of age exhibited greater variation in gut microbiota composition than younger individuals. Specifically, the gut microbiota of older individuals tends to have lower levels of Firmicutes, including Clostridium and Faecalibacterium, as well as Bifidobacterium. Conversely, there is an increase in Proteobacteria and Streptococcaceae, which is associated with exacerbations in inflammation, frailty, and multimorbidity [111]. These microbial shifts have been linked to sarcopenia-related muscle loss and functional decline. In contrast, the presence of different microbial clusters may correspond with enhanced cardiovascular and metabolic health, possibly acting as indicators of healthy aging and longevity [112]. Furthermore, age-related changes in the gut microbiota impact its metabolic potential, with increased proteolytic activity and variations in compound degradation, which are most likely driven by lifestyle and environmental exposures [111].
Recent studies suggest a possible role of the gut microbiota in the pathophysiology of muscle loss [113,114,115,116]. In a cross-sectional analysis of 728 female twins in a community setting, higher frailty status correlated with decreased levels of anti-inflammatory short-chain fatty acids (SCFAs)-producing microbes like Faecalibacterium prausnitzii and an increase in pro-inflammatory Enterobacteriaceae [117]. Frailer individuals also exhibited heightened lipopolysaccharide synthesis, indicating an elevated local and systemic pro-inflammatory state [118]. The ELDERMET study found that institutionalization and muscle loss were associated with a dysbiosis, reduced anti-inflammatory microbial metabolites such as butyrate, and elevated levels of circulating pro-inflammatory markers like TNF-α, IL-6, IL-8, and C-reactive protein (CRP) [109]. Pro-inflammatory bacteria, including Eggerthella lenta, were found in frail patients, possibly contributing to sarcopenia [117]. Sarcopenic and frail individuals exhibited a similar gut microbial profile, marked by enrichment in Oscillospira and Ruminococcus, and reductions in members of Barnesiellaceae and Christenellaceae families [119]. Animal studies support these findings, showing that butyrate administration could help restore lean muscle mass [120]. Conversely, medications like proton pump inhibitors and NSAIDs, commonly used by older adults, were associated with dysbiosis, potentially exacerbating frailty and its complications [111]. While animal studies on butyrate administration are promising, clinical evidence in humans remains limited. The optimal way to increase SCFA production through diet or supplementation is not yet well established, and the long-term effects of manipulating microbiota for muscle health require more extensive trials in older populations.
Research was carried out to investigate treatments, such as diet modifications, prebiotics, and probiotics, aimed at restoring a balanced composition of gut microbiota. The NU-AGE project demonstrated that a Mediterranean diet intervention in older adults led to increased levels of beneficial microbial communities, which was associated with reduced frailty and improved cognitive function, along with decreased inflammatory markers such as CRP and IL-17. Additionally, there was an increase in the circulating levels of SCFAs/branched-chain fatty acids and a decrease in the production of secondary bile acids, p-cresols, ethanol, and carbon dioxide [121]. Similarly, dietary supplementation with galacto-oligosaccharides showed promise in restoring beneficial Bifidobacterium spp. levels in prefrail older individuals [122]. Increasing dietary fiber intake is highlighted as a beneficial strategy for preventing age-related decline in skeletal muscle mass and strength [111]. Interventional studies, like one led by Lee et al. [123], observed improvements in handgrip strength and overall functional performance in frail older adults following a minimum of six weeks of continuous L. plantarum supplementation. While diet can positively influence gut microbiota, individual responses to dietary changes vary significantly. Moreover, the long-term effects of microbiota-targeted dietary interventions on muscle health are not fully understood. Additional longitudinal studies are necessary to determine the most effective dietary strategies for maintaining muscle mass in older adults. Further research with larger cohorts and diverse microbiome analysis methods is needed to fully understand and utilize gut microbiota modulation for promoting healthy aging and counteracting sarcopenia.

Summary of Evidence and Current Clinical Recommendations

The gut microbiota impacts muscle health by influencing nutrient absorption, inflammation, and metabolism. In older adults, changes in gut microbiota composition, often marked by a reduction in beneficial bacteria and an increase in pro-inflammatory microbes, contribute to the onset of sarcopenia and frailty.
Dietary measures, particularly the Mediterranean diet and an increased fiber intake, as part of multimodal interventions, improve muscle function and reduce the risk of frailty. Prebiotic and probiotic supplementation may offer additional benefits, but further research is needed to confirm their effectiveness in targeting muscle loss.

7. Nutrient Flow in Sarcopenia

Nutrient flow refers to the delivery and utilization of essential nutrients that support muscle health, including proteins, amino acids, vitamins, and minerals. A proper nutrient flow is crucial for maintaining muscle protein synthesis (MPS) and preventing muscle protein breakdown (MPB) [124]. Disruptions in nutrient flow, whether due to inadequate dietary intake, impaired digestion, or altered nutrient metabolism and delivery contribute to the pathogenesis of sarcopenia [125,126].
Endothelial cells and smooth muscle cells in microvascular units adapt to the metabolic demands of muscle fibers in response to insulin [127,128]. The muscle microvascular system undergoes structural and functional adaptations essential for regulating nutrient delivery and trans-endothelial transport processes, sustaining muscle metabolism [129]. Studies have demonstrated the critical role of the microvascular system in efficient nutrient delivery to muscles, highlighting the importance of maintaining vascular health for muscle regeneration. Metabolic–vascular coupling in skeletal muscle dynamically coordinates blood flow with metabolic demands, especially during muscle contraction [130]. Blood vessel dilation in response to increased metabolism ensures efficient oxygen and nutrient delivery and waste removal, supporting muscle function and performance [130]. During exercise, the transition from anaerobic to aerobic metabolism requires increased oxygen and nutrient supply, which are facilitated by enhanced blood flow to active muscle tissues [131]. Despite this knowledge, the long-term effects of impaired nutrient flow on muscle health in sarcopenia are not fully understood. Older individuals with cardiovascular disease or multimorbidity may show severe microvascular involvement. Therefore, interventions targeting the microvascular system have yet to demonstrate consistent efficacy in older adults with sarcopenia.
Studies have also explored the role of hormones like glucagon-like peptide 1 (GLP-1) and insulin in regulating the muscle microvascular system and nutrient delivery [132]. GLP-1 and insulin increase muscle endothelial surface area and promote the delivery of oxygen, nutrients, and hormones to muscle tissues, thereby optimizing nutrient transport for muscle function [132]. Regulating blood flow volume may help modulate anabolic resistance in muscle, emphasizing the importance of adequate blood flow for supporting muscle protein synthesis [133]. Age-related changes in muscle blood flow and nutrient transport can affect muscle protein metabolism and anabolic responses to exercise [134]. Impaired microcirculatory blood flow can contribute to anabolic resistance by limiting the delivery of essential amino acids to skeletal muscle, highlighting the importance of optimizing blood flow for muscle health [134]. Adequate blood flow is crucial for modulating MPS and MPB, as it facilitates the transport of metabolic precursors and products, underscoring its role in supporting muscle metabolism [135]. The ability of hormones like insulin and GLP-1 to regulate nutrient delivery and improve endothelial function offers a promising therapeutic pathway for combating sarcopenia. However, the effects of these hormones can be diminished in older adults, particularly in those with insulin resistance, making the benefits of hormone-based strategies less reliable in these populations.
Protein intake and their digestion into amino acids are vital for MPS. Essential amino acids, especially leucine, stimulate the mTORC signaling pathway, a key regulator of MPS. In sarcopenic individuals, the efficiency of this pathway can be compromised, leading to an imbalance between MPS and MPB [124]. Ensuring an adequate intake of high-quality protein, rich in essential amino acids, is thus a key strategy for combating sarcopenia. Studies have shown that older adults often have reduced anabolic responses to protein ingestion, a phenomenon known as anabolic resistance [136]. This resistance can be mitigated by consuming higher quantities of protein or by distributing protein intake evenly across meals to optimize MPS throughout the day [137]. Studies have consistently shown that adequate protein intake, particularly when rich in leucine, can significantly improve MPS and combat muscle loss. However, the effectiveness of protein supplementation is often limited by the degree of anabolic resistance in older adults, necessitating further research to optimize dosing strategies and determine the most effective approaches for mitigating MPB.
The molecular mechanisms underlying nutrient flow and its impact on sarcopenia are complex and involve multiple signaling pathways. The mTORC pathway, as previously mentioned, is central to the regulation of muscle growth and atrophy. In addition to mTORC, the AMPK pathway, which is activated under conditions of low energy availability, can inhibit MPS and promote MPB [138]. IGF-1 is another critical regulator that promotes muscle growth by activating the mTORC pathway and inhibiting the Forkhead box O (FoxO) transcription factors that drive muscle atrophy [139]. Nutrient deficiencies or imbalances can disrupt these signaling pathways, exacerbating muscle loss in sarcopenia. The mTORC and AMPK pathways offer key therapeutic targets for regulating muscle metabolism and promoting muscle health. Nevertheless, interventions that aim to modulate these pathways are still experimental and require more extensive clinical validation before they can be applied to older adults with sarcopenia.
Given the centrality of nutrient flow in the pathogenesis of sarcopenia, targeted nutritional interventions are a promising therapeutic approach. Potential approaches include enhancing protein intake, optimizing the intake of key micronutrients, and incorporating bioactive compounds that can modulate muscle metabolism. For instance, leucine supplementation has been shown to directly stimulate MPS [65,140], while HMB can reduce MPB [54]. Combined supplementation of protein and omega-3 fatty acids has been found to have synergistic effects on muscle health [53]. Some nitrate-rich foods and amino acids, such as arginine and beetroot juice, have vasodilatory properties through their action on the synthesis and/or bioavailability of nitric oxide (NO) [58,62]. Arginine serves as the primary substrate for endothelial NO synthase and is the key precursor for NO production within the vascular endothelium [58]. The consumption of nitrate-rich foods, such as beetroot juice, has been shown to enhance the bioavailability of NO through an alternative nitrate-nitrite-NO pathway, where nitrate is converted to nitrite by commensal bacteria in the oral cavity [63]. By elevating circulating NO levels, beetroot juice and arginine modulate mitochondrial respiration, increase the efficiency of oxygen use during exercise, improve muscle contractility, and enhance blood and nutrient supply to muscle [58,141]. Arginine has also been found to activate mTORC1 by binding to the cytosolic arginine sensor for the mTORC1 subunit 1 (CASTOR1) [59], thus stimulating MPS [142]. Beetroot juice is rich in biologically active substances with anti-inflammatory and antioxidant properties, including betalains, carotenoids, organic acids, and polyphenols, which have beneficial effects for muscle and vessel health [143]. Indeed, the consumption of beetroot juice has been demonstrated to improve endothelial function and tolerance of exercise in older individuals [144]. Nutritional strategies, such as the use of leucine, HMB, and beetroot juice, show promise in improving nutrient flow, muscle metabolism, and overall muscle health. These strategies offer non-invasive, low-risk interventions that may help mitigate muscle loss. However, the evidence for long-term benefits of these interventions in older adults is limited, and further research is needed to confirm their efficacy, particularly in clinical settings.
Exercise has been shown to promote skeletal muscle microvascular blood flow, improving nutrient transport and disposal in muscle tissues [145]. In contrast, high-glucose mixed-nutrient meals have been observed to restrict muscle microvascular blood flow, potentially contributing to acute hyperglycemia-induced insulin resistance [146].
Emerging research is also exploring the role of gut microbiota in nutrient flow and muscle health. Probiotics and prebiotics, which modulate the gut microbiota, are being investigated for their potential to improve nutrient absorption and muscle health [147].
Additionally, the concept of “nutritional periodization”, which involves adjusting nutrient intake to match the body’s needs at different times (such as pre- and post-exercise), is gaining attention as a strategy to maximize the anabolic response to nutrients [148,149].

Summary of Evidence and Current Clinical Recommendations

Nutrient flow, which ensures the delivery of essential nutrients like amino acids and glucose to muscles, is critical for maintaining MPS. Impaired nutrient delivery, often due to endothelial dysfunction and insulin resistance, contributes to anabolic resistance. Nutrient flow is regulated by pathways such as mTORC and AMPK, with leucine and other essential amino acids playing crucial roles in activating MPS.
In clinical practice, optimizing nutrient flow through increased protein intake and the consumption of leucine-rich foods is key to improving MPS. Exercise, especially resistance training, helps enhance blood flow and nutrient delivery. Supplements like arginine, beetroot juice, and omega-3 fatty acids may further support nutrient flow by improving vascular function.

8. Anabolic Resistance and Anabolic Regulators in Sarcopenia

Anabolic resistance is a critical element in sarcopenia and is characterized by a diminished MPS response to anabolic stimuli such as nutrient ingestion and exercise [150]. This phenomenon is attributed, at least partly, to disruptions in the signaling pathways essential for muscle growth and repair, notably the mTORC pathway [150]. Research indicates that older adults show decreased phosphorylation of mTORC and its downstream targets in response to anabolic signals, leading to diminished MPS and impaired muscle mass maintenance [150].
Several pathophysiological mechanisms underpin anabolic resistance, including insulin resistance, chronic low-grade inflammation, and mitochondrial dysfunction [151]. Studies have identified insulin resistance and inflammation as major contributors to anabolic resistance, providing key therapeutic targets for improving muscle health. However, the complex interplay of these mechanisms implies that addressing one factor alone may not fully overcome anabolic resistance in older adults. Insulin resistance, prevalent among older adults, impairs the muscle’s ability to respond to anabolic signals, thereby decreasing MPS [151,152]. Chronic low-grade inflammation, often referred to as “inflammaging”, involves elevated levels of proinflammatory cytokines such as TNF-α and IL-6, which interfere with anabolic signaling and exacerbate muscle degradation [153]. Mitochondrial dysfunction contributes by reducing energy production and increasing oxidative stress, impairing muscle cell function and regeneration [154].
Addressing anabolic resistance involves multiple strategies, including optimizing protein intake, with greater protein quantities per meal necessary to effectively stimulate MPS in older individuals [155]. Optimizing protein intake, particularly leucine-rich protein, has consistently been shown to enhance MPS in older adults, helping counteract the effects of anabolic resistance. Supplementation with essential amino acids, particularly leucine, has shown promise in enhancing MPS due to its role in activating the mTORC pathway [155]. Nevertheless, anabolic resistance remains a significant barrier, and higher protein intakes are often necessary to achieve the desired results, which may be difficult for some older adults to sustain.
Pharmacological approaches include myostatin inhibitors [20], which are currently undergoing clinical trials, and selective androgen receptor modulators (SARMs) [85,156], which promote muscle growth without the adverse effects of traditional anabolic steroids. These pharmacological interventions, particularly SARMs, offer a potentially safer alternative to traditional anabolic therapies. However, both myostatin inhibitors and SARMs are still in experimental phases, and their long-term safety and efficacy in humans remain to be established.
Resistance training is the most effective intervention for combating anabolic resistance, enhancing insulin sensitivity, reducing inflammation, and directly stimulating MPS through mechanical loading of the muscles [155]. Combining resistance exercise with protein supplementation can synergistically improve muscle mass and function [156].
Anti-inflammatory treatments, such as omega-3 fatty acids, are being explored for their potential to reduce systemic inflammation and improve muscle anabolic response [157]. Recently, the administration of an experimental drug targeting IL-11, a pro-inflammatory cytokine of the IL-6 family, increased muscle mass and strength in old mice, which was associated with up to 25% life extension [158]. The potential for anti-inflammatory therapies, particularly novel treatments targeting specific cytokines like IL-11, offers a promising approach to improving anabolic response and muscle mass in aging populations. Despite these exciting findings in preclinical models, human studies are still limited, and concerns about safety in long-term use remain. More research is needed before these treatments can be applied in clinical practice.
Hormonal therapies involving growth hormone (GH) and IGF-1 are also investigated for their anabolic effects, although they are not currently employed in clinical practice due to significant safety concerns, including the risk of cancer, diabetes, and other adverse effects, particularly in older adults [159].

Summary of Evidence and Current Clinical Recommendations

Anabolic resistance refers to a diminished ability of muscle tissue to respond to anabolic stimuli, such as dietary protein and exercise. In older adults, anabolic resistance is a key contributor to the development and progression of sarcopenia. This resistance is largely mediated by impaired nutrient sensing, insulin resistance, and chronic low-grade inflammation, which reduce MPS. Key signaling pathways, such as mTORC1, play a central role in regulating MPS, and their activation can be compromised in the setting of anabolic resistance. The intake of leucine-rich proteins and the use of anabolic regulators, such as IGF-1 and testosterone, have shown potential in overcoming anabolic resistance.
To mitigate anabolic resistance in older adults, increasing protein intake is recommended, with a particular focus on leucine-rich sources, and distributing protein intake evenly across meals to enhance MPS throughout the day. Resistance training remains a highly effective strategy for improving anabolic sensitivity. In some cases, supplementation with anabolic regulators such as HMB may be considered. Nutritional strategies and exercise should be tailored to individual needs to optimize the anabolic response.

9. Neuromuscular Junction Transmission in Sarcopenia

In recent years, neuromuscular junction (NMJ) alterations have emerged as a key determinant in the pathogenesis of sarcopenia [160]. The NMJ is a complex structure involving presynaptic motor neuron terminals, the synaptic cleft, and postsynaptic muscle membrane. Efficient transmission at the NMJ is dependent on the release of the neurotransmitter acetylcholine (ACh) from synaptic vesicles in the presynaptic terminal. This process is tightly regulated by several proteins, including synaptotagmin, SNAP receptor (SNARE) complexes, and voltage-gated calcium channels. Upon release, ACh traverses the synaptic cleft and binds to nicotinic acetylcholine receptors (nAChRs) on the muscle membrane, leading to depolarization and subsequent muscle contraction [161].
With aging, several alterations in NMJ structure and function contribute to sarcopenia (Figure 2). One significant change is the reduction in the number and function of motor neurons, leading to denervation of muscle fibers. This denervation results in the fragmentation and simplification of the NMJ, impairing efficient neurotransmission [160]. Additionally, advancing age is associated with a decline in the expression and clustering of nAChRs on the muscle membrane, further compromising synaptic efficacy [162]. Mitochondrial dysfunction within both neurons and muscle fibers also plays a role, as oxidative stress can damage NMJ components and exacerbate synaptic decline [160]. At the molecular level, several signaling pathways and molecules are implicated in the maintenance and degradation of the NMJ. The agrin-MuSK (muscle-specific kinase) pathway is critical for the formation and maintenance of NMJs. Agrin, released by motor neurons, activates MuSK on the muscle membrane, promoting the clustering of nAChRs and stabilizing the NMJ structure. With aging, there is a reduction in agrin levels and MuSK activity, leading to NMJ destabilization [163]. Another important pathway involves neuregulin-1 (NRG1) and its receptors, ErbB2/4, which are essential for the survival and function of motor neurons and muscle fibers. Decreased NRG1-ErbB signaling is associated with NMJ degeneration in aged muscles [164]. These findings provide insight into the molecular mechanisms underlying NMJ degeneration, offering promising therapeutic targets, such as agrin and MuSK signaling, that can potentially stabilize NMJs and improve muscle function in sarcopenia. However, the clinical translation of these molecular targets is still in its early stages, and more research is needed to evaluate their effectiveness in human populations.
To address sarcopenia through NMJ-targeted therapies, several strategies can be considered. One promising approach is the use of compounds that enhance NMJ stability and function. For example, acetylcholinesterase inhibitors, which prevent the breakdown of ACh in the synaptic cleft, can enhance neurotransmission and have shown potential in mitigating muscle weakness in sarcopenic mice [165]. Additionally, drugs that mimic or boost agrin and MuSK signaling could help maintain NMJ integrity [166]. Experimental therapies using recombinant agrin or small molecules that activate MuSK are currently under investigation [163,167]. These approaches show promise in preclinical models, particularly in improving NMJ function and muscle strength. However, their long-term safety and efficacy in humans have yet to be established, and they remain experimental.
In a recent study, Schellino et al. [168] tested the effects of ActR-Fc-nLG3, a protein that combines the soluble activin receptor, a potent myostatin inhibitor, with the C-terminal agrin nLG3 domain, in old mice. The authors found that this dual approach enhanced NMJ stability, improved muscle function, and increased overall endurance [168], suggesting a potential therapeutic strategy to counteract age-related muscle decline and sarcopenia. The dual action of myostatin inhibition and agrin enhancement represents an innovative approach that could offer synergistic benefits for NMJ and overall muscle health. However, this remains a preclinical finding, and further studies in humans are necessary to determine its therapeutic potential.
Gene therapy also holds potential for targeting NMJ degeneration. Delivering genes that encode for key NMJ proteins, such as agrin or NRG1, directly to muscle tissues using viral vectors could restore normal NMJ function [169,170]. This approach could provide long-term benefits by addressing the underlying molecular deficits in sarcopenia. However, gene therapy is still in its early stages, and concerns about safety, delivery, and cost remain significant barriers to clinical application.
Another area of intervention is the modulation of oxidative stress and mitochondrial function. Antioxidants and compounds that improve mitochondrial health may protect NMJ components from age-related damage. For example, mitochondrial-targeted antioxidants such as MitoQ have shown promise in both preclinical models of sarcopenia and humans [171,172]. Enhancing mitochondrial biogenesis by activating pathways such as PGC-1α may also support NMJ maintenance [173,174]. Targeting mitochondrial dysfunction through antioxidants and PGC-1α activation holds promise for preventing NMJ decline and improving muscle endurance. However, the long-term efficacy of these interventions, particularly in reversing established NMJ degeneration, requires further investigation.
Physical activity and exercise remain the most effective non-pharmacological interventions for sarcopenia. Physical activity stimulates NMJ remodeling and enhances synaptic transmission efficiency [175,176]. Resistance training, in particular, has been shown to increase the expression of NMJ-related genes and improve neuromuscular function in older adults [73]. Combining exercise with pharmacological or gene-based therapies could synergistically enhance their effectiveness.
Emerging research is also exploring the role of exosomes, small extracellular vesicles that mediate intercellular communication, in NMJ maintenance [177,178]. Exosomes derived from stem cells or young muscle tissues contain factors that can rejuvenate aged NMJs [179]. Developing exosome-based therapies could offer a novel approach for treating sarcopenia by delivering bioactive molecules that promote NMJ repair and regeneration [180]. Exosome-based therapies offer a novel and potentially less invasive approach to NMJ repair, leveraging the natural regenerative properties of stem cells. However, this approach is still in its experimental phase, and further research is needed to optimize the use of exosomes in clinical applications.

Summary of Evidence and Current Clinical Recommendations

With aging, NMJ transmission efficiency declines, contributing to muscle weakness and sarcopenia. Structural and functional alterations in the NMJ impair signal transmission, leading to decreased muscle strength. This degeneration is associated with both aging and disuse, with chronic inflammation and oxidative stress playing significant roles. Strategies to preserve NMJ integrity, such as exercise, nutritional support, and neuromodulatory agents, are being investigated.
Exercise, particularly resistance and neuromuscular training, is recommended to maintain or restore NMJ function. Nutritional interventions that include omega-3 fatty acids and antioxidants may help preserve NMJ integrity by reducing inflammation and oxidative stress. In some cases, pharmacological agents that enhance acetylcholine receptor function or protect NMJ transmission may be considered, although more research is needed to confirm their clinical utility.

10. Conclusions

Sarcopenia is a complex and multifactorial clinical condition in which multiple determinants may have a different weight from person to person [181]. As a result, each individual with sarcopenia may have a unique substrate on which to intervene and this does not only depend on the features of this disease, but also on the great variability in terms of health status and functional capacities within the population of older adults and the coexistence of chronic diseases and geriatric syndromes [182,183,184]. Regarding the long-standing issue of the definition of sarcopenia, we are at a point where the main diagnostic algorithms have been developed, with the next steps being to further refine current definitions and diagnostic workups, as well as the relationships linking it to other conditions resulting from changes in body composition, such as osteoporosis (osteosarcopenia) and obesity (sarcopenic obesity) [185]. Nutritional interventions and exercise have been established as key treatments for sarcopenia. Emerging therapeutic strategies, including pharmacological interventions, gene therapy, and mitochondrial-targeted antioxidants, offer promising avenues for mitigating sarcopenia (Table 3). However, more research is needed to validate the efficacy and safety of these therapies in clinical trials and real-world settings, particularly in older adults with multiple comorbidities. The combination of conventional interventions with pharmacological or gene-based therapies appears particularly promising for enhancing muscle function and NMJ stability. Yet, further clinical trials are essential to explore how these combinations can be optimized for long-term outcomes and to address concerns about potential side-effects. Exosome-based therapies and the modulation of oxidative stress and mitochondrial function are intriguing areas of ongoing research. While early studies have shown potential, more rigorous, long-term studies are required to establish their effectiveness and determine their role in standard sarcopenia treatment protocols. Addressing the molecular deficits underlying sarcopenia through these innovative approaches holds potential for significantly improving the quality of life and physical function in the aging population. However, it is essential to focus on addressing the existing research gaps, particularly by conducting long-term clinical trials, real-life cohort studies, and advancing translational research. This will be crucial for gaining a deeper understanding of the long-term efficacy, safety, and practical applicability of these emerging therapies in clinical settings.

Author Contributions

Conceptualization, S.C. and F.L.; methodology, A.P. and R.C.; validation, R.C. and E.M.; writing—original draft preparation, S.C., I.E., C.M. and G.G.; writing—review and editing, R.C., A.P., M.T., E.M. and F.L.; visualization, S.C. and R.C.; supervision, E.M. and F.L.; funding acquisition, E.M. and F.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Università Cattolica del Sacro Cuore (D1.2023) and the nonprofit research foundation “Centro Studi Achille e Linda Lorenzon” (N/A). This work was also supported by the European Union-Next Generation EU PRIN 2022YNENP3 to E.M. The authors acknowledge co-funding from Next Generation EU, in the context of the National Recovery and Resilience Plan, Investment PE8—Project Age-It: “Ageing Well in an Ageing Society”. This resource was co-financed by Next Generation EU (DM 1557 11 October 2022). The views and opinions expressed are only those of the authors and do not necessarily reflect those of the European Union or the European Commission. Neither the European Union nor the European Commission can be held responsible for them.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

Conflicts of Interest

The funders had no role in the writing of the manuscript or in the decision to publish it. The authors declare no conflicts of interest.

References

  1. Cruz-Jentoft, A.J.; Bahat, G.; Bauer, J.; Boirie, Y.; Bruyere, O.; Cederholm, T.; Cooper, C.; Landi, F.; Rolland, Y.; Sayer, A.A.; et al. Sarcopenia: Revised European consensus on definition and diagnosis. Age Ageing 2019, 48, 16–31. [Google Scholar] [CrossRef]
  2. Evans, W.J.; Guralnik, J.; Cawthon, P.; Appleby, J.; Landi, F.; Clarke, L.; Vellas, B.; Ferrucci, L.; Roubenoff, R. Sarcopenia: No consensus, no diagnostic criteria, and no approved indication—How did we get here? Geroscience 2024, 46, 183–190. [Google Scholar] [CrossRef]
  3. Anker, S.D.; Morley, J.E.; von Haehling, S. Welcome to the ICD-10 code for sarcopenia. J. Cachexia Sarcopenia Muscle 2016, 7, 512–514. [Google Scholar] [CrossRef]
  4. Cesari, M.; Landi, F.; Calvani, R.; Cherubini, A.; Di Bari, M.; Kortebein, P.; Del Signore, S.; Le Lain, R.; Vellas, B.; Pahor, M.; et al. Rationale for a preliminary operational definition of physical frailty and sarcopenia in the SPRINTT trial. Aging Clin. Exp. Res. 2017, 29, 81–88. [Google Scholar] [CrossRef]
  5. Cacciatore, S.; Calvani, R.; Marzetti, E.; Picca, A.; Russo, A.; Tosato, M.; Landi, F. Physical performance is associated with long-term survival in adults 80 years and older: Results from the ilSIRENTE study. J. Am. Geriatr. Soc. 2024, 72, 2585–2589. [Google Scholar] [CrossRef]
  6. Beaudart, C.; Demonceau, C.; Reginster, J.Y.; Locquet, M.; Cesari, M.; Cruz Jentoft, A.J.; Bruyere, O. Sarcopenia and health-related quality of life: A systematic review and meta-analysis. J. Cachexia Sarcopenia Muscle 2023, 14, 1228–1243. [Google Scholar] [CrossRef]
  7. Beaudart, C.; Zaaria, M.; Pasleau, F.; Reginster, J.Y.; Bruyere, O. Health Outcomes of Sarcopenia: A Systematic Review and Meta-Analysis. PLoS ONE 2017, 12, e0169548. [Google Scholar] [CrossRef]
  8. Petermann-Rocha, F.; Balntzi, V.; Gray, S.R.; Lara, J.; Ho, F.K.; Pell, J.P.; Celis-Morales, C. Global prevalence of sarcopenia and severe sarcopenia: A systematic review and meta-analysis. J. Cachexia Sarcopenia Muscle 2022, 13, 86–99. [Google Scholar] [CrossRef]
  9. Ethgen, O.; Beaudart, C.; Buckinx, F.; Bruyere, O.; Reginster, J.Y. The Future Prevalence of Sarcopenia in Europe: A Claim for Public Health Action. Calcif. Tissue Int. 2017, 100, 229–234. [Google Scholar] [CrossRef]
  10. Cesari, M.; Bernabei, R.; Vellas, B.; Fielding, R.A.; Rooks, D.; Azzolino, D.; Mariani, J.; Oliva, A.A.; Bhasin, S.; Rolland, Y. Challenges in the Development of Drugs for Sarcopenia and Frailty—Report from the International Conference on Frailty and Sarcopenia Research (ICFSR) Task Force. J. Frailty Aging 2022, 11, 135–142. [Google Scholar] [CrossRef]
  11. Calvani, R.; Picca, A.; Coelho-Junior, H.J.; Tosato, M.; Marzetti, E.; Landi, F. Diet for the prevention and management of sarcopenia. Metabolism 2023, 146, 155637. [Google Scholar] [CrossRef]
  12. Bernabei, R.; Landi, F.; Calvani, R.; Cesari, M.; Del Signore, S.; Anker, S.D.; Bejuit, R.; Bordes, P.; Cherubini, A.; Cruz-Jentoft, A.J.; et al. Multicomponent intervention to prevent mobility disability in frail older adults: Randomised controlled trial (SPRINTT project). BMJ 2022, 377, e068788. [Google Scholar] [CrossRef]
  13. Picca, A.; Calvani, R.; Sirago, G.; Coelho-Junior, H.J.; Marzetti, E. Molecular routes to sarcopenia and biomarker development: Per aspera ad astra. Curr. Opin. Pharmacol. 2021, 57, 140–147. [Google Scholar] [CrossRef]
  14. Rolland, Y.; Dray, C.; Vellas, B.; Barreto, P.S. Current and investigational medications for the treatment of sarcopenia. Metabolism 2023, 149, 155597. [Google Scholar] [CrossRef]
  15. Hong, X.; Campanario, S.; Ramirez-Pardo, I.; Grima-Terren, M.; Isern, J.; Munoz-Canoves, P. Stem cell aging in the skeletal muscle: The importance of communication. Ageing Res. Rev. 2022, 73, 101528. [Google Scholar] [CrossRef]
  16. Sousa-Victor, P.; Garcia-Prat, L.; Munoz-Canoves, P. Control of satellite cell function in muscle regeneration and its disruption in ageing. Nat. Rev. Mol. Cell Biol. 2022, 23, 204–226. [Google Scholar] [CrossRef]
  17. Yoshida, T.; Delafontaine, P. Mechanisms of IGF-1-Mediated Regulation of Skeletal Muscle Hypertrophy and Atrophy. Cells 2020, 9, 1970. [Google Scholar] [CrossRef]
  18. Ryan, A.S.; Li, G. Skeletal muscle myostatin gene expression and sarcopenia in overweight and obese middle-aged and older adults. JCSM Clin. Rep. 2021, 6, 137–142. [Google Scholar] [CrossRef]
  19. Whittemore, L.A.; Song, K.; Li, X.; Aghajanian, J.; Davies, M.; Girgenrath, S.; Hill, J.J.; Jalenak, M.; Kelley, P.; Knight, A.; et al. Inhibition of myostatin in adult mice increases skeletal muscle mass and strength. Biochem. Biophys. Res. Commun. 2003, 300, 965–971. [Google Scholar] [CrossRef]
  20. Lee, S.J.; Bhasin, S.; Klickstein, L.; Krishnan, V.; Rooks, D. Challenges and Future Prospects of Targeting Myostatin/Activin A Signaling to Treat Diseases of Muscle Loss and Metabolic Dysfunction. J. Gerontol. A Biol. Sci. Med. Sci. 2023, 78, 32–37. [Google Scholar] [CrossRef]
  21. EU Clinical Trials Register. Available online: https://www.clinicaltrialsregister.eu/ctr-search/trial/2013-003134-33/results (accessed on 2 July 2024).
  22. Becker, C.; Lord, S.R.; Studenski, S.A.; Warden, S.J.; Fielding, R.A.; Recknor, C.P.; Hochberg, M.C.; Ferrari, S.L.; Blain, H.; Binder, E.F.; et al. Myostatin antibody (LY2495655) in older weak fallers: A proof-of-concept, randomised, phase 2 trial. Lancet Diabetes Endocrinol. 2015, 3, 948–957. [Google Scholar] [CrossRef] [PubMed]
  23. Woodhouse, L.; Gandhi, R.; Warden, S.J.; Poiraudeau, S.; Myers, S.L.; Benson, C.T.; Hu, L.; Ahmad, Q.I.; Linnemeier, P.; Gomez, E.V.; et al. A Phase 2 Randomized Study Investigating the Efficacy and Safety of Myostatin Antibody LY2495655 versus Placebo in Patients Undergoing Elective Total Hip Arthroplasty. J. Frailty Aging 2016, 5, 62–70. [Google Scholar] [CrossRef]
  24. Rooks, D.; Swan, T.; Goswami, B.; Filosa, L.A.; Bunte, O.; Panchaud, N.; Coleman, L.A.; Miller, R.R.; Garcia Garayoa, E.; Praestgaard, J.; et al. Bimagrumab vs. Optimized Standard of Care for Treatment of Sarcopenia in Community-Dwelling Older Adults: A Randomized Clinical Trial. JAMA Netw. Open 2020, 3, e2020836. [Google Scholar] [CrossRef] [PubMed]
  25. Rooks, D.; Praestgaard, J.; Hariry, S.; Laurent, D.; Petricoul, O.; Perry, R.G.; Lach-Trifilieff, E.; Roubenoff, R. Treatment of Sarcopenia with Bimagrumab: Results from a Phase II, Randomized, Controlled, Proof-of-Concept Study. J. Am. Geriatr. Soc. 2017, 65, 1988–1995. [Google Scholar] [CrossRef] [PubMed]
  26. Hofbauer, L.C.; Witvrouw, R.; Varga, Z.; Shiota, N.; Cremer, M.; Tanko, L.B.; Rooks, D.; Auberson, L.Z.; Arkuszewski, M.; Fretault, N.; et al. Bimagrumab to improve recovery after hip fracture in older adults: A multicentre, double-blind, randomised, parallel-group, placebo-controlled, phase 2a/b trial. Lancet Healthy Longev. 2021, 2, e263–e274. [Google Scholar] [CrossRef]
  27. Tu, H.; Li, Y.L. Inflammation balance in skeletal muscle damage and repair. Front. Immunol. 2023, 14, 1133355. [Google Scholar] [CrossRef] [PubMed]
  28. Belizario, J.E.; Fontes-Oliveira, C.C.; Borges, J.P.; Kashiabara, J.A.; Vannier, E. Skeletal muscle wasting and renewal: A pivotal role of myokine IL-6. SpringerPlus 2016, 5, 619. [Google Scholar] [CrossRef] [PubMed]
  29. Alvarez, A.M.; DeOcesano-Pereira, C.; Teixeira, C.; Moreira, V. IL-1beta and TNF-alpha Modulation of Proliferated and Committed Myoblasts: IL-6 and COX-2-Derived Prostaglandins as Key Actors in the Mechanisms Involved. Cells 2020, 9, 2005. [Google Scholar] [CrossRef]
  30. Landi, F.; Marzetti, E.; Liperoti, R.; Pahor, M.; Russo, A.; Martone, A.M.; Colloca, G.; Capoluongo, E.; Bernabei, R. Nonsteroidal anti-inflammatory drug (NSAID) use and sarcopenia in older people: Results from the ilSIRENTE study. J. Am. Med. Dir. Assoc. 2013, 14, 626.e9–626.e13. [Google Scholar] [CrossRef]
  31. Trappe, T.A.; Carroll, C.C.; Dickinson, J.M.; LeMoine, J.K.; Haus, J.M.; Sullivan, B.E.; Lee, J.D.; Jemiolo, B.; Weinheimer, E.M.; Hollon, C.J. Influence of acetaminophen and ibuprofen on skeletal muscle adaptations to resistance exercise in older adults. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2011, 300, R655–R662. [Google Scholar] [CrossRef]
  32. Ribeiro, H.; Rodrigues, I.; Napoleao, L.; Lira, L.; Marques, D.; Verissimo, M.; Andrade, J.P.; Dourado, M. Non-steroidal anti-inflammatory drugs (NSAIDs), pain and aging: Adjusting prescription to patient features. Biomed. Pharmacother. 2022, 150, 112958. [Google Scholar] [CrossRef] [PubMed]
  33. Cai, L.; Shi, L.; Peng, Z.; Sun, Y.; Chen, J. Ageing of skeletal muscle extracellular matrix and mitochondria: Finding a potential link. Ann. Med. 2023, 55, 2240707. [Google Scholar] [CrossRef] [PubMed]
  34. Melouane, A.; Yoshioka, M.; St-Amand, J. Extracellular matrix/mitochondria pathway: A novel potential target for sarcopenia. Mitochondrion 2020, 50, 63–70. [Google Scholar] [CrossRef] [PubMed]
  35. Kumar, L.; Bisen, M.; Khan, A.; Kumar, P.; Patel, S.K.S. Role of Matrix Metalloproteinases in Musculoskeletal Diseases. Biomedicines 2022, 10, 2477. [Google Scholar] [CrossRef]
  36. Barakat, N.; Jangir, H.; Gallo, L.; Grillo, M.; Guo, X.; Hickman, J. Inhibition of Metalloproteinases Extends Longevity and Function of In Vitro Human iPSC-Derived Skeletal Muscle. Biomedicines 2024, 12, 856. [Google Scholar] [CrossRef]
  37. Loreti, M.; Sacco, A. The jam session between muscle stem cells and the extracellular matrix in the tissue microenvironment. NPJ Regen. Med. 2022, 7, 16. [Google Scholar] [CrossRef]
  38. Brunet, A.; Goodell, M.A.; Rando, T.A. Ageing and rejuvenation of tissue stem cells and their niches. Nat. Rev. Mol. Cell Biol. 2023, 24, 45–62. [Google Scholar] [CrossRef]
  39. Boyer, O.; Butler-Browne, G.; Chinoy, H.; Cossu, G.; Galli, F.; Lilleker, J.B.; Magli, A.; Mouly, V.; Perlingeiro, R.C.R.; Previtali, S.C.; et al. Myogenic Cell Transplantation in Genetic and Acquired Diseases of Skeletal Muscle. Front. Genet. 2021, 12, 702547. [Google Scholar] [CrossRef]
  40. Eugenis, I.; Wu, D.; Rando, T.A. Cells, scaffolds, and bioactive factors: Engineering strategies for improving regeneration following volumetric muscle loss. Biomaterials 2021, 278, 121173. [Google Scholar] [CrossRef]
  41. Mihaly, E.; Altamirano, D.E.; Tuffaha, S.; Grayson, W. Engineering skeletal muscle: Building complexity to achieve functionality. Semin. Cell Dev. Biol. 2021, 119, 61–69. [Google Scholar] [CrossRef]
  42. Weng, S.; Gao, F.; Wang, J.; Li, X.; Chu, B.; Wang, J.; Yang, G. Improvement of muscular atrophy by AAV-SaCas9-mediated myostatin gene editing in aged mice. Cancer Gene Ther. 2020, 27, 960–975. [Google Scholar] [CrossRef] [PubMed]
  43. Wohlwend, M.; Laurila, P.P.; Williams, K.; Romani, M.; Lima, T.; Pattawaran, P.; Benegiamo, G.; Salonen, M.; Schneider, B.L.; Lahti, J.; et al. The exercise-induced long noncoding RNA CYTOR promotes fast-twitch myogenesis in aging. Sci. Transl. Med. 2021, 13, eabc7367. [Google Scholar] [CrossRef] [PubMed]
  44. Rashid, M.I.; Ito, T.; Miya, F.; Shimojo, D.; Arimoto, K.; Onodera, K.; Okada, R.; Nagashima, T.; Yamamoto, K.; Khatun, Z.; et al. Simple and efficient differentiation of human iPSCs into contractible skeletal muscles for muscular disease modeling. Sci. Rep. 2023, 13, 8146. [Google Scholar] [CrossRef]
  45. Caron, L.; Testa, S.; Magdinier, F. Induced Pluripotent Stem Cells for Modeling Physiological and Pathological Striated Muscle Complexity. J. Neuromuscul. Dis. 2023, 10, 761–776. [Google Scholar] [CrossRef]
  46. Tompkins, B.A.; DiFede, D.L.; Khan, A.; Landin, A.M.; Schulman, I.H.; Pujol, M.V.; Heldman, A.W.; Miki, R.; Goldschmidt-Clermont, P.J.; Goldstein, B.J.; et al. Allogeneic Mesenchymal Stem Cells Ameliorate Aging Frailty: A Phase II Randomized, Double-Blind, Placebo-Controlled Clinical Trial. J. Gerontol. A Biol. Sci. Med. Sci. 2017, 72, 1513–1522. [Google Scholar] [CrossRef]
  47. Golpanian, S.; DiFede, D.L.; Khan, A.; Schulman, I.H.; Landin, A.M.; Tompkins, B.A.; Heldman, A.W.; Miki, R.; Goldstein, B.J.; Mushtaq, M.; et al. Allogeneic Human Mesenchymal Stem Cell Infusions for Aging Frailty. J. Gerontol. A Biol. Sci. Med. Sci. 2017, 72, 1505–1512. [Google Scholar] [CrossRef]
  48. Aranega, A.E.; Lozano-Velasco, E.; Rodriguez-Outeirino, L.; Ramirez de Acuna, F.; Franco, D.; Hernandez-Torres, F. MiRNAs and Muscle Regeneration: Therapeutic Targets in Duchenne Muscular Dystrophy. Int. J. Mol. Sci. 2021, 22, 4236. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, L.; Xu, Z.; Ling, D.; Li, J.; Wang, Y.; Shan, T. The regulatory role of dietary factors in skeletal muscle development, regeneration and function. Crit. Rev. Food Sci. Nutr. 2022, 62, 764–782. [Google Scholar] [CrossRef]
  50. Beaudry, K.M.; Binet, E.R.; Collao, N.; De Lisio, M. Nutritional Regulation of Muscle Stem Cells in Exercise and Disease: The Role of Protein and Amino Acid Dietary Supplementation. Front. Physiol. 2022, 13, 915390. [Google Scholar] [CrossRef]
  51. Ko, C.H.; Wu, S.J.; Wang, S.T.; Chang, Y.F.; Chang, C.S.; Kuan, T.S.; Chuang, H.Y.; Chang, C.M.; Chou, W.; Wu, C.H. Effects of enriched branched-chain amino acid supplementation on sarcopenia. Aging 2020, 12, 15091–15103. [Google Scholar] [CrossRef]
  52. Linton, C.; Wright, H.H.; Wadsworth, D.P.; Schaumberg, M.A. Dietary Inflammatory Index and Associations with Sarcopenia Symptomology in Community-Dwelling Older Adults. Nutrients 2022, 14, 5319. [Google Scholar] [CrossRef] [PubMed]
  53. McGlory, C.; Calder, P.C.; Nunes, E.A. The Influence of Omega-3 Fatty Acids on Skeletal Muscle Protein Turnover in Health, Disuse, and Disease. Front. Nutr. 2019, 6, 144. [Google Scholar] [CrossRef] [PubMed]
  54. Kaczka, P.; Michalczyk, M.M.; Jastrzab, R.; Gawelczyk, M.; Kubicka, K. Mechanism of Action and the Effect of Beta-Hydroxy-Beta-Methylbutyrate (HMB) Supplementation on Different Types of Physical Performance—A Systematic Review. J. Hum. Kinet. 2019, 68, 211–222. [Google Scholar] [CrossRef]
  55. Landi, F.; Calvani, R.; Picca, A.; Marzetti, E. Beta-hydroxy-beta-methylbutyrate and sarcopenia: From biological plausibility to clinical evidence. Curr. Opin. Clin. Nutr. Metab. Care 2019, 22, 37–43. [Google Scholar] [CrossRef] [PubMed]
  56. Candow, D.G.; Chilibeck, P.D.; Forbes, S.C.; Fairman, C.M.; Gualano, B.; Roschel, H. Creatine supplementation for older adults: Focus on sarcopenia, osteoporosis, frailty and Cachexia. Bone 2022, 162, 116467. [Google Scholar] [CrossRef]
  57. Sobolewski, E.J.; Thompson, B.J.; Smith, A.E.; Ryan, E.D. The Physiological Effects of Creatine Supplementation on Hydration: A Review. Am. J. Lifestyle Med. 2011, 5, 320–327. [Google Scholar] [CrossRef]
  58. Lundberg, J.O.; Weitzberg, E. Nitric oxide signaling in health and disease. Cell 2022, 185, 2853–2878. [Google Scholar] [CrossRef]
  59. Kamei, Y.; Hatazawa, Y.; Uchitomi, R.; Yoshimura, R.; Miura, S. Regulation of Skeletal Muscle Function by Amino Acids. Nutrients 2020, 12, 261. [Google Scholar] [CrossRef]
  60. Volkert, D.; Beck, A.M.; Cederholm, T.; Cruz-Jentoft, A.; Hooper, L.; Kiesswetter, E.; Maggio, M.; Raynaud-Simon, A.; Sieber, C.; Sobotka, L.; et al. ESPEN practical guideline: Clinical nutrition and hydration in geriatrics. Clin. Nutr. 2022, 41, 958–989. [Google Scholar] [CrossRef]
  61. Chen, Q.; Huang, W.; Capanoglu, E.; Amrouche, A.T.; Lu, B. Targeting mitochondrial quality control in muscle aging: Natural dietary products as potential interventions. Food Front. 2023, 4, 1206–1241. [Google Scholar] [CrossRef]
  62. Chen, L.; Zhu, Y.; Hu, Z.; Wu, S.; Jin, C. Beetroot as a functional food with huge health benefits: Antioxidant, antitumor, physical function, and chronic metabolomics activity. Food Sci. Nutr. 2021, 9, 6406–6420. [Google Scholar] [CrossRef] [PubMed]
  63. Kapil, V.; Khambata, R.S.; Jones, D.A.; Rathod, K.; Primus, C.; Massimo, G.; Fukuto, J.M.; Ahluwalia, A. The Noncanonical Pathway for In Vivo Nitric Oxide Generation: The Nitrate-Nitrite-Nitric Oxide Pathway. Pharmacol. Rev. 2020, 72, 692–766. [Google Scholar] [CrossRef] [PubMed]
  64. Cacciatore, S.; Calvani, R.; Marzetti, E.; Picca, A.; Coelho-Junior, H.J.; Martone, A.M.; 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. [Google Scholar] [CrossRef] [PubMed]
  65. Galluzzo, V.; Zazzara, M.B.; Ciciarello, F.; Savera, G.; Pais, C.; Calvani, R.; Picca, A.; Marzetti, E.; Landi, F.; Tosato, M.; et al. Fatigue in COVID-19 survivors: The potential impact of a nutritional supplement on muscle strength and function. Clin. Nutr. ESPEN 2022, 51, 215–221. [Google Scholar] [CrossRef]
  66. Song, Z.; Pan, T.; Tong, X.; Yang, Y.; Zhang, Z. The effects of nutritional supplementation on older sarcopenic individuals who engage in resistance training: A meta-analysis. Front. Nutr. 2023, 10, 1109789. [Google Scholar] [CrossRef]
  67. Heidari, D.; Shirvani, H.; Bazgir, B.; Shamsoddini, A. The Resistance Training Effects on Skeletal Muscle Stem Cells in Older Adult: A Systematic Review and Meta-Analysis. Cell J. 2023, 25, 513–523. [Google Scholar] [CrossRef] [PubMed]
  68. Hirabara, S.M.; Marzuca-Nassr, G.N.; Cury-Boaventura, M.F. Nutrition and Exercise Interventions on Skeletal Muscle Physiology, Injury and Recovery: From Mechanisms to Therapy. Nutrients 2024, 16, 293. [Google Scholar] [CrossRef]
  69. Ahsan, M.; Garneau, L.; Aguer, C. The bidirectional relationship between AMPK pathway activation and myokine secretion in skeletal muscle: How it affects energy metabolism. Front. Physiol. 2022, 13, 1040809. [Google Scholar] [CrossRef]
  70. Grevendonk, L.; Connell, N.J.; McCrum, C.; Fealy, C.E.; Bilet, L.; Bruls, Y.M.H.; Mevenkamp, J.; Schrauwen-Hinderling, V.B.; Jorgensen, J.A.; Moonen-Kornips, E.; et al. Impact of aging and exercise on skeletal muscle mitochondrial capacity, energy metabolism, and physical function. Nat. Commun. 2021, 12, 4773. [Google Scholar] [CrossRef]
  71. Ribeiro, A.S.; Avelar, A.; Schoenfeld, B.J.; Ritti Dias, R.M.; Altimari, L.R.; Cyrino, E.S. Resistance training promotes increase in intracellular hydration in men and women. Eur. J. Sport Sci. 2014, 14, 578–585. [Google Scholar] [CrossRef]
  72. Flor-Rufino, C.; Barrachina-Igual, J.; Perez-Ros, P.; Pablos-Monzo, A.; Sanz-Requena, R.; Martinez-Arnau, F.M. Fat infiltration and muscle hydration improve after high-intensity resistance training in women with sarcopenia. A randomized clinical trial. Maturitas 2023, 168, 29–36. [Google Scholar] [CrossRef] [PubMed]
  73. Kreko-Pierce, T.; Eaton, B.A. Rejuvenation of the aged neuromuscular junction by exercise. Cell Stress 2018, 2, 25–33. [Google Scholar] [CrossRef] [PubMed]
  74. Lopez-Otin, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of aging: An expanding universe. Cell 2023, 186, 243–278. [Google Scholar] [CrossRef] [PubMed]
  75. Marzetti, E.; Calvani, R.; Coelho-Junior, H.J.; Landi, F.; Picca, A. Mitochondrial Quantity and Quality in Age-Related Sarcopenia. Int. J. Mol. Sci. 2024, 25, 2052. [Google Scholar] [CrossRef] [PubMed]
  76. Affourtit, C.; Carre, J.E. Mitochondrial involvement in sarcopenia. Acta Physiol. 2024, 240, e14107. [Google Scholar] [CrossRef] [PubMed]
  77. Amorim, J.A.; Coppotelli, G.; Rolo, A.P.; Palmeira, C.M.; Ross, J.M.; Sinclair, D.A. Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat. Rev. Endocrinol. 2022, 18, 243–258. [Google Scholar] [CrossRef]
  78. Halling, J.F.; Pilegaard, H. PGC-1alpha-mediated regulation of mitochondrial function and physiological implications. Appl. Physiol. Nutr. Metab. 2020, 45, 927–936. [Google Scholar] [CrossRef]
  79. Ji, Z.; Liu, G.H.; Qu, J. Mitochondrial sirtuins, metabolism, and aging. J. Genet. Genom. 2022, 49, 287–298. [Google Scholar] [CrossRef]
  80. Goel, S.; Singh, R.; Singh, V.; Singh, H.; Kumari, P.; Chopra, H.; Sharma, R.; Nepovimova, E.; Valis, M.; Kuca, K.; et al. Metformin: Activation of 5′ AMP-activated protein kinase and its emerging potential beyond anti-hyperglycemic action. Front. Genet. 2022, 13, 1022739. [Google Scholar] [CrossRef]
  81. Marco-Bonilla, M.; Fresnadillo, M.; Largo, R.; Herrero-Beaumont, G.; Mediero, A. Energy Regulation in Inflammatory Sarcopenia by the Purinergic System. Int. J. Mol. Sci. 2023, 24, 6904. [Google Scholar] [CrossRef]
  82. Xie, G.; Jin, H.; Mikhail, H.; Pavel, V.; Yang, G.; Ji, B.; Lu, B.; Li, Y. Autophagy in sarcopenia: Possible mechanisms and novel therapies. Biomed. Pharmacother. 2023, 165, 115147. [Google Scholar] [CrossRef] [PubMed]
  83. Hidalgo-Gutierrez, A.; Gonzalez-Garcia, P.; Diaz-Casado, M.E.; Barriocanal-Casado, E.; Lopez-Herrador, S.; Quinzii, C.M.; Lopez, L.C. Metabolic Targets of Coenzyme Q10 in Mitochondria. Antioxidants 2021, 10, 520. [Google Scholar] [CrossRef] [PubMed]
  84. Fielding, R.; Riede, L.; Lugo, J.P.; Bellamine, A. l-Carnitine Supplementation in Recovery after Exercise. Nutrients 2018, 10, 349. [Google Scholar] [CrossRef]
  85. Huang, L.T.; Wang, J.H. The Therapeutic Intervention of Sex Steroid Hormones for Sarcopenia. Front. Med. 2021, 8, 739251. [Google Scholar] [CrossRef]
  86. Lorenzo, I.; Serra-Prat, M.; Yebenes, J.C. The Role of Water Homeostasis in Muscle Function and Frailty: A Review. Nutrients 2019, 11, 1857. [Google Scholar] [CrossRef] [PubMed]
  87. Seifter, J.L. Body Fluid Compartments, Cell Membrane Ion Transport, Electrolyte Concentrations, and Acid-Base Balance. Semin. Nephrol. 2019, 39, 368–379. [Google Scholar] [CrossRef]
  88. Serra-Prat, M.; Lorenzo, I.; Palomera, E.; Ramirez, S.; Yebenes, J.C. Total Body Water and Intracellular Water Relationships with Muscle Strength, Frailty and Functional Performance in an Elderly Population. J. Nutr. Health Aging 2019, 23, 96–101. [Google Scholar] [CrossRef]
  89. Serra-Prat, M.; Lorenzo, I.; Palomera, E.; Yebenes, J.C.; Campins, L.; Cabre, M. Intracellular Water Content in Lean Mass is Associated with Muscle Strength, Functional Capacity, and Frailty in Community-Dwelling Elderly Individuals. A Cross-Sectional Study. Nutrients 2019, 11, 661. [Google Scholar] [CrossRef]
  90. Hioka, A.; Akazawa, N.; Okawa, N.; Nagahiro, S. Influence of aging on extracellular water-to-total body water ratio in community-dwelling females. Clin. Nutr. ESPEN 2024, 60, 73–78. [Google Scholar] [CrossRef]
  91. Yamada, Y.; Schoeller, D.A.; Nakamura, E.; Morimoto, T.; Kimura, M.; Oda, S. Extracellular water may mask actual muscle atrophy during aging. J. Gerontol. A Biol. Sci. Med. Sci. 2010, 65, 510–516. [Google Scholar] [CrossRef]
  92. Schliess, F.; Richter, L.; vom Dahl, S.; Haussinger, D. Cell hydration and mTOR-dependent signalling. Acta Physiol. 2006, 187, 223–229. [Google Scholar] [CrossRef] [PubMed]
  93. Yamada, Y.; Yoshida, T.; Yokoyama, K.; Watanabe, Y.; Miyake, M.; Yamagata, E.; Yamada, M.; Kimura, M.; Kyoto-Kameoka, S. The Extracellular to Intracellular Water Ratio in Upper Legs is Negatively Associated with Skeletal Muscle Strength and Gait Speed in Older People. J. Gerontol. A Biol. Sci. Med. Sci. 2017, 72, 293–298. [Google Scholar] [CrossRef] [PubMed]
  94. Aslesh, T.; Al-Aghbari, A.; Yokota, T. Assessing the Role of Aquaporin 4 in Skeletal Muscle Function. Int. J. Mol. Sci. 2023, 24, 1489. [Google Scholar] [CrossRef] [PubMed]
  95. Pirkmajer, S.; Chibalin, A.V. Na,K-ATPase regulation in skeletal muscle. Am. J. Physiol. Endocrinol. Metab. 2016, 311, E1–E31. [Google Scholar] [CrossRef] [PubMed]
  96. Lindinger, M.I. A century of exercise physiology: Key concepts in muscle cell volume regulation. Eur. J. Appl. Physiol. 2022, 122, 541–559. [Google Scholar] [CrossRef]
  97. Rebelo-Marques, A.; Coelho-Ribeiro, B.; De Sousa Lages, A.; Andrade, R.; Afonso, J.; Pereira, R.; Batista, A.S.; Teixeira, V.H.; Jacome, C. Trends and Missing Links in (De)Hydration Research: A Narrative Review. Nutrients 2024, 16, 1709. [Google Scholar] [CrossRef]
  98. Sumukadas, D.; Struthers, A.D.; McMurdo, M.E. Sarcopenia—A potential target for Angiotensin-converting enzyme inhibition? Gerontology 2006, 52, 237–242. [Google Scholar] [CrossRef]
  99. Masot, O.; Miranda, J.; Santamaria, A.L.; Paraiso Pueyo, E.; Pascual, A.; Botigue, T. Fluid Intake Recommendation Considering the Physiological Adaptations of Adults over 65 Years: A Critical Review. Nutrients 2020, 12, 3383. [Google Scholar] [CrossRef]
  100. Bakhtiar, S.M.; LeBlanc, J.G.; Salvucci, E.; Ali, A.; Martin, R.; Langella, P.; Chatel, J.M.; Miyoshi, A.; Bermudez-Humaran, L.G.; Azevedo, V. Implications of the human microbiome in inflammatory bowel diseases. FEMS Microbiol. Lett. 2013, 342, 10–17. [Google Scholar] [CrossRef]
  101. LeBlanc, J.G.; Milani, C.; de Giori, G.S.; Sesma, F.; van Sinderen, D.; Ventura, M. Bacteria as vitamin suppliers to their host: A gut microbiota perspective. Curr. Opin. Biotechnol. 2013, 24, 160–168. [Google Scholar] [CrossRef]
  102. Hooper, L.V.; Littman, D.R.; Macpherson, A.J. Interactions between the microbiota and the immune system. Science 2012, 336, 1268–1273. [Google Scholar] [CrossRef] [PubMed]
  103. O’Connor, E.M. The role of gut microbiota in nutritional status. Curr. Opin. Clin. Nutr. Metab. Care 2013, 16, 509–516. [Google Scholar] [CrossRef]
  104. Ghosh, T.S.; Shanahan, F.; O’Toole, P.W. The gut microbiome as a modulator of healthy ageing. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 565–584. [Google Scholar] [CrossRef]
  105. Wilmanski, T.; Diener, C.; Rappaport, N.; Patwardhan, S.; Wiedrick, J.; Lapidus, J.; Earls, J.C.; Zimmer, A.; Glusman, G.; Robinson, M.; et al. Gut microbiome pattern reflects healthy ageing and predicts survival in humans. Nat. Metab. 2021, 3, 274–286. [Google Scholar] [CrossRef]
  106. Barone, M.; D’Amico, F.; Rampelli, S.; Brigidi, P.; Turroni, S. Age-related diseases, therapies and gut microbiome: A new frontier for healthy aging. Mech. Ageing Dev. 2022, 206, 111711. [Google Scholar] [CrossRef] [PubMed]
  107. Bosco, N.; Noti, M. The aging gut microbiome and its impact on host immunity. Genes Immun. 2021, 22, 289–303. [Google Scholar] [CrossRef]
  108. Claesson, M.J.; Cusack, S.; O’Sullivan, O.; Greene-Diniz, R.; de Weerd, H.; Flannery, E.; Marchesi, J.R.; Falush, D.; Dinan, T.; Fitzgerald, G.; et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4586–4591. [Google Scholar] [CrossRef]
  109. Claesson, M.J.; Jeffery, I.B.; Conde, S.; Power, S.E.; O’Connor, E.M.; Cusack, S.; Harris, H.M.; Coakley, M.; Lakshminarayanan, B.; O’Sullivan, O.; et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 2012, 488, 178–184. [Google Scholar] [CrossRef] [PubMed]
  110. Ghosh, T.S.; Das, M.; Jeffery, I.B.; O’Toole, P.W. Adjusting for age improves identification of gut microbiome alterations in multiple diseases. Elife 2020, 9, e50240. [Google Scholar] [CrossRef]
  111. D’Amico, F.; Barone, M.; Brigidi, P.; Turroni, S. Gut microbiota in relation to frailty and clinical outcomes. Curr. Opin. Clin. Nutr. Metab. Care 2023, 26, 219–225. [Google Scholar] [CrossRef]
  112. Tavella, T.; Rampelli, S.; Guidarelli, G.; Bazzocchi, A.; Gasperini, C.; Pujos-Guillot, E.; Comte, B.; Barone, M.; Biagi, E.; Candela, M.; et al. Elevated gut microbiome abundance of Christensenellaceae, Porphyromonadaceae and Rikenellaceae is associated with reduced visceral adipose tissue and healthier metabolic profile in Italian elderly. Gut Microbes 2021, 13, 1880221. [Google Scholar] [CrossRef] [PubMed]
  113. Strasser, B.; Ticinesi, A. Intestinal microbiome in normal ageing, frailty and cognition decline. Curr. Opin. Clin. Nutr. Metab. Care 2023, 26, 8–16. [Google Scholar] [CrossRef] [PubMed]
  114. Rashidah, N.H.; Lim, S.M.; Neoh, C.F.; Majeed, A.B.A.; Tan, M.P.; Khor, H.M.; Tan, A.H.; Rehiman, S.H.; Ramasamy, K. Differential gut microbiota and intestinal permeability between frail and healthy older adults: A systematic review. Ageing Res. Rev. 2022, 82, 101744. [Google Scholar] [CrossRef] [PubMed]
  115. Xu, Y.; Liu, X.; Liu, X.; Chen, D.; Wang, M.; Jiang, X.; Xiong, Z. The Roles of the Gut Microbiota and Chronic Low-Grade Inflammation in Older Adults With Frailty. Front. Cell. Infect. Microbiol. 2021, 11, 675414. [Google Scholar] [CrossRef] [PubMed]
  116. Picca, A.; Fanelli, F.; Calvani, R.; Mule, G.; Pesce, V.; Sisto, A.; Pantanelli, C.; Bernabei, R.; Landi, F.; Marzetti, E. Gut Dysbiosis and Muscle Aging: Searching for Novel Targets against Sarcopenia. Mediators Inflamm. 2018, 2018, 7026198. [Google Scholar] [CrossRef] [PubMed]
  117. Jackson, M.A.; Jeffery, I.B.; Beaumont, M.; Bell, J.T.; Clark, A.G.; Ley, R.E.; O’Toole, P.W.; Spector, T.D.; Steves, C.J. Signatures of early frailty in the gut microbiota. Genome Med. 2016, 8, 8. [Google Scholar] [CrossRef]
  118. Haran, J.P.; Bucci, V.; Dutta, P.; Ward, D.; McCormick, B. The nursing home elder microbiome stability and associations with age, frailty, nutrition and physical location. J. Med. Microbiol. 2018, 67, 40–51. [Google Scholar] [CrossRef]
  119. Picca, A.; Ponziani, F.R.; Calvani, R.; Marini, F.; Biancolillo, A.; Coelho-Junior, H.J.; Gervasoni, J.; Primiano, A.; Putignani, L.; Del Chierico, F.; et al. Gut Microbial, Inflammatory and Metabolic Signatures in Older People with Physical Frailty and Sarcopenia: Results from the BIOSPHERE Study. Nutrients 2019, 12, 65. [Google Scholar] [CrossRef]
  120. Wu, C.S.; Wei, Q.; Wang, H.; Kim, D.M.; Balderas, M.; Wu, G.; Lawler, J.; Safe, S.; Guo, S.; Devaraj, S.; et al. Protective Effects of Ghrelin on Fasting-Induced Muscle Atrophy in Aging Mice. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 621–630. [Google Scholar] [CrossRef]
  121. Ghosh, T.S.; Rampelli, S.; Jeffery, I.B.; Santoro, A.; Neto, M.; Capri, M.; Giampieri, E.; Jennings, A.; Candela, M.; Turroni, S.; et al. Mediterranean diet intervention alters the gut microbiome in older people reducing frailty and improving health status: The NU-AGE 1-year dietary intervention across five European countries. Gut 2020, 69, 1218–1228. [Google Scholar] [CrossRef]
  122. Wilms, E.; An, R.; Smolinska, A.; Stevens, Y.; Weseler, A.R.; Elizalde, M.; Drittij, M.J.; Ioannou, A.; van Schooten, F.J.; Smidt, H.; et al. Galacto-oligosaccharides supplementation in prefrail older and healthy adults increased faecal bifidobacteria, but did not impact immune function and oxidative stress. Clin. Nutr. 2021, 40, 3019–3031. [Google Scholar] [CrossRef] [PubMed]
  123. Lee, M.C.; Tu, Y.T.; Lee, C.C.; Tsai, S.C.; Hsu, H.Y.; Tsai, T.Y.; Liu, T.H.; Young, S.L.; Lin, J.S.; Huang, C.C. Lactobacillus plantarum TWK10 Improves Muscle Mass and Functional Performance in Frail Older Adults: A Randomized, Double-Blind Clinical Trial. Microorganisms 2021, 9, 1466. [Google Scholar] [CrossRef]
  124. Witard, O.C.; Wardle, S.L.; Macnaughton, L.S.; Hodgson, A.B.; Tipton, K.D. Protein Considerations for Optimising Skeletal Muscle Mass in Healthy Young and Older Adults. Nutrients 2016, 8, 181. [Google Scholar] [CrossRef]
  125. Prior, S.J.; Ryan, A.S.; Blumenthal, J.B.; Watson, J.M.; Katzel, L.I.; Goldberg, A.P. Sarcopenia Is Associated with Lower Skeletal Muscle Capillarization and Exercise Capacity in Older Adults. J. Gerontol. A Biol. Sci. Med. Sci. 2016, 71, 1096–1101. [Google Scholar] [CrossRef]
  126. Biolo, G.; Ciocchi, B.; Stulle, M.; Bosutti, A.; Barazzoni, R.; Zanetti, M.; Antonione, R.; Lebenstedt, M.; Platen, P.; Heer, M.; et al. Calorie restriction accelerates the catabolism of lean body mass during 2 wk of bed rest. Am. J. Clin. Nutr. 2007, 86, 366–372. [Google Scholar] [CrossRef] [PubMed]
  127. Wagenmakers, A.J.; Strauss, J.A.; Shepherd, S.O.; Keske, M.A.; Cocks, M. Increased muscle blood supply and transendothelial nutrient and insulin transport induced by food intake and exercise: Effect of obesity and ageing. J. Physiol. 2016, 594, 2207–2222. [Google Scholar] [CrossRef] [PubMed]
  128. Barrett, E.J.; Wang, H.; Upchurch, C.T.; Liu, Z. Insulin regulates its own delivery to skeletal muscle by feed-forward actions on the vasculature. Am. J. Physiol. Endocrinol. Metab. 2011, 301, E252–E263. [Google Scholar] [CrossRef]
  129. Kusters, Y.H.; Barrett, E.J. Muscle microvasculature’s structural and functional specializations facilitate muscle metabolism. Am. J. Physiol. Endocrinol. Metab. 2016, 310, E379–E387. [Google Scholar] [CrossRef]
  130. Attrill, E.; Ramsay, C.; Ross, R.; Richards, S.; Sutherland, B.A.; Keske, M.A.; Eringa, E.; Premilovac, D. Metabolic-vascular coupling in skeletal muscle: A potential role for capillary pericytes? Clin. Exp. Pharmacol. Physiol. 2020, 47, 520–528. [Google Scholar] [CrossRef]
  131. Hirabara, S.M.; Silveira, L.R.; Abdulkader, F.R.; Alberici, L.C.; Procopio, J.; Carvalho, C.R.; Pithon-Curi, T.C.; Curi, R. Role of fatty acids in the transition from anaerobic to aerobic metabolism in skeletal muscle during exercise. Cell Biochem. Funct. 2006, 24, 475–481. [Google Scholar] [CrossRef]
  132. Tan, A.W.K.; Subaran, S.C.; Sauder, M.A.; Chai, W.; Jahn, L.A.; Fowler, D.E.; Patrie, J.T.; Aylor, K.W.; Basu, A.; Liu, Z. GLP-1 and Insulin Recruit Muscle Microvasculature and Dilate Conduit Artery Individually But Not Additively in Healthy Humans. J. Endocr. Soc. 2018, 2, 190–206. [Google Scholar] [CrossRef] [PubMed]
  133. Zempo, H.; Isobe, M.; Naito, H. Link between blood flow and muscle protein metabolism in elderly adults. J. Sports Med. Phys. Fitness 2017, 6, 25–31. [Google Scholar] [CrossRef]
  134. Durham, W.J.; Casperson, S.L.; Dillon, E.L.; Keske, M.A.; Paddon-Jones, D.; Sanford, A.P.; Hickner, R.C.; Grady, J.J.; Sheffield-Moore, M. Age-related anabolic resistance after endurance-type exercise in healthy humans. FASEB J. 2010, 24, 4117–4127. [Google Scholar] [CrossRef] [PubMed]
  135. Zhang, X.J.; Irtun, O.; Chinkes, D.L.; Wolfe, R.R. Acute responses of muscle protein metabolism to reduced blood flow reflect metabolic priorities for homeostasis. Am. J. Physiol. Endocrinol. Metab. 2008, 294, E551–E557. [Google Scholar] [CrossRef] [PubMed]
  136. Paulussen, K.J.M.; McKenna, C.F.; Beals, J.W.; Wilund, K.R.; Salvador, A.F.; Burd, N.A. Anabolic Resistance of Muscle Protein Turnover Comes in Various Shapes and Sizes. Front. Nutr. 2021, 8, 615849. [Google Scholar] [CrossRef]
  137. Moore, D.R. Keeping older muscle “young” through dietary protein and physical activity. Adv. Nutr. 2014, 5, 599S–607S. [Google Scholar] [CrossRef]
  138. Martin, K.A.; Merenick, B.L.; Ding, M.; Fetalvero, K.M.; Rzucidlo, E.M.; Kozul, C.D.; Brown, D.J.; Chiu, H.Y.; Shyu, M.; Drapeau, B.L.; et al. Rapamycin promotes vascular smooth muscle cell differentiation through insulin receptor substrate-1/phosphatidylinositol 3-kinase/Akt2 feedback signaling. J. Biol. Chem. 2007, 282, 36112–36120. [Google Scholar] [CrossRef]
  139. Chen, K.; Gao, P.; Li, Z.; Dai, A.; Yang, M.; Chen, S.; Su, J.; Deng, Z.; Li, L. Forkhead Box O Signaling Pathway in Skeletal Muscle Atrophy. Am. J. Pathol. 2022, 192, 1648–1657. [Google Scholar] [CrossRef]
  140. Paoli, A.; Cerullo, G.; Bianco, A.; Neri, M.; Gennaro, F.; Charrier, D.; Moro, T. Not Only Protein: Dietary Supplements to Optimize the Skeletal Muscle Growth Response to Resistance Training: The Current State of Knowledge. J. Hum. Kinet. 2024, 91, 225–244. [Google Scholar] [CrossRef]
  141. Jones, A.M.; Vanhatalo, A.; Seals, D.R.; Rossman, M.J.; Piknova, B.; Jonvik, K.L. Dietary Nitrate and Nitric Oxide Metabolism: Mouth, Circulation, Skeletal Muscle, and Exercise Performance. Med. Sci. Sports Exerc. 2021, 53, 280–294. [Google Scholar] [CrossRef]
  142. Wang, R.; Jiao, H.; Zhao, J.; Wang, X.; Lin, H. L-Arginine Enhances Protein Synthesis by Phosphorylating mTOR (Thr 2446) in a Nitric Oxide-Dependent Manner in C2C12 Cells. Oxid. Med. Cell. Longev. 2018, 2018, 7569127. [Google Scholar] [CrossRef] [PubMed]
  143. Milton-Laskibar, I.; Martinez, J.A.; Portillo, M.P. Current Knowledge on Beetroot Bioactive Compounds: Role of Nitrate and Betalains in Health and Disease. Foods 2021, 10, 1314. [Google Scholar] [CrossRef] [PubMed]
  144. Walker, M.A.; Bailey, T.G.; McIlvenna, L.; Allen, J.D.; Green, D.J.; Askew, C.D. Acute Dietary Nitrate Supplementation Improves Flow Mediated Dilatation of the Superficial Femoral Artery in Healthy Older Males. Nutrients 2019, 11, 954. [Google Scholar] [CrossRef]
  145. Parker, L.; Morrison, D.J.; Wadley, G.D.; Shaw, C.S.; Betik, A.C.; Roberts-Thomson, K.; Kaur, G.; Keske, M.A. Prior exercise enhances skeletal muscle microvascular blood flow and mitigates microvascular flow impairments induced by a high-glucose mixed meal in healthy young men. J. Physiol. 2021, 599, 83–102. [Google Scholar] [CrossRef] [PubMed]
  146. Parker, L.; Morrison, D.J.; Betik, A.C.; Roberts-Thomson, K.; Kaur, G.; Wadley, G.D.; Shaw, C.S.; Keske, M.A. High-glucose mixed-nutrient meal ingestion impairs skeletal muscle microvascular blood flow in healthy young men. Am. J. Physiol. Endocrinol. Metab. 2020, 318, E1014–E1021. [Google Scholar] [CrossRef]
  147. Tominaga, K.; Tsuchiya, A.; Nakano, O.; Kuroki, Y.; Oka, K.; Minemura, A.; Matsumoto, A.; Takahashi, M.; Kadota, Y.; Tochio, T.; et al. Increase in muscle mass associated with the prebiotic effects of 1-kestose in super-elderly patients with sarcopenia. Biosci. Microbiota Food Health 2021, 40, 150–155. [Google Scholar] [CrossRef]
  148. Conlon, J.A.; Newton, R.U.; Tufano, J.J.; Banyard, H.G.; Hopper, A.J.; Ridge, A.J.; Haff, G.G. Periodization Strategies in Older Adults: Impact on Physical Function and Health. Med. Sci. Sports Exerc. 2016, 48, 2426–2436. [Google Scholar] [CrossRef]
  149. Oikawa, S.Y.; Brisbois, T.D.; van Loon, L.J.C.; Rollo, I. Eat like an athlete: Insights of sports nutrition science to support active aging in healthy older adults. Geroscience 2021, 43, 2485–2495. [Google Scholar] [CrossRef]
  150. Haran, P.H.; Rivas, D.A.; Fielding, R.A. Role and potential mechanisms of anabolic resistance in sarcopenia. J. Cachexia Sarcopenia Muscle 2012, 3, 157–162. [Google Scholar] [CrossRef]
  151. Guillet, C.; Masgrau, A.; Walrand, S.; Boirie, Y. Impaired protein metabolism: Interlinks between obesity, insulin resistance and inflammation. Obes. Rev. 2012, 13 (Suppl. S2), 51–57. [Google Scholar] [CrossRef]
  152. Smeuninx, B.; McKendry, J.; Wilson, D.; Martin, U.; Breen, L. Age-Related Anabolic Resistance of Myofibrillar Protein Synthesis Is Exacerbated in Obese Inactive Individuals. J. Clin. Endocrinol. Metab. 2017, 102, 3535–3545. [Google Scholar] [CrossRef] [PubMed]
  153. Draganidis, D.; Jamurtas, A.Z.; Chondrogianni, N.; Mastorakos, G.; Jung, T.; Grune, T.; Papadopoulos, C.; Papanikolaou, K.; Papassotiriou, I.; Papaevgeniou, N.; et al. Low-Grade Systemic Inflammation Interferes with Anabolic and Catabolic Characteristics of the Aged Human Skeletal Muscle. Oxid. Med. Cell. Longev. 2021, 2021, 8376915. [Google Scholar] [CrossRef]
  154. 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]
  155. Tezze, C.; Sandri, M.; Tessari, P. Anabolic Resistance in the Pathogenesis of Sarcopenia in the Elderly: Role of Nutrition and Exercise in Young and Old People. Nutrients 2023, 15, 4073. [Google Scholar] [CrossRef] [PubMed]
  156. Gharahdaghi, N.; Phillips, B.E.; Szewczyk, N.J.; Smith, K.; Wilkinson, D.J.; Atherton, P.J. Links between Testosterone, Oestrogen, and the Growth Hormone/Insulin-like Growth Factor Axis and Resistance Exercise Muscle Adaptations. Front. Physiol. 2020, 11, 621226. [Google Scholar] [CrossRef] [PubMed]
  157. Bano, G.; Trevisan, C.; Carraro, S.; Solmi, M.; Luchini, C.; Stubbs, B.; Manzato, E.; Sergi, G.; Veronese, N. Inflammation and sarcopenia: A systematic review and meta-analysis. Maturitas 2017, 96, 10–15. [Google Scholar] [CrossRef] [PubMed]
  158. Widjaja, A.A.; Lim, W.W.; Viswanathan, S.; Chothani, S.; Corden, B.; Dasan, C.M.; Goh, J.W.T.; Lim, R.; Singh, B.K.; Tan, J.; et al. Inhibition of IL-11 signalling extends mammalian healthspan and lifespan. Nature 2024, 632, 157–165. [Google Scholar] [CrossRef]
  159. Trobec, K.; von Haehling, S.; Anker, S.D.; Lainscak, M. Growth hormone, insulin-like growth factor 1, and insulin signaling-a pharmacological target in body wasting and cachexia. J. Cachexia Sarcopenia Muscle 2011, 2, 191–200. [Google Scholar] [CrossRef]
  160. Marzetti, E.; Lozanoska-Ochser, B.; Calvani, R.; Landi, F.; Coelho-Junior, H.J.; Picca, A. Restoring Mitochondrial Function and Muscle Satellite Cell Signaling: Remedies against Age-Related Sarcopenia. Biomolecules 2024, 14, 415. [Google Scholar] [CrossRef]
  161. Rodriguez Cruz, P.M.; Cossins, J.; Beeson, D.; Vincent, A. The Neuromuscular Junction in Health and Disease: Molecular Mechanisms Governing Synaptic Formation and Homeostasis. Front. Mol. Neurosci. 2020, 13, 610964. [Google Scholar] [CrossRef]
  162. Bao, Z.; Cui, C.; Chow, S.K.; Qin, L.; Wong, R.M.Y.; Cheung, W.H. AChRs Degeneration at NMJ in Aging-Associated Sarcopenia-A Systematic Review. Front. Aging Neurosci. 2020, 12, 597811. [Google Scholar] [CrossRef] [PubMed]
  163. Wu, J.; Ding, P.; Wu, H.; Yang, P.; Guo, H.; Tian, Y.; Meng, L.; Zhao, Q. Sarcopenia: Molecular regulatory network for loss of muscle mass and function. Front. Nutr. 2023, 10, 1037200. [Google Scholar] [CrossRef] [PubMed]
  164. Ham, D.J.; Borsch, A.; Lin, S.; Thurkauf, M.; Weihrauch, M.; Reinhard, J.R.; Delezie, J.; Battilana, F.; Wang, X.; Kaiser, M.S.; et al. The neuromuscular junction is a focal point of mTORC1 signaling in sarcopenia. Nat. Commun. 2020, 11, 4510. [Google Scholar] [CrossRef]
  165. Tezze, C.; Amendolagine, F.I.; Nogara, L.; Baraldo, M.; Ciciliot, S.; Arcidiacono, D.; Zaramella, A.; Masiero, G.; Ferrarese, G.; Realdon, S.; et al. A combination of metformin and galantamine exhibits synergistic benefits in the treatment of sarcopenia. JCI Insight 2023, 8, e168787. [Google Scholar] [CrossRef]
  166. Ohno, K.; Ohkawara, B.; Ito, M. Agrin-LRP4-MuSK signaling as a therapeutic target for myasthenia gravis and other neuromuscular disorders. Expert Opin. Ther. Targets 2017, 21, 949–958. [Google Scholar] [CrossRef]
  167. Hettwer, S.; Lin, S.; Kucsera, S.; Haubitz, M.; Oliveri, F.; Fariello, R.G.; Ruegg, M.A.; Vrijbloed, J.W. Injection of a soluble fragment of neural agrin (NT-1654) considerably improves the muscle pathology caused by the disassembly of the neuromuscular junction. PLoS ONE 2014, 9, e88739. [Google Scholar] [CrossRef]
  168. Schellino, R.; Boido, M.; Vrijbloed, J.W.; Fariello, R.G.; Vercelli, A. Synergistically Acting on Myostatin and Agrin Pathways Increases Neuromuscular Junction Stability and Endurance in Old Mice. Aging Dis. 2024, 15, 893–910. [Google Scholar] [CrossRef] [PubMed]
  169. Ueta, R.; Sugita, S.; Minegishi, Y.; Shimotoyodome, A.; Ota, N.; Ogiso, N.; Eguchi, T.; Yamanashi, Y. DOK7 Gene Therapy Enhances Neuromuscular Junction Innervation and Motor Function in Aged Mice. iScience 2020, 23, 101385. [Google Scholar] [CrossRef]
  170. Ozes, B.; Tong, L.; Myers, M.; Moss, K.; Ridgley, A.; Sahenk, Z. AAV1.NT-3 gene therapy prevents age-related sarcopenia. Aging 2023, 15, 1306–1329. [Google Scholar] [CrossRef]
  171. Pin, F.; Huot, J.R.; Bonetto, A. The Mitochondria-Targeting Agent MitoQ Improves Muscle Atrophy, Weakness and Oxidative Metabolism in C26 Tumor-Bearing Mice. Front. Cell. Dev. Biol. 2022, 10, 861622. [Google Scholar] [CrossRef]
  172. Braakhuis, A.J.; Nagulan, R.; Somerville, V. The Effect of MitoQ on Aging-Related Biomarkers: A Systematic Review and Meta-Analysis. Oxid. Med. Cell. Longev. 2018, 2018, 8575263. [Google Scholar] [CrossRef] [PubMed]
  173. Fu, Y.; Tao, L.; Wang, X.; Wang, B.; Qin, W.; Song, L. PGC-1alpha participates in regulating mitochondrial function in aged sarcopenia through effects on the Sestrin2-mediated mTORC1 pathway. Exp. Gerontol. 2024, 190, 112428. [Google Scholar] [CrossRef]
  174. Song, L.; Xue, J.; Xu, L.; Cheng, L.; Zhang, Y.; Wang, X. Muscle-specific PGC-1alpha modulates mitochondrial oxidative stress in aged sarcopenia through regulating Nrf2. Exp. Gerontol. 2024, 193, 112468. [Google Scholar] [CrossRef] [PubMed]
  175. Wang, Q.; Cui, C.; Zhang, N.; Lin, W.; Chai, S.; Chow, S.K.; Wong, R.M.Y.; Hu, Y.; Law, S.W.; Cheung, W.H. Effects of physical exercise on neuromuscular junction degeneration during ageing: A systematic review. J. Orthop. Translat. 2024, 46, 91–102. [Google Scholar] [CrossRef]
  176. Monti, E.; Tagliaferri, S.; Zampieri, S.; Sarto, F.; Sirago, G.; Franchi, M.V.; Ticinesi, A.; Longobucco, Y.; Adorni, E.; Lauretani, F.; et al. Effects of a 2-year exercise training on neuromuscular system health in older individuals with low muscle function. J. Cachexia Sarcopenia Muscle 2023, 14, 794–804. [Google Scholar] [CrossRef]
  177. Xhuti, D.; Nilsson, M.I.; Manta, K.; Tarnopolsky, M.A.; Nederveen, J.P. Circulating exosome-like vesicle and skeletal muscle microRNAs are altered with age and resistance training. J. Physiol. 2023, 601, 5051–5073. [Google Scholar] [CrossRef]
  178. Aoi, W.; Tanimura, Y. Roles of Skeletal Muscle-Derived Exosomes in Organ Metabolic and Immunological Communication. Front. Endocrinol. 2021, 12, 697204. [Google Scholar] [CrossRef]
  179. Li, X.; Zhang, H.; Wang, X.; Lu, M.; Ding, Q.; Chen, A.F.; Xiang, M.; Chen, S. iPSC-derived exosomes promote angiogenesis in naturally aged mice. Aging 2023, 15, 5854–5872. [Google Scholar] [CrossRef]
  180. Wan, R.; Liu, S.; Feng, X.; Luo, W.; Zhang, H.; Wu, Y.; Chen, S.; Shang, X. The Revolution of exosomes: From biological functions to therapeutic applications in skeletal muscle diseases. J. Orthop. Translat. 2024, 45, 132–139. [Google Scholar] [CrossRef]
  181. Yuan, S.; Larsson, S.C. Epidemiology of sarcopenia: Prevalence, risk factors, and consequences. Metabolism 2023, 144, 155533. [Google Scholar] [CrossRef]
  182. Field, D.; Gueldner, S.H. The oldest-old: How do they differ from the old-old? J. Gerontol. Nurs. 2001, 27, 20–27. [Google Scholar] [CrossRef] [PubMed]
  183. Tsoi, C.S.; Chow, J.Y.; Choi, K.S.; Li, H.W.; Nie, J.X.; Tracy, C.S.; Wang, L.; Upshur, R.E. Medical characteristics of the oldest old: Retrospective chart review of patients aged 85+ in an academic primary care centre. BMC Res. Notes 2014, 7, 340. [Google Scholar] [CrossRef] [PubMed]
  184. Salinas-Rodriguez, A.; Palazuelos-Gonzalez, R.; Gonzalez-Bautista, E.; Manrique-Espinoza, B. Sarcopenia, Cognitive Function, and the Heterogeneity in Aging. J. Nutr. Health Aging 2023, 27, 240–242. [Google Scholar] [CrossRef] [PubMed]
  185. Cacciatore, S.; Duque, G.; Marzetti, E. Osteosarcopenic obesity: A triple threat for older adults? Eur. Geriatr. Med. 2023, 14, 1191–1193. [Google Scholar] [CrossRef]
Figure 1. Overview of emerging pathways and potential therapeutic approaches for age-related sarcopenia. Abbreviations: Ach, acetylcholine; AchE, acetylcholinesterase; Akt, protein kinase B; ATP, adenosine triphosphate; IGF-1, insulin-like growth factor 1; mTOR: mechanistic target of rapamycin; MuSK, muscle-specific kinase; NAD+, nicotinamide adenine dinucleotide; NMJ, neuromuscular junction; PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1-alpha; PI3K, phosphoinositide 3-kinase; SCFA, short-chain fatty acids.
Figure 1. Overview of emerging pathways and potential therapeutic approaches for age-related sarcopenia. Abbreviations: Ach, acetylcholine; AchE, acetylcholinesterase; Akt, protein kinase B; ATP, adenosine triphosphate; IGF-1, insulin-like growth factor 1; mTOR: mechanistic target of rapamycin; MuSK, muscle-specific kinase; NAD+, nicotinamide adenine dinucleotide; NMJ, neuromuscular junction; PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1-alpha; PI3K, phosphoinositide 3-kinase; SCFA, short-chain fatty acids.
Nutrients 16 03271 g001
Figure 2. Age-related changes in neuromuscular junction potentially contributing to sarcopenia. Abbreviations: nAchR, nicotinic acetylcholine receptors; MuSK, muscle-specific kinase.
Figure 2. Age-related changes in neuromuscular junction potentially contributing to sarcopenia. Abbreviations: nAchR, nicotinic acetylcholine receptors; MuSK, muscle-specific kinase.
Nutrients 16 03271 g002
Table 1. Nutritional strategies for sarcopenia and their effects on muscle health.
Table 1. Nutritional strategies for sarcopenia and their effects on muscle health.
Nutritional
Intervention
Key Nutrients/
Components
Effects on MPSEffects on
Inflammation
Effects on Muscle
Regeneration
References
Protein intakeHigh-quality protein (e.g., whey, casein)Stimulates MPS, essential for muscle repair and growthMay reduce inflammation when combined with exerciseSupports overall muscle regeneration[49,50]
BCAA supplementationBCAA, particularly leucineStimulates MPS via the mTORC1 pathwayReduces markers of inflammationEnhances satellite cell activation and muscle repair[50,51]
Omega-3 fatty acidsEPA, DHAModulate MPS, especially in response to resistance trainingStrong anti-inflammatory effects, reduce cytokine levelsEnhance muscle regeneration through improved mitochondrial function[52,53]
HMBMetabolite of leucinePromotes MPS, decreases muscle protein breakdownAnti-inflammatory effects by inhibiting NF-κB signalingEnhances muscle mass and function in older adults[54,55]
CreatineCreatine monohydrateSupports rapid ATP production, increases training capacityIndirectly reduces inflammation by improving muscle bioenergeticsEnhances satellite cell proliferation and muscle fiber repair[56,57]
ArginineAmino acid, precursor of NOPromotes MPS via the mTORC1 pathwayVasodilatory and anti-inflammatory effects via increased NO productionImproves blood flow to muscles, supporting regeneration and recovery[58,59]
Vitamin DCholecalciferol, ergocalciferolEnhances MPS, especially in individuals with low baseline levelsModulates immune response, potentially reducing inflammationSupports muscle cell growth and differentiation[49,60]
AntioxidantsVitamin C, Vitamin E, Coenzyme Q10May support MPS indirectly by reducing oxidative damage to muscle cellsReduce oxidative stress and inflammationProtect muscle cells from oxidative damage, supports recovery[52,61]
Dietary nitratesBeetroot juice, spinach, arugulaImprove mitochondrial efficiency, indirectly supporting MPSReduce blood pressure and inflammationEnhance blood flow to muscles, supports muscle endurance[62,63]
Abbreviations: ATP, adenosine triphosphate; BCAA, branched-chain amino acid; DHA, docosahexaenoic acid; EPA, eicosapentaenoic acid; HMB, β-hydroxy β-methylbutyrate; MPS, muscle protein synthesis; mTORC1, mechanistic target of rapamycin complex 1; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NO, nitric oxide.
Table 3. Emerging pharmacological interventions for sarcopenia.
Table 3. Emerging pharmacological interventions for sarcopenia.
Pharmacological
Intervention
Mechanism(s)
of Action
Potential BenefitsChallenges/
Side-Effects
References
Myostatin inhibitorsBlock myostatin signaling, which inhibits muscle growthIncrease muscle mass and strengthInconsistent results in improving muscle function and reducing physical frailty in clinical trials. Significant side-effects.[19,25]
Anti-inflammatory therapiesReduce chronic inflammation which impairs muscle regenerationPotentially enhance muscle regeneration and reduce muscle atrophySignificant side-effects, especially in older adults (e.g., NSAIDs),[30,157]
Stem cell therapyPromotes muscle regeneration by leveraging stem cell populationsEnhances muscle repair and regeneration through cell-based therapiesPromising results from clinical trials; further data on efficacy and safety are needed.[44,46]
Gene editing (CRISPR/Cas9)Targets genetic pathways to enhance muscle growth and repairPotential to correct genetic mutations and increase muscle massEthical considerations and early-stage clinical applications.[42,43]
Mitochondrial enhancersImprove mitochondrial biogenesis and functionEnhance muscle energy metabolism and reduce muscle fatigueRequire further validation in clinical settings.[61,78]
Selective androgen
receptor modulators
Promote muscle growth without the adverse effects of anabolic steroidsIncrease muscle mass and strengthPotential risks and long-term safety concerns.[85,156]
Acetylcholinesterase inhibitorsPrevent the breakdown of acetylcholine, enhancing neuromuscular junction functionMitigate muscle weakness associated with sarcopeniaSide-effects related to acetylcholine metabolism.[165,168]
MicroRNA modulationModulates gene expression to enhance muscle regenerationNovel approach to target specific pathways involved in muscle regenerationEarly stage of research; potential off-target effects.[43,48]
Exosome-based therapiesUtilize small extracellular vesicles for targeted delivery of bioactive moleculesPromote NMJ repair and muscle regenerationStill in early research stages; potential delivery challenges.[177,180]
Abbreviations: CRISPR, Clustered Regularly Interspaced Short Palindromic Repeats, NMJ, neuromuscular junction; NSAIDs, non-steroidal anti-inflammatory drugs.
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

Cacciatore, S.; Calvani, R.; Esposito, I.; Massaro, C.; Gava, G.; Picca, A.; Tosato, M.; Marzetti, E.; Landi, F. Emerging Targets and Treatments for Sarcopenia: A Narrative Review. Nutrients 2024, 16, 3271. https://doi.org/10.3390/nu16193271

AMA Style

Cacciatore S, Calvani R, Esposito I, Massaro C, Gava G, Picca A, Tosato M, Marzetti E, Landi F. Emerging Targets and Treatments for Sarcopenia: A Narrative Review. Nutrients. 2024; 16(19):3271. https://doi.org/10.3390/nu16193271

Chicago/Turabian Style

Cacciatore, Stefano, Riccardo Calvani, Ilaria Esposito, Claudia Massaro, Giordana Gava, Anna Picca, Matteo Tosato, Emanuele Marzetti, and Francesco Landi. 2024. "Emerging Targets and Treatments for Sarcopenia: A Narrative Review" Nutrients 16, no. 19: 3271. https://doi.org/10.3390/nu16193271

APA Style

Cacciatore, S., Calvani, R., Esposito, I., Massaro, C., Gava, G., Picca, A., Tosato, M., Marzetti, E., & Landi, F. (2024). Emerging Targets and Treatments for Sarcopenia: A Narrative Review. Nutrients, 16(19), 3271. https://doi.org/10.3390/nu16193271

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