Abstract

In an ageing society, the importance of maintaining healthy life expectancy has been emphasized. As a result of age-related decline in functional reserve, frailty is a state of increased vulnerability and susceptibility to adverse health outcomes with a serious impact on healthy life expectancy. The decline in skeletal muscle mass and function, also known as sarcopenia, is key in the development of physical frailty. Both frailty and sarcopenia are highly prevalent in patients not only with advanced age but also in patients with illnesses that exacerbate their progression like heart failure (HF), cancer, or dementia, with the prevalence of frailty and sarcopenia in HF patients reaching up to 50–75% and 19.5–47.3%, respectively, resulting in 1.5–3 times higher 1-year mortality. The biological mechanisms of frailty and sarcopenia are multifactorial, complex, and not yet fully elucidated, ranging from DNA damage, proteostasis impairment, and epigenetic changes to mitochondrial dysfunction, cellular senescence, and environmental factors, many of which are further linked to cardiac disease. Currently, there is no gold standard for the treatment of frailty and sarcopenia, however, growing evidence supports that a combination of exercise training and nutritional supplement improves skeletal muscle function and frailty, with a variety of other therapies being devised based on the underlying pathophysiology. In this review, we address the involvement of frailty and sarcopenia in cardiac disease and describe the latest insights into their biological mechanisms as well as the potential for intervention through exercise, diet, and specific therapies.

Schematic diagram of the biological mechanisms of the development of frailty and sarcopenia, their comorbidities, and potential reversibility by therapeutic intervention. CAD, coronary artery disease; HF, heart failure.
Graphical Abstract

Schematic diagram of the biological mechanisms of the development of frailty and sarcopenia, their comorbidities, and potential reversibility by therapeutic intervention. CAD, coronary artery disease; HF, heart failure.

1. Introduction

The rapid ageing of societies worldwide leads to attention increasingly focusing on frailty, a state of increased vulnerability to health problems due to age-related functional decline and loss of reserve capacity.1 By 2050, the percentage of the population aged 65 and over is expected to double as compared to 2015 levels,2 and the prevalence of frailty is consequently expected to continue to increase for the foreseeable future.3 Whilst frailty embraces many aspects including physical, social, and psychological factors, one phenotype associated with the occurrence of physical frailty is sarcopenia, a geriatric syndrome defined as age-related (primary sarcopenia) or chronic disease-related (secondary sarcopenia) loss of skeletal muscle mass and muscle strength.4,5 Recently, frailty and sarcopenia have been reported to exacerbate the clinical outcomes of numerous illnesses, and the same has been shown for major cardiac diseases, such as heart failure (HF), and coronary artery disease (CAD).6,7 On the other hand, since sarcopenia and frailty are potentially reversible with early intervention,8 a better understanding of their biological basis and appropriate therapeutic interventions are crucial to improve prognosis. This review outlines the current findings on the biological basis of frailty and sarcopenia, their association with cardiac disease, and some promising therapeutic approaches (Table 1).

Table 1

Confirmed and unresolved issues in sarcopenia and frailty

IssuesWhether the issues are confirmed or unresolvedReferences
Definition of sarcopenia and frailtyWith numerous definitions of sarcopenia and frailty, the precise and appropriate methods and cut-off values for their diagnosis remain an unmet challenge.1,9–12
Involvement in frailty and sarcopenia in cardiac diseaseFrailty and sarcopenia are independently associated with the risk of developing CAD and HF.13–16
Genomic instability: There is some evidence that genomic instability is associated with sarcopenia and frailty, although the causality is not elucidated.17,18
Telomere attrition: Telomere length is short in patients with HF and has been associated with disease severity. However, there is no consistent evidence of a relationship between telomere shortening and sarcopenia or frailty.19–22
Epigenetic alterations: Several studies have shown that changes in DNA methylation levels, methylation patterns, and miRNAs during the lifespan may play a role in frailty and age-related skeletal muscle dysfunction, although the causal relationship remains unclear.23,24
Loss of proteostasis
Molecular chaperones: Several HSP maintains muscle function and regulates muscle atrophy.
25,26
Proteasomal degradation: E3 ubiquitin ligase activity is elevated and is associated with decreased cardiac contractility in chronic HF. However, conflicting evidence exists for the role of UPS in sarcopenia.27–31
Autophagy: Autophagy declines with age. Autophagy inhibition induces cardiac hypertrophy, left ventricular dilatation, and cardiac dysfunction. Autophagy dysfunction, i.e. both under- and over-activation can contribute to the development of sarcopenia.32–36
Biological mechanisms of frailty and sarcopeniaDeregulated nutrient sensing: Low GH, IGF-1, and testosterone levels are associated with impaired cardiac performance and exercise capacity in HF patients. GH and IGF-1 play a pivotal role in protein synthesis and muscle growth, and the association of declines in anabolic hormones with sarcopenia and frailty has been widely examined.37–40
Mitochondrial dysfunction: Mitochondrial dysfunction is a common feature of many aspects of ageing, and is associated with the development of various age-related metabolic diseases, including HF and sarcopenia. Oxidative stress is enhanced in patients with HF, contributing to disease severity and exercise intolerance by causing intracellular Ca2+ overload in cardiac myocytes, hypertrophy, apoptosis, and endothelial dysfunction. The role of ROS in mitochondrial dysfunction, however, remains a significant challenge in ageing research.41–49
Cellular senescence and stem cell exhaustion: p16INK4a and p19ARF are both encoded by the INK4a/ARF locus, which has been identified as the locus most genetically related to age-related diseases such as CAD and frailty. Excessive senescent cell accumulation plays a crucial role in driving tissue senescence, which contributes to the development of several age-related pathologies such as frailty and sarcopenia. The role of satellite cells in sarcopenia and frailty remains controversial.50–54
Altered intercellular communication: Chronic low-level inflammation associated with ageing is related to factors closely associated with physical frailty, such as loss of muscle mass, muscle strength, and bone mineral density. HF patients have chronic low-level systemic inflammation, which promotes cardiac dysfunction and remodelling, further exacerbating HF itself. Enhanced RAS can contribute to muscle wasting by increasing ROS production and inducing the expression of TGF-β1 and connective tissue growth factor.55–59
Environmental factors: Smoking, poor dietary patterns, lack of exercise, and a sedentary lifestyle have been implicated in the development and exacerbation of CAD and frailty.60–65
Treatment strategiesCurrently, there is no gold standard for the treatment of frailty and sarcopenia, however, exercise training, nutritional therapies, and disease-specific therapies are certainly the primary treatments for frailty and sarcopenia.
Exercise: Resistance training has beneficial and significant effects on functional capacity, muscle mass, and strength for the prevention and treatment of sarcopenia. The multicomponent intervention is associated with a reduction in the incidence of mobility disability in frail sarcopenic elderly subjects. Data from several exercise intervention trials have shown that supervised exercise training is safe and useful for improving exercise capacity and quality of life in patients with HF.66–68
Nutrition: Energy expenditure is reduced as a protective mechanism in malnutrition whereas it remains inappropriately high in sarcopenia and muscle wasting. Also, increased resting energy expenditure has been shown in HF. High protein intake is generally recommended in elderly patients with sarcopenia and HF, but special attention needs to consider chronic kidney disease, which frequently coexists with these conditions.69–72
Drug therapy: There are no pharmacologic agents approved worldwide for the treatment of frailty and sarcopenia. Hormonal treatment is still controversial in its practical application due to uncertainties in efficacy, the need for continuous administration, and side effects. The therapeutic effectiveness of myostatin inhibitors on sarcopenia has only been limited. Ghrelin administration could improve functional capacity, muscle strength, and mass as well as left ventricular ejection fraction in sarcopenic HF patients. GDF-15 neutralization is a transformative therapeutic approach that may restore muscle function and physical performance.73–79
IssuesWhether the issues are confirmed or unresolvedReferences
Definition of sarcopenia and frailtyWith numerous definitions of sarcopenia and frailty, the precise and appropriate methods and cut-off values for their diagnosis remain an unmet challenge.1,9–12
Involvement in frailty and sarcopenia in cardiac diseaseFrailty and sarcopenia are independently associated with the risk of developing CAD and HF.13–16
Genomic instability: There is some evidence that genomic instability is associated with sarcopenia and frailty, although the causality is not elucidated.17,18
Telomere attrition: Telomere length is short in patients with HF and has been associated with disease severity. However, there is no consistent evidence of a relationship between telomere shortening and sarcopenia or frailty.19–22
Epigenetic alterations: Several studies have shown that changes in DNA methylation levels, methylation patterns, and miRNAs during the lifespan may play a role in frailty and age-related skeletal muscle dysfunction, although the causal relationship remains unclear.23,24
Loss of proteostasis
Molecular chaperones: Several HSP maintains muscle function and regulates muscle atrophy.
25,26
Proteasomal degradation: E3 ubiquitin ligase activity is elevated and is associated with decreased cardiac contractility in chronic HF. However, conflicting evidence exists for the role of UPS in sarcopenia.27–31
Autophagy: Autophagy declines with age. Autophagy inhibition induces cardiac hypertrophy, left ventricular dilatation, and cardiac dysfunction. Autophagy dysfunction, i.e. both under- and over-activation can contribute to the development of sarcopenia.32–36
Biological mechanisms of frailty and sarcopeniaDeregulated nutrient sensing: Low GH, IGF-1, and testosterone levels are associated with impaired cardiac performance and exercise capacity in HF patients. GH and IGF-1 play a pivotal role in protein synthesis and muscle growth, and the association of declines in anabolic hormones with sarcopenia and frailty has been widely examined.37–40
Mitochondrial dysfunction: Mitochondrial dysfunction is a common feature of many aspects of ageing, and is associated with the development of various age-related metabolic diseases, including HF and sarcopenia. Oxidative stress is enhanced in patients with HF, contributing to disease severity and exercise intolerance by causing intracellular Ca2+ overload in cardiac myocytes, hypertrophy, apoptosis, and endothelial dysfunction. The role of ROS in mitochondrial dysfunction, however, remains a significant challenge in ageing research.41–49
Cellular senescence and stem cell exhaustion: p16INK4a and p19ARF are both encoded by the INK4a/ARF locus, which has been identified as the locus most genetically related to age-related diseases such as CAD and frailty. Excessive senescent cell accumulation plays a crucial role in driving tissue senescence, which contributes to the development of several age-related pathologies such as frailty and sarcopenia. The role of satellite cells in sarcopenia and frailty remains controversial.50–54
Altered intercellular communication: Chronic low-level inflammation associated with ageing is related to factors closely associated with physical frailty, such as loss of muscle mass, muscle strength, and bone mineral density. HF patients have chronic low-level systemic inflammation, which promotes cardiac dysfunction and remodelling, further exacerbating HF itself. Enhanced RAS can contribute to muscle wasting by increasing ROS production and inducing the expression of TGF-β1 and connective tissue growth factor.55–59
Environmental factors: Smoking, poor dietary patterns, lack of exercise, and a sedentary lifestyle have been implicated in the development and exacerbation of CAD and frailty.60–65
Treatment strategiesCurrently, there is no gold standard for the treatment of frailty and sarcopenia, however, exercise training, nutritional therapies, and disease-specific therapies are certainly the primary treatments for frailty and sarcopenia.
Exercise: Resistance training has beneficial and significant effects on functional capacity, muscle mass, and strength for the prevention and treatment of sarcopenia. The multicomponent intervention is associated with a reduction in the incidence of mobility disability in frail sarcopenic elderly subjects. Data from several exercise intervention trials have shown that supervised exercise training is safe and useful for improving exercise capacity and quality of life in patients with HF.66–68
Nutrition: Energy expenditure is reduced as a protective mechanism in malnutrition whereas it remains inappropriately high in sarcopenia and muscle wasting. Also, increased resting energy expenditure has been shown in HF. High protein intake is generally recommended in elderly patients with sarcopenia and HF, but special attention needs to consider chronic kidney disease, which frequently coexists with these conditions.69–72
Drug therapy: There are no pharmacologic agents approved worldwide for the treatment of frailty and sarcopenia. Hormonal treatment is still controversial in its practical application due to uncertainties in efficacy, the need for continuous administration, and side effects. The therapeutic effectiveness of myostatin inhibitors on sarcopenia has only been limited. Ghrelin administration could improve functional capacity, muscle strength, and mass as well as left ventricular ejection fraction in sarcopenic HF patients. GDF-15 neutralization is a transformative therapeutic approach that may restore muscle function and physical performance.73–79

CAD, coronary artery disease; GDF-15, Growth differentiation factor-15; GH, growth hormone; HF, heart failure; HSP, heat-shock family of proteins; IGF-1, insulin-like growth factor-1; miRNAs, microRNAs; RAS, renin-angiotensin system; ROS, reactive oxygen species; TGF, transforming growth factor; TNF, tumour necrosis factor; UPS, ubiquitin-proteasome system.

Table 1

Confirmed and unresolved issues in sarcopenia and frailty

IssuesWhether the issues are confirmed or unresolvedReferences
Definition of sarcopenia and frailtyWith numerous definitions of sarcopenia and frailty, the precise and appropriate methods and cut-off values for their diagnosis remain an unmet challenge.1,9–12
Involvement in frailty and sarcopenia in cardiac diseaseFrailty and sarcopenia are independently associated with the risk of developing CAD and HF.13–16
Genomic instability: There is some evidence that genomic instability is associated with sarcopenia and frailty, although the causality is not elucidated.17,18
Telomere attrition: Telomere length is short in patients with HF and has been associated with disease severity. However, there is no consistent evidence of a relationship between telomere shortening and sarcopenia or frailty.19–22
Epigenetic alterations: Several studies have shown that changes in DNA methylation levels, methylation patterns, and miRNAs during the lifespan may play a role in frailty and age-related skeletal muscle dysfunction, although the causal relationship remains unclear.23,24
Loss of proteostasis
Molecular chaperones: Several HSP maintains muscle function and regulates muscle atrophy.
25,26
Proteasomal degradation: E3 ubiquitin ligase activity is elevated and is associated with decreased cardiac contractility in chronic HF. However, conflicting evidence exists for the role of UPS in sarcopenia.27–31
Autophagy: Autophagy declines with age. Autophagy inhibition induces cardiac hypertrophy, left ventricular dilatation, and cardiac dysfunction. Autophagy dysfunction, i.e. both under- and over-activation can contribute to the development of sarcopenia.32–36
Biological mechanisms of frailty and sarcopeniaDeregulated nutrient sensing: Low GH, IGF-1, and testosterone levels are associated with impaired cardiac performance and exercise capacity in HF patients. GH and IGF-1 play a pivotal role in protein synthesis and muscle growth, and the association of declines in anabolic hormones with sarcopenia and frailty has been widely examined.37–40
Mitochondrial dysfunction: Mitochondrial dysfunction is a common feature of many aspects of ageing, and is associated with the development of various age-related metabolic diseases, including HF and sarcopenia. Oxidative stress is enhanced in patients with HF, contributing to disease severity and exercise intolerance by causing intracellular Ca2+ overload in cardiac myocytes, hypertrophy, apoptosis, and endothelial dysfunction. The role of ROS in mitochondrial dysfunction, however, remains a significant challenge in ageing research.41–49
Cellular senescence and stem cell exhaustion: p16INK4a and p19ARF are both encoded by the INK4a/ARF locus, which has been identified as the locus most genetically related to age-related diseases such as CAD and frailty. Excessive senescent cell accumulation plays a crucial role in driving tissue senescence, which contributes to the development of several age-related pathologies such as frailty and sarcopenia. The role of satellite cells in sarcopenia and frailty remains controversial.50–54
Altered intercellular communication: Chronic low-level inflammation associated with ageing is related to factors closely associated with physical frailty, such as loss of muscle mass, muscle strength, and bone mineral density. HF patients have chronic low-level systemic inflammation, which promotes cardiac dysfunction and remodelling, further exacerbating HF itself. Enhanced RAS can contribute to muscle wasting by increasing ROS production and inducing the expression of TGF-β1 and connective tissue growth factor.55–59
Environmental factors: Smoking, poor dietary patterns, lack of exercise, and a sedentary lifestyle have been implicated in the development and exacerbation of CAD and frailty.60–65
Treatment strategiesCurrently, there is no gold standard for the treatment of frailty and sarcopenia, however, exercise training, nutritional therapies, and disease-specific therapies are certainly the primary treatments for frailty and sarcopenia.
Exercise: Resistance training has beneficial and significant effects on functional capacity, muscle mass, and strength for the prevention and treatment of sarcopenia. The multicomponent intervention is associated with a reduction in the incidence of mobility disability in frail sarcopenic elderly subjects. Data from several exercise intervention trials have shown that supervised exercise training is safe and useful for improving exercise capacity and quality of life in patients with HF.66–68
Nutrition: Energy expenditure is reduced as a protective mechanism in malnutrition whereas it remains inappropriately high in sarcopenia and muscle wasting. Also, increased resting energy expenditure has been shown in HF. High protein intake is generally recommended in elderly patients with sarcopenia and HF, but special attention needs to consider chronic kidney disease, which frequently coexists with these conditions.69–72
Drug therapy: There are no pharmacologic agents approved worldwide for the treatment of frailty and sarcopenia. Hormonal treatment is still controversial in its practical application due to uncertainties in efficacy, the need for continuous administration, and side effects. The therapeutic effectiveness of myostatin inhibitors on sarcopenia has only been limited. Ghrelin administration could improve functional capacity, muscle strength, and mass as well as left ventricular ejection fraction in sarcopenic HF patients. GDF-15 neutralization is a transformative therapeutic approach that may restore muscle function and physical performance.73–79
IssuesWhether the issues are confirmed or unresolvedReferences
Definition of sarcopenia and frailtyWith numerous definitions of sarcopenia and frailty, the precise and appropriate methods and cut-off values for their diagnosis remain an unmet challenge.1,9–12
Involvement in frailty and sarcopenia in cardiac diseaseFrailty and sarcopenia are independently associated with the risk of developing CAD and HF.13–16
Genomic instability: There is some evidence that genomic instability is associated with sarcopenia and frailty, although the causality is not elucidated.17,18
Telomere attrition: Telomere length is short in patients with HF and has been associated with disease severity. However, there is no consistent evidence of a relationship between telomere shortening and sarcopenia or frailty.19–22
Epigenetic alterations: Several studies have shown that changes in DNA methylation levels, methylation patterns, and miRNAs during the lifespan may play a role in frailty and age-related skeletal muscle dysfunction, although the causal relationship remains unclear.23,24
Loss of proteostasis
Molecular chaperones: Several HSP maintains muscle function and regulates muscle atrophy.
25,26
Proteasomal degradation: E3 ubiquitin ligase activity is elevated and is associated with decreased cardiac contractility in chronic HF. However, conflicting evidence exists for the role of UPS in sarcopenia.27–31
Autophagy: Autophagy declines with age. Autophagy inhibition induces cardiac hypertrophy, left ventricular dilatation, and cardiac dysfunction. Autophagy dysfunction, i.e. both under- and over-activation can contribute to the development of sarcopenia.32–36
Biological mechanisms of frailty and sarcopeniaDeregulated nutrient sensing: Low GH, IGF-1, and testosterone levels are associated with impaired cardiac performance and exercise capacity in HF patients. GH and IGF-1 play a pivotal role in protein synthesis and muscle growth, and the association of declines in anabolic hormones with sarcopenia and frailty has been widely examined.37–40
Mitochondrial dysfunction: Mitochondrial dysfunction is a common feature of many aspects of ageing, and is associated with the development of various age-related metabolic diseases, including HF and sarcopenia. Oxidative stress is enhanced in patients with HF, contributing to disease severity and exercise intolerance by causing intracellular Ca2+ overload in cardiac myocytes, hypertrophy, apoptosis, and endothelial dysfunction. The role of ROS in mitochondrial dysfunction, however, remains a significant challenge in ageing research.41–49
Cellular senescence and stem cell exhaustion: p16INK4a and p19ARF are both encoded by the INK4a/ARF locus, which has been identified as the locus most genetically related to age-related diseases such as CAD and frailty. Excessive senescent cell accumulation plays a crucial role in driving tissue senescence, which contributes to the development of several age-related pathologies such as frailty and sarcopenia. The role of satellite cells in sarcopenia and frailty remains controversial.50–54
Altered intercellular communication: Chronic low-level inflammation associated with ageing is related to factors closely associated with physical frailty, such as loss of muscle mass, muscle strength, and bone mineral density. HF patients have chronic low-level systemic inflammation, which promotes cardiac dysfunction and remodelling, further exacerbating HF itself. Enhanced RAS can contribute to muscle wasting by increasing ROS production and inducing the expression of TGF-β1 and connective tissue growth factor.55–59
Environmental factors: Smoking, poor dietary patterns, lack of exercise, and a sedentary lifestyle have been implicated in the development and exacerbation of CAD and frailty.60–65
Treatment strategiesCurrently, there is no gold standard for the treatment of frailty and sarcopenia, however, exercise training, nutritional therapies, and disease-specific therapies are certainly the primary treatments for frailty and sarcopenia.
Exercise: Resistance training has beneficial and significant effects on functional capacity, muscle mass, and strength for the prevention and treatment of sarcopenia. The multicomponent intervention is associated with a reduction in the incidence of mobility disability in frail sarcopenic elderly subjects. Data from several exercise intervention trials have shown that supervised exercise training is safe and useful for improving exercise capacity and quality of life in patients with HF.66–68
Nutrition: Energy expenditure is reduced as a protective mechanism in malnutrition whereas it remains inappropriately high in sarcopenia and muscle wasting. Also, increased resting energy expenditure has been shown in HF. High protein intake is generally recommended in elderly patients with sarcopenia and HF, but special attention needs to consider chronic kidney disease, which frequently coexists with these conditions.69–72
Drug therapy: There are no pharmacologic agents approved worldwide for the treatment of frailty and sarcopenia. Hormonal treatment is still controversial in its practical application due to uncertainties in efficacy, the need for continuous administration, and side effects. The therapeutic effectiveness of myostatin inhibitors on sarcopenia has only been limited. Ghrelin administration could improve functional capacity, muscle strength, and mass as well as left ventricular ejection fraction in sarcopenic HF patients. GDF-15 neutralization is a transformative therapeutic approach that may restore muscle function and physical performance.73–79

CAD, coronary artery disease; GDF-15, Growth differentiation factor-15; GH, growth hormone; HF, heart failure; HSP, heat-shock family of proteins; IGF-1, insulin-like growth factor-1; miRNAs, microRNAs; RAS, renin-angiotensin system; ROS, reactive oxygen species; TGF, transforming growth factor; TNF, tumour necrosis factor; UPS, ubiquitin-proteasome system.

2. Definition of frailty and sarcopenia

Frailty is defined as a geriatric syndrome characterized by reduced reserve and ability to cope with endogenous and exogenous stressors that lead to a cumulative impairment in response from multiple physiological systems, resulting in susceptibility and vulnerability to adverse outcomes.1,9 Although a plethora of different methods has been applied to identify frailty in a cardiovascular disease setting,9,10 the definition of frailty that has been widely used is based on the physical frailty phenotype, which includes parameters of unintentional weight loss, self-reported exhaustion, weakness, slow walking speed, and low level of physical activity.1 The diagnosis of frailty is confirmed when patients meet ≥ 3 criteria, while pre-frailty is identified as the presence of only one or two of the aforementioned criteria. Other models based on a multidimensional approach including functional, psychological, cognitive, and social components have been developed to overcome a simplified physical approach to frailty.10–12 To encompass all the physiological systems that frailty can affect, the Heart Failure Association of the European Society of Cardiology recently proposed an integral conceptual model of frailty in patients with HF based on four domains: clinical, physical-functional, psycho-cognitive, and social (right circle in Figure 1).9 On the other hand, the validity and reliability of frailty assessment tools are highly dependent on the setting and population in which they are developed and validated,80 and frailty assessment in critically ill patients, in particular, poses special challenges. During critical illness, there is often a reliance on proxy responders, and proxy assessments do not necessarily correspond to the patient's own evaluation of his or her functioning and quality of life.81 Frailty itself is also an independent risk factor for delirium, which can coexist with cognitive impairment and is associated with disability.82,83 Furthermore, there is an inherent risk of mistakenly attributing features of acute illness to underlying frailty. Challenges such as these inherent in the assessment of frailty in critically ill patients raise the need for further research on the feasibility and reliability of frailty assessment tools.

Comprehensive conceptual framework of frailty and sarcopenia. ADL, activities of daily living; ASM, appendicular skeletal muscle mass; SARC-F, strength, assistance walking, rise from a chair, climb stairs, and falls; SPPB, short physical performance battery; TUG, Timed-Up and Go test.
Figure 1

Comprehensive conceptual framework of frailty and sarcopenia. ADL, activities of daily living; ASM, appendicular skeletal muscle mass; SARC-F, strength, assistance walking, rise from a chair, climb stairs, and falls; SPPB, short physical performance battery; TUG, Timed-Up and Go test.

Sarcopenia is characterized by a global and progressive decline in muscle strength, skeletal muscle mass, and physical performance (left circle in Figure 1), often associated with increased risk of falls, physical disability, hospitalization, comorbidities, and premature death.84–86 In patients with HF, sarcopenia is prevalent in 19.5–47.3% of patients87–89 and associated with reduced bone mineral density, exercise capacity,87,90–92 endothelial dysfunction,93 and independently with poor survival.94 Prevalence values appear to be similar in patients with HF with preserved and reduced ejection fraction. Since 2016, sarcopenia has also been recognized as a disease and received an ICD-10 code (M62.84).95 This recognition has led to an increased awareness of sarcopenia with better screening and diagnostic tools as well as more interest from pharmaceutical companies in the development of new compounds to prevent and treat sarcopenia. Nonetheless, it is worth mentioning that a consensus about the definition of sarcopenia (e.g. methods, cut-off values, and adjustments used to determine the reduction in skeletal muscle mass and impaired muscle function) has not been met yet.96

In 1998, Baumgartner and colleagues proposed the first method to assess sarcopenia based on the sum of appendicular skeletal muscle mass (ASM) in both arms and legs divided by the height in metres squared (ASM/height2).97 Although, in accordance with the revised definition from the European Working Group on Sarcopenia in Older People (EWGSOP 2), muscle strength moved up as the first criterion to screen for sarcopenia with grip strength < 27 kg for men and grip strength < 16 kg for women, ASM/height2 is still used to confirm the diagnosis of sarcopenia (i.e. ASM/height2 ≤ 7.0 kg/m2 and ≤5.5 kg/m2 for men and women, respectively).84 In addition, physical performance assessed by gait speed (≤0.8 m/s), the short physical performance battery (SPPB ≤ 8 point score) test, the Timed-Up and Go test (TUG ≥ 20 s), or the 400 m walk test (≥6 min for completion) can be applied to determine the severity of sarcopenia.84 However, other authors proposed adjustments for skeletal muscle mass by total body mass,98 by body mass index (BMI),99 and by total fat mass100 with the aim to overcome limitations in the equation proposed by Baumgartner and colleagues that underestimated body fat mass, limiting its applicability in patients with combined sarcopenia and obesity, which has lately been termed sarcopenic obesity.101 In patients with chronic HF, a recent study has demonstrated that detecting sarcopenia with three different methods to correct the reduction in skeletal muscle mass (ASM/height2 vs. ASM/total body fat vs. ASM/BMI) confirmed earlier prevalence values of sarcopenia around 21% across all methods regardless of BMI.102 Thus, the methods adjusting ASM for fat mass and BMI may be better to detect sarcopenia in obese patients than Baumgartner’s method with ASM adjusted for height, showing that obese patients can be as affected by sarcopenia as lean patients when appropriate indices are applied.

The methods used to identify frailty and sarcopenia are still evolving to determine precise cut-points for parameters assessed in each condition. Some authors have also described sarcopenia as the central biological substrate for frailty, suggesting that the two conditions should be merged into a new clinical entity, called physical frailty and sarcopenia.103 Although there is a complex and multidimensional overlap between sarcopenia and frailty (Figure 1), there are gender differences in the contribution of sarcopenia to the frailty phenotype, and not all frail patients develop sarcopenia.104,105 Therefore, future studies should focus on applying accurate methods to classify as well as define the border between frailty and sarcopenia in patients with cardiac disease.

3. Involvement of frailty and sarcopenia in cardiac disease

The incidence of cardiovascular disease increases significantly with age, which coincides with changes in functional capacity and body composition due to the ageing process and progressing disease states. Moreover, the improved management of risk factors for cardiovascular disease as well as improved survival in patients with HF and CAD are leading to a longer life expectancy that modified the clinical profile of these patients towards more cumulative age-related conditions. Therefore, all patients with cardiac disease should be screened for frailty and sarcopenia regardless of chronological age.

Comorbidities, such as diabetes mellitus, hypertension, and other conventional risk factors for cardiovascular disease, are frequently associated with frailty in older adults, suggesting that there may be a shared pathophysiological mechanism possibly due to increased inflammation and metabolic dysregulation that can lead to a higher risk of cardiac disease in frail individuals. Indeed, frailty has been independently associated with the risk of developing peripheral vascular disease, acute myocardial infarction, CAD, and HF in the general population,13,14 suggesting that frailty, and even pre-frailty, can be recognized as risk factors for cardiovascular disease.106,107 The prevalence of frailty can reach up to 50–75% in patients with HF depending on the assessment tool used,108,109 whereas the prevalence of frailty in patients with CAD is around 25%.110 Additionally, sarcopenia has been implicated as both cause and consequence of diabetes mellitus,111 associated with cognitive impairment and depression,112,113 and associated with an increased risk for ischaemic heart disease.15,16 As may be noted, both frailty and sarcopenia are emerging as risk factors of cardiovascular disease and, due to their interweaving relationship, both conditions, associated or not, play an important role in the development of cardiac disease.

In patients with HF, a recent study comparing sex differences in prevalence and prognostic impact of both conditions showed that frailty was somewhat more prevalent in women (i.e. 61% vs. 54%) and sarcopenia had a higher prevalence in men (i.e. 24% vs. 14%), whereas frailty was associated with 1-year mortality only in men, and sarcopenia was associated with 1-year mortality in both sexes.114 Moreover, elderly patients with HF with preserved ejection fraction (HFpEF) showed a higher prevalence of frailty than patients with HF with reduced ejection fraction (HFrEF) (55% vs. 47%).115 Most of the differences between HFpEF and HFrEF were driven by deficits in the physical-functional domains such as handgrip strength (71.4% vs. 52.1%) and gait speed (82.6% vs. 65.2%), whereas no differences were detected in social and cognitive impairment.115 Thus, considering that women have a higher life expectancy, frailty, a condition strongly associated with age, may affect more women than men, whereas sarcopenia should be watched closer in males taking into account that sarcopenia can be an initial sign of alterations in the physical and functional component of frailty in patients with cardiac disease.

Although frailty may be characterized by a decline in reserve and response from many physiological systems to biological stressors, it has been shown that frailty is predominantly associated with left ventricular hypertrophy and impaired left ventricular systolic and diastolic function after accounting for abnormalities in other organ systems.116 Moreover, a SARC-F (Strength, Assistance walking, Rise from a chair, Climb stairs, and Falls) score ≥ 4 was associated with diastolic dysfunction accompanied by increased markers of inflammation in community-dwelling older adults.117 Indeed, there is an extremely strong connection between frailty, sarcopenia, and cardiac disease with a common pathophysiological mechanism that seems to be linked to increased circulating pro-inflammatory cytokines described across all these conditions.118 Below, we elaborate on several of the possible mechanisms that may explain the interplay among frailty, sarcopenia, and especially HF.

4. Biological mechanisms of frailty and sarcopenia

The pathophysiology of frailty and sarcopenia is inextricably involved with ageing, which is characterized by an inevitable time-dependent decline in physiological function. The hallmarks of ageing are classified into three main categories. Primary hallmarks: damage to cellular functions, such as genomic instability, telomere loss, epigenetic changes, and loss of proteostasis. Antagonistic hallmarks: metabolic responses to the primary hallmarks of ageing, include deregulated nutrient sensing, altered mitochondrial function, and cellular senescence. Integrative hallmarks: the physiological consequences of the primary and antagonistic hallmarks of ageing, ultimately resulting in age-related functional decline, which is mainly characterized by stem cell exhaustion and altered intercellular communication.119 These hallmarks of ageing contribute robustly to reduced physical performance and loss of reserve capacity. Environmental factors may also cause or exacerbate many of these mechanisms that may be triggers of frailty and sarcopenia.120 At the same time, the pathophysiology described above may be closely associated with the development and/or exacerbation of cardiac disease (Table 2).7,27,28,37,121–124 Furthermore, the combination of cardiac disease and geriatric syndromes can exacerbate one another and worsen prognosis significantly,7,121 making it crucial to comprehend the pathophysiology underlying these conditions in greater depth. The following overview provides possible biological mechanisms of frailty and sarcopenia based on well-established hallmarks of ageing, addressing their relevance to the pathogenesis of HF with recent findings.

Table 2

Shared biological mechanisms between frailty, sarcopenia, and HF

Biological mechanismsChanges in vivoContribution to the pathogenesis of HFReferences
Mitochondrial dysfunction↑ ROS
↓ ATP
Dysfunctional mitochondria increase ROS production, causing a mismatch between ATP production and demand, which can result in cardiac remodelling, inflammation, and diastolic dysfunction.122,125–127
Oxidative damage↑ mtDNA damageOxidative damage contributes to the severity of HF by causing intracellular Ca2+ overload in cardiac myocytes, cardiac hypertrophy, apoptosis, and endothelial dysfunction.44–46,125
Increased protein degradation↑ UPS (MuRF-1 and MAFbx)
↓ Autophagy
MuRF-1 and MAFbx are associated with impaired cardiac contractility under the regulation of TNF. Autophagy impairment leads to the accumulation of abnormal proteins, resulting in cardiac hypertrophy, left ventricular dilatation, and contractile dysfunction.27,28,123,128–130
Hormonal changes↓ GH/IGF-1
↓ Testosterone
↑ RAS
↑ GDF-15
Low GH, IGF-1, and testosterone levels are associated with impaired cardiac performance and exercise capacity in HF patients. Enhanced RAS activity advances cardiac dysfunction/remodelling. GDF-15 is expressed in myocytes in response to myocardial ischaemia and pressure overload.37,40,59,131–138
Inflammation↑ IL-1, IL-6, TNF
↑ NF-κB
↑ UPS
HF patients have chronic low-grade systemic inflammation. Inflammation promotes cardiac dysfunction/remodelling and exacerbates HF itself.55,57,58,139–143
Biological mechanismsChanges in vivoContribution to the pathogenesis of HFReferences
Mitochondrial dysfunction↑ ROS
↓ ATP
Dysfunctional mitochondria increase ROS production, causing a mismatch between ATP production and demand, which can result in cardiac remodelling, inflammation, and diastolic dysfunction.122,125–127
Oxidative damage↑ mtDNA damageOxidative damage contributes to the severity of HF by causing intracellular Ca2+ overload in cardiac myocytes, cardiac hypertrophy, apoptosis, and endothelial dysfunction.44–46,125
Increased protein degradation↑ UPS (MuRF-1 and MAFbx)
↓ Autophagy
MuRF-1 and MAFbx are associated with impaired cardiac contractility under the regulation of TNF. Autophagy impairment leads to the accumulation of abnormal proteins, resulting in cardiac hypertrophy, left ventricular dilatation, and contractile dysfunction.27,28,123,128–130
Hormonal changes↓ GH/IGF-1
↓ Testosterone
↑ RAS
↑ GDF-15
Low GH, IGF-1, and testosterone levels are associated with impaired cardiac performance and exercise capacity in HF patients. Enhanced RAS activity advances cardiac dysfunction/remodelling. GDF-15 is expressed in myocytes in response to myocardial ischaemia and pressure overload.37,40,59,131–138
Inflammation↑ IL-1, IL-6, TNF
↑ NF-κB
↑ UPS
HF patients have chronic low-grade systemic inflammation. Inflammation promotes cardiac dysfunction/remodelling and exacerbates HF itself.55,57,58,139–143

All these mechanisms up-regulate frailty and sarcopenia.

ATP, Adenosine triphosphate; GDF-15, Growth differentiation factor-15; GH, growth hormone; HF, heart failure; IGF-1, insulin-like growth factor-1; IL, interleukin; mtDNA, mitochondrial DNA; MuRF-1, muscle; NF-κB, nuclear factor-kappa B; RAS, renin-angiotensin system; TNF, tumour necrosis factor; UPS, ubiquitin-proteasome system.

Table 2

Shared biological mechanisms between frailty, sarcopenia, and HF

Biological mechanismsChanges in vivoContribution to the pathogenesis of HFReferences
Mitochondrial dysfunction↑ ROS
↓ ATP
Dysfunctional mitochondria increase ROS production, causing a mismatch between ATP production and demand, which can result in cardiac remodelling, inflammation, and diastolic dysfunction.122,125–127
Oxidative damage↑ mtDNA damageOxidative damage contributes to the severity of HF by causing intracellular Ca2+ overload in cardiac myocytes, cardiac hypertrophy, apoptosis, and endothelial dysfunction.44–46,125
Increased protein degradation↑ UPS (MuRF-1 and MAFbx)
↓ Autophagy
MuRF-1 and MAFbx are associated with impaired cardiac contractility under the regulation of TNF. Autophagy impairment leads to the accumulation of abnormal proteins, resulting in cardiac hypertrophy, left ventricular dilatation, and contractile dysfunction.27,28,123,128–130
Hormonal changes↓ GH/IGF-1
↓ Testosterone
↑ RAS
↑ GDF-15
Low GH, IGF-1, and testosterone levels are associated with impaired cardiac performance and exercise capacity in HF patients. Enhanced RAS activity advances cardiac dysfunction/remodelling. GDF-15 is expressed in myocytes in response to myocardial ischaemia and pressure overload.37,40,59,131–138
Inflammation↑ IL-1, IL-6, TNF
↑ NF-κB
↑ UPS
HF patients have chronic low-grade systemic inflammation. Inflammation promotes cardiac dysfunction/remodelling and exacerbates HF itself.55,57,58,139–143
Biological mechanismsChanges in vivoContribution to the pathogenesis of HFReferences
Mitochondrial dysfunction↑ ROS
↓ ATP
Dysfunctional mitochondria increase ROS production, causing a mismatch between ATP production and demand, which can result in cardiac remodelling, inflammation, and diastolic dysfunction.122,125–127
Oxidative damage↑ mtDNA damageOxidative damage contributes to the severity of HF by causing intracellular Ca2+ overload in cardiac myocytes, cardiac hypertrophy, apoptosis, and endothelial dysfunction.44–46,125
Increased protein degradation↑ UPS (MuRF-1 and MAFbx)
↓ Autophagy
MuRF-1 and MAFbx are associated with impaired cardiac contractility under the regulation of TNF. Autophagy impairment leads to the accumulation of abnormal proteins, resulting in cardiac hypertrophy, left ventricular dilatation, and contractile dysfunction.27,28,123,128–130
Hormonal changes↓ GH/IGF-1
↓ Testosterone
↑ RAS
↑ GDF-15
Low GH, IGF-1, and testosterone levels are associated with impaired cardiac performance and exercise capacity in HF patients. Enhanced RAS activity advances cardiac dysfunction/remodelling. GDF-15 is expressed in myocytes in response to myocardial ischaemia and pressure overload.37,40,59,131–138
Inflammation↑ IL-1, IL-6, TNF
↑ NF-κB
↑ UPS
HF patients have chronic low-grade systemic inflammation. Inflammation promotes cardiac dysfunction/remodelling and exacerbates HF itself.55,57,58,139–143

All these mechanisms up-regulate frailty and sarcopenia.

ATP, Adenosine triphosphate; GDF-15, Growth differentiation factor-15; GH, growth hormone; HF, heart failure; IGF-1, insulin-like growth factor-1; IL, interleukin; mtDNA, mitochondrial DNA; MuRF-1, muscle; NF-κB, nuclear factor-kappa B; RAS, renin-angiotensin system; TNF, tumour necrosis factor; UPS, ubiquitin-proteasome system.

4.1 Genomic instability

The integrity and stability of human DNA are always exposed to exogenous stressors, such as radiation and chemicals, and endogenous agents, including DNA replication errors, spontaneous hydrolysis reactions, and reactive oxygen species (ROS).144 The genetic lesions arising from mutagens vary extensively—small point mutations, translocations, chromosome gains, and breaks, telomere shortening, etc.—the organism has developed a complex network of DNA repair mechanisms that can fix most of these damages inflicted on DNA.145 However, some lesions are eventually not fully repaired, allowing DNA damage and mutations to accumulate over time. This is why genomic instability is a major hallmark of ageing, and genomic instability has been reported to be involved in a variety of ageing-related cardiovascular and neurodegenerative diseases.146,147 Skeletal muscle development and regeneration are regulated by skeletal muscle stem cells, called satellite cells (SCs).148 Therefore, the maintenance of satellite cell genomic integrity is a critical process directly related to the organization of skeletal muscle tissue. However, with ageing, DNA damage protection and repair systems begin to fail, and passenger mutations accumulate throughout the genome of SCs, leading to reduced replication capacity and ultimately to skeletal muscle defects. Taken together, a causal relationship between genomic instability and sarcopenia is expected; however, the ageing process in skeletal muscle is extremely complex, making a causal relationship with genomic instability difficult to clarify. Nevertheless, some evidence links genomic instability to sarcopenia and frailty: Higher levels of 8-oxo-7,8-dihydroguanine (8-oxoG), a key marker of DNA damage, in the vastus lateralis were observed in elderly compared to younger subjects.17 Sedentary older adults were also identified to have higher levels of 8-oxoG than those who were more active.17 Moreover, Sánchez-Flores et al.18 revealed that genomic instability, as assessed by lymphocyte micronucleus frequencies, was associated with frailty in the elderly. These findings are intriguing because they not only indicate that DNA instability, a hallmark of ageing, may be involved in the development of sarcopenia and frailty but also suggest a possible gene-protective effect of physical activity.

4.2 Telomere attrition

Telomeres are gene-poor nuclear protein structures located at the ends of linear chromosomes that shorten with age,149,150 and are associated not only with cellular ageing but also with many age-related diseases, including cancer, diabetes, and HF.19,151 In addition, telomere length is associated with disease severity in HF.19 Therefore, telomere length is widely considered a possible senescence biomarker, although it is still not fully elucidated whether telomere shortening is a cause or a consequence of physiological ageing or age-related diseases. The clinical significance of telomere shortening in sarcopenia and frailty has also been much discussed but still remains inconclusive. Based on nine articles that examined the association between telomere length of leucocytes and sarcopenia and frailty, Lorenzi et al.20 reported that there might be a correlation between telomere length and skeletal muscle mass, but not with muscle strength or frailty. Yet, since then, several findings have also contradicted this report.21,22 Such inconsistencies may be due not only to the fact that telomere length measured in peripheral blood not accurately reflecting that of skeletal muscle but may also imply that only a single biomarker of telomere shortening, caused by a variety of factors including oxidative stress, inflammation, and repeated cell replication, is insufficient to explain the extreme diversity of pathophysiology underlying the development of sarcopenia and frailty.

4.3 Epigenetic alterations

Epigenetics is defined as the study of mechanisms that can alter gene expression irrespective of changes in the DNA sequence,152 including DNA methylation, microRNAs (miRNAs), and histone modifications.153 These epigenetic mechanisms are influenced by environmental factors and play a part in the adaptive homeostasis of the organism. There is considerable evidence implying that epigenetic changes underlie age-related cellular deterioration,154 and Pal and Tyler155 mentioned that reduced bulk levels of the core histones, altered patterns of histone post-translational modifications and DNA methylation, replacement of canonical histones with histone variants, and altered non-coding RNA expression, are the characteristics of the aforementioned changes. DNA methylation has been identified as a possible biomarker of ageing that predicts the age of human cells and tissues, irrespective of health conditions, lifestyle, and other known genetic factors.156,157 Histone demethylases also regulate lifespan by targeting components such as the insulin/insulin-like growth factor-1 (IGF-1) signalling pathway.158 Moreover, miRNAs are involved in the regulation of insulin/IGF-1 and mammalian target of rapamycin (mTOR) signalling pathways, and their conserved role in ageing is well established.159,160 As can be seen from these findings, epigenetic alterations are one hallmark of ageing, whereas it has also been suggested that they are closely related to the degree of frailty and the development of sarcopenia. Some studies have demonstrated that frail individuals have lower levels of global DNA methylation than non-frail individuals and that methylation patterns and miRNA species could be involved in age-related impairment of skeletal muscle function.23,24 Although the causal relationship between changes in epigenetic patterns during the lifespan and the development of age-related diseases is still unclear, advances in this area of research should lead to a better understanding of the biological mechanisms of frailty in the elderly.

4.4 Loss of proteostasis

Proteostasis is a collection of cellular mechanisms involved in the maintenance of human protein homeostasis.161 Proteostasis fidelity is maintained by a proteostasis network consisting of chaperone-mediated folding, proteasomal degradation, and autophagy. All of these systems function in concert to repair the structure of misfolded polypeptides, degrading and removing them, preventing the accumulation of dysfunctional intracellular proteins, and ensuring the maintenance of proteome integrity.119 Many studies have demonstrated that deterioration of proteostasis with ageing results in misfolded protein aggregates and inclusions in tissues of the aged organisms, contributing to the development of age-related diseases such as Alzheimer's disease, Parkinson's disease, and cataracts.162 Similarly, skeletal muscle proteostasis impairment contributes significantly to the progression of muscle atrophy in sarcopenia.

Molecular chaperones are small proteins that bind to polypeptide chains in an unfolded, denatured state and assist in their folding into the proper protein structure and acquisition of normal function.163 Chaperone synthesis is severely impaired with ageing,164 and the most important molecular chaperones are the heat-shock family of proteins (HSP). In mammalian animals, the transcription factor heat shock factor 1 (HSF-1) is considered the master regulator of the heat shock response, and deacetylation of HSF-1 by Sirtuin-1 (SIRT-1) is known to potentiate the transcriptional activity of heat shock genes such as Hsp70.165 Meanwhile, pharmacological induction of the Hsp72, an inducible member of the Hsp70, preserved muscle function and slowed the progression of dystrophic pathology in mice models of Duchenne muscular dystrophy.25 In experiments using treadmill running, Hsp70 was induced in the vastus lateralis in an exercise intensity-dependent manner.26 Activation of SIRT-1 and/or inhibition of poly (ADP-ribose) polymerase-1 (PARP-1) have also been demonstrated to improve skeletal muscle mitochondrial biogenesis and performance in ageing mice.166 Up-regulation of molecular chaperones may therefore be a promising intervention to improve skeletal muscle performance in conditions such as age-related muscle atrophy and sarcopenia.

Increased protein degradation has been recognized as one of the possible mechanisms of skeletal muscle atrophy, with the ubiquitin-proteasome system (UPS) and the autophagy-lysosome system mainly regulating intracellular protein degradation in skeletal muscle cells (Figure 2).128 The degradation process is activated primarily by the p38 mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) pathways, as well as the glucocorticoid pathway.167,168 p38 MAPK activates the expression of a group of enzymes called muscle-specific E3 ubiquitin ligases, such as muscle RING-finger protein-1 (MuRF-1) and muscle atrophy F-Box (MAFbx), which mediate proteolysis by UPS,129,169 and elevated levels of these ubiquitin ligases are associated with muscle atrophy.130 Meanwhile, MuRF-1 and proteasome activity are elevated in the diaphragm and quadriceps of rats with chronic HF.27 Also, MuRF-1 and MAFbx are regulated by tumour necrosis factor (TNF) and associated with impaired cardiac contractility in chronic HF patients.28 These findings suggest that UPS may be one of the shared biological mechanisms in sarcopenia and HF. The transcription factor, NF-κB is activated by oxidative stress and inflammatory cytokines such as TNF,170 and its role in skeletal muscle atrophy is revealed by its ability to activate MuRF-1 expression.171 Activation of NF-κB in the muscle-specific transgenic expression of activated IkappaB kinase (IKK)-β mice caused marked muscle wasting due to accelerated proteolysis mediated by overexpression of MuRF-1, whereas pharmacological or genetic inhibition of the IKK-β/NF-κB/MuRF-1 pathway reversed muscle atrophy.171

Alterations in skeletal muscle metabolism with ageing and molecular mechanisms of frailty and sarcopenia. Bnip3, Bcl-2/adenovirus E1B 19 kDa-interacting protein 3; FOXO, forkhead box; GSK3, glycogen synthase kinase 3; IGF-1, insulin-like growth factor-1; KLF15, Krüppel-like factor 15; LC3, light chain 3; MAFbx, muscle atrophy F-Box; MAPK, mitogen-activated protein kinase; mTORC1, mammalian target of rapamycin complex-1; MuRF-1, RING-finger protein-1; NF-κB, nuclear factor-kappa B; PGC-1α, peroxisome proliferator-activated receptor gamma coactivator-1 alpha; PI3-K/Akt, Phosphoinositide 3-kinase/protein kinase B; Smad, suppressor of mothers against decapentaplegic; UPS, ubiquitin-proteasome system.
Figure 2

Alterations in skeletal muscle metabolism with ageing and molecular mechanisms of frailty and sarcopenia. Bnip3, Bcl-2/adenovirus E1B 19 kDa-interacting protein 3; FOXO, forkhead box; GSK3, glycogen synthase kinase 3; IGF-1, insulin-like growth factor-1; KLF15, Krüppel-like factor 15; LC3, light chain 3; MAFbx, muscle atrophy F-Box; MAPK, mitogen-activated protein kinase; mTORC1, mammalian target of rapamycin complex-1; MuRF-1, RING-finger protein-1; NF-κB, nuclear factor-kappa B; PGC-1α, peroxisome proliferator-activated receptor gamma coactivator-1 alpha; PI3-K/Akt, Phosphoinositide 3-kinase/protein kinase B; Smad, suppressor of mothers against decapentaplegic; UPS, ubiquitin-proteasome system.

The glucocorticoid pathway is another major pathway of protein catabolism. The forkhead box (FOXO) family of transcription factors that regulate genes involved in cellular processes such as cell cycle, apoptosis, and metabolism are elevated at gene expression levels in response to glucocorticoid treatment.172,173 Activated FOXO transcription factors up-regulate MuRF-1 and MAFbx to exert skeletal muscle catabolic effects.174,175 Glucocorticoids also induce transcription and stability of myostatin,176 a member of the transforming growth factor (TGF)-β family and a central negative regulator of muscle growth that inhibits protein synthesis by activating suppressor of mothers against decapentaplegic (Smad) proteins (Figure 2),177,178 thereby further pronouncing proteolysis. Furthermore, glucocorticoids regulate MAFbx and MuRF-1 expression via a transcription factor known as Krüppel-like factor 15 (KLF15).179 Ligand-bound glucocorticoid receptors induce transcription of KLF15, which interacts with KLF15-responsive elements in the promoter regions of MAFbx and MURF1 and induces their expression.179 KLF15 also triggers activation of MuRF-1 and MAFbx via stimulation of FOXO1 and FOXO3a expression. Taken together, the glucocorticoid pathway activates MAFbx and MuRF1 through a network of transcription factors, including the FOXO family and KLF15, which ultimately regulate muscle atrophy.179,180 Thus, UPS is a fundamental mechanism for protein degradation, but there is conflicting evidence for the role of UPS in ageing and sarcopenia. The expression levels of MAFbx and MuRF-1 in aged muscles have shown inconsistent results across studies, with an increase, no change, or even a decrease.29–31 Because even muscle hypertrophy increases both protein degradation and synthesis, the role of UPS in ageing and sarcopenia needs to be further elucidated, and the present data appear to suggest that other proteolytic pathways besides UPS play a more important role in protein degradation in these pathologies.

The autophagy-lysosome system is another major regulator of proteolysis. Removal of dysfunctional organelles and misfolded proteins by lysosomes plays a pivotal role in maintaining cell and tissue homeostasis.181 Age-related decline of this system is expected to be caused by decreased expression of autophagy-related genes,32 reduced levels of autophagy core components,182 and sustained mammalian target of rapamycin complex-1 (mTORC1) signalling, a well-established autophagy inhibitor.183 Autophagy is likewise important for the regulation of SCs,184 and age-related decline of autophagy function not only reduces the clearance of damaged and dysfunctional proteins but also leads to loss of SCs function, resulting in skeletal muscle loss from dysfunctional myogenesis.33 Furthermore, autophagy inhibition in autophagy-related 7 (Atg7) knockout mice induces up-regulation of MuRF-1 and MAFbx, accompanied by loss of myofibre size.185 This may explain the potential contribution of age-related blunting of lysosomal activity to sarcopenia, accompanied by a compensatory increase in proteolysis by the UPS.

On the other hand, a recent study suggests that general control non-depressible 2 (GCN2)/eukaryotic translation initiation factor 2A (eIF2A) signalling up-regulates autophagy in response to short-term deficiency of single essential amino acid, indicating that this pathway may contribute to cell survival and adaptation by regulating protein homeostasis under nutritional stress conditions.186 Some autophagy-related markers can be elevated in skeletal muscle cells in the catabolic state,34 and FOXO3 has been established as an important transcription factor that activates autophagy by regulating the expression of many autophagy-related genes, including microtubule-associated protein light chain 3 (LC3) and Bcl-2/adenovirus E1B 19 kDa-interacting protein 3 (Bnip3).187 Therefore, autophagy dysfunction, i.e. both under- and over-activation can contribute to the development of sarcopenia. Several effects of autophagy dysfunction on myocardium have also been reported; inhibition of autophagy in cardiomyocytes by autophagy-related 5 (Atg5) deletion induces hypertrophy, left ventricular dilatation, and contractile dysfunction.35 Administration of the autophagy inhibitor bafilomycin A1 to mice causes marked cardiac dysfunction and left ventricular dilatation in starved mice.36

4.5 Deregulated nutrient sensing

The nutrient-sensing pathway is responsible for regulating protein levels by controlling both protein production and degradation. Various anabolic and neurohormonal hormones are involved in protein metabolism, especially anabolic hormones such as insulin, growth hormone (GH), and its secondary mediator IGF-1, which play a pivotal role in protein synthesis and muscle growth.188 IGF-1 binding to transmembrane tyrosine kinase receptors on myocyte membranes activates the Phosphoinositide 3-kinase (PI3-K)/protein kinase B (Akt) pathway, which promotes protein synthesis and muscle hypertrophy by inhibiting glycogen synthase kinase 3 (GSK3) and activation of mTORC1 (Figure 2).189 In addition, activated PI3K/Akt phosphorylates and inactivates the downstream transcription factors FOXO, preventing protein degradation in skeletal muscle by suppressing MuRF-1 and MAFbx gene expression.175 Intriguingly, the FOXO transcription factors not only regulate transcription of MuRF-1 and MAFbx, which induce proteolysis by UPS, but also regulate transcription of core components such as Bnip3, which promotes the autophagy-lysosome system,187 whilst myostatin reverses the PI3K/Akt pathway by inhibiting Akt-phosphorylation and induces atrophy-related genes by increasing FOXO1 expression.190 Therefore, inhibition of the FOXO transcription factors, which play a crucial role in the development of skeletal muscle proteolysis, is a fascinating approach to sarcopenia, and it was recently reported that vitamin D not only suppresses the transcriptional activity of FOXO1 in myoblasts but also the gene expression of ubiquitin ligase and lysosomal proteolytic enzymes.191 On the other hand, the association of declines in anabolic hormones such as GH, IGF-1, and androgens with sarcopenia and frailty has been widely examined, and the replacement of these anabolic hormones has attracted much interest as a treatment for these pathologies.38,39,192,193 Low GH, IGF-1, and testosterone levels are also associated with impaired cardiac performance and exercise capacity in HF patients.37,40 However, the uncertainty of efficacy, the need for continuous administration, and its side effects, unfortunately, make its practical application controversial.

4.6 Mitochondrial dysfunction

Mitochondrial dysfunction is a common feature of various aspects of ageing, and reduced mitochondrial energy metabolism is associated with the development of various age-related metabolic diseases, including HF.41,42 A key regulator of mitochondrial biogenesis is the peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α),194 which is modulated by various regulators including AMP-activated protein kinase (AMPK), mTOR, SIRT-1, Akt, and GSK3β.195 On the other hand, PGC-1α suppresses the transcription factor Foxo3 and prevents proteolysis by controlling UPS (Figure 2).196 Whilst PGC-1α levels in skeletal muscle decline dramatically with age,195 there is consensus that exercise increases PGC-1α expression in skeletal muscle through the AMPK-SIRT-1 signalling pathway among others.197,198 Moreover, in experimental models of HF, mitochondrial dysfunction in cardiac and skeletal muscle has been demonstrated to be associated with lower levels of PGC-1α expression.199 In light of these findings, PGC-1α may hold promise as a target for preventing the development of sarcopenia and HF through the regulation of mitochondrial biosynthesis.

ROS are generated primarily in mitochondria as a by-product of the electron transfer chain and increase as the organism ages, contributing to mitochondrial degradation and cellular senescence.43 Mitochondrial DNA (mtDNA) is located next to the ROS generator and is more transcriptionally active than nuclear DNA, rendering it more prone to damage than nuclear DNA and a major target for ROS.125 Myocytes are inherently susceptible to mitochondria-derived ROS injury because they account for a large proportion of total oxygen consumption in the organism.200 Furthermore, skeletal muscle is composed of post-mitotic cells, making it one of the tissues particularly susceptible to oxidative damage, along with the central nervous system.201 It is well known that ROS production is increased in dysfunctional mitochondria of aged muscle.126 The accumulation of oxidative stress with ageing alters excitation-contraction coupling and calcium homeostasis in skeletal muscle, causing fibre loss via apoptosis, atrophy of remaining fibres, dysfunction of SCs, and concomitant impairment of muscle degeneration or muscle regeneration, which may eventually lead to sarcopenia.202,203 Increased ROS production by dysfunctional mitochondria causes a mismatch between ATP production and demand, which can also lead to cardiac remodelling, inflammation, and diastolic dysfunction.122,127 Moreover, oxidative stress is enhanced in patients with HF, contributing to disease severity and exercise intolerance by causing intracellular Ca2+ overload in cardiac myocytes, hypertrophy, apoptosis, and endothelial dysfunction.44–46 However, against the so-called ‘mitochondrial free radical theory’ described above,43 evidence is also accumulating in the field of intracellular signal transduction regarding the role of ROS in triggering cell proliferation and survival signals in response to physiological signals and stress conditions.47–49 Recently, Viña et al.204 proposed a ‘free radical theory of frailty’ in which oxidative damage is associated with frailty rather than chronological age, especially in the elderly. Thus, elucidating the details of the role of mitochondrial dysfunction and ROS in ageing remains a significant challenge in ageing research.

4.7 Cellular senescence and stem cell exhaustion

Cellular senescence, defined as the stable arrest of the cell cycle and the concomitant change in canonical phenotype,205 is caused by telomere shortening,206 but several pathways have been implicated in the road to senescence independently of this process. Most prominent among them are the DNA damage response pathways, with the p16INK4a/Rb and p19ARF/p53 tumour suppressor pathways appearing to be crucial in age-related senescence.207 Both p16INK4a and p19ARF are encoded at the same genomic locus, the INK4a/ARF locus, which has been identified as the genomic locus most genetically associated with age-related conditions such as frailty, diabetes, and CAD.50,51,208 Actually, selective removal of INK4a-expressing senescent cells can delay the onset or even halt the progression of age-related diseases in mice.209 Senescent cells are usually eliminated by immune surveillance but accumulate with age.210 Whilst cyclin-dependent kinase (CKD) inhibitors such as p16 and p21, tumour suppressor proteins expressed in senescent cells prevent potential cancer cells from proliferating by arresting the cell cycle,211 senescent cells may also disrupt tissue integrity and make its repair difficult.212 As such, excessive senescent cell accumulation plays a crucial role in driving tissue senescence, which contributes to the development of several age-related pathologies such as frailty and sarcopenia.52 In addition, senescent cells form what is termed the senescence-associated secretory phenotype (SASP) and secrete pro-inflammatory cytokines, chemokines, matrix-remodelling proteases, TGF-β, and ROS, all of which may contribute to the development of frailty and sarcopenia.213,214 The SASP can also be induced locally in normal neighbouring cells via autocrine or paracrine mechanisms of senescent cells.215 A Recent study in mice has also revealed a ‘bystander effect’ in which transplantation of senescent cells adjacent to normal skeletal muscle increases senescence markers in the muscle and induces thinning of muscle fibres indicative of sarcopenia.216

The decline in the regenerative potential of tissues by stem cell exhaustion is one of the obvious features of ageing. While there is a study indicating that depletion of SCs impairs muscle regenerative capacity without affecting sarcopenia,53 a number of studies have reported that exercise therapy, the most effective treatment for sarcopenia and frailty, protects SCs against exhaustion and increases their number and activity.54,217,218 A better understanding of the role of senescent cells and SCs in sarcopenia and frailty will facilitate the realization of new therapeutic approaches to combat these pathologies.

4.8 Altered intercellular communication

The mechanisms discussed above address alterations at the cellular intrinsic level with ageing. However, ageing also leads to changes at the level of intercellular communication, including endocrine, neuroendocrine, and inflammatory signalling. Age-related chronic low-level inflammation is referred to as ‘inflamm-ageing’,219 and several pro-inflammatory cytokines like interleukin (IL)-1, IL-6, and TNF are known to be elevated during the ageing process.220 Meanwhile, systemic inflammation is related to factors closely associated with physical frailty, such as loss of muscle mass, muscle strength, and bone mineral density, as well as many age-related diseases.55,56,221 Elevated IL-1, IL-6, and TNF result in impaired protein synthesis from decreased IGF-1 and associated suppression of the PI3K/Akt pathway,222 and down-regulation of Foxo3 phosphorylation.223 They also enhance mechanisms involved in protein catabolism, such as the p38 MAPK pathway,224 the NF-κB pathway,140,141 the glucocorticoid pathway via activation of the hypothalamic-pituitary-adrenal (HPA) axis,225 and UPS via increased levels of E3 ubiquitin ligase,226 which in turn shifts skeletal muscle towards catabolism altogether (Figure 2). Furthermore, whilst there are some reports of no changes in any inflammatory cytokines in anorexic patients,139,227 central or peripheral administration of IL-1β or TNF induces anorexia, which has an essential role in the development of cachexia.228–230 Given the above findings, these pro-inflammatory cytokines have recently emerged as promising candidates for the screening and monitoring of sarcopenia and frailty. Meanwhile, patients with HF have chronic low-grade systemic inflammation, with elevated levels of the aforementioned inflammatory biomarkers involved in skeletal muscle wasting.57,142,143 Chronic inflammation promotes cardiac dysfunction/remodelling, further exacerbating HF itself.58 Therefore, inflammation may also be a shared pathogenesis contributing to the exacerbation of HF itself and the development of physical frailty in patients with HF.

As for the age-related neurohormonal changes, activation of the renin-angiotensin system (RAS) accelerates cellular senescence by inducing the uncontrolled generation of ROS, telomere shortening, and cell cycle arrest by angiotensin II (Ang II).231–233 Whilst an enhanced RAS is well known to be found in HF patients, there is emerging evidence that it can contribute to muscle wasting through increased production of ROS and induction of TGF-β1 and connective tissue growth factor expression.59 Recently, RAS inhibition was also demonstrated to be associated with a lower prevalence of muscle wasting in patients with HF.131

Growth differentiation factor-15 (GDF-15) is a stress-induced mitokine and not a hormone, but deserves mention here as a potential therapeutic target for frailty.131 This mitokine is a remote member of the TGF-β superfamily, mirrors inflammation and oxidative stress, and its elevated circulating levels have been identified in ageing, cancer, and cachexia, making it a powerful prognostic biomarker for these conditions.132,133 GDF-15 is also expressed in myocytes in response to myocardial ischaemia and pressure overload,134 and its elevated levels are associated with the severity and prognosis of acute coronary syndromes and chronic HF.135,136 Furthermore, Bettencourt et al.137 demonstrated that higher GDF-15 levels in acute HF were associated with worse prognosis independent of B-type natriuretic peptide (BNP) levels. Most recently, neutralization of GDF-15 with a rat model of monocrotaline-induced cardiac cachexia was reported to prevent anorexia and weight loss, indicating that inhibition of GDF-15 is a potential therapeutic strategy for cardiac cachexia.138

4.9 Environmental factors

Frailty is thought to be initiated by a combination of multiple biological mechanisms as mentioned above, yet it can also be induced and exacerbated by a variety of environmental factors from birth onward. Smoking is a predictor of worsening frailty status among community-dwelling elderly people,234 with possible mechanisms including reduced DNA methylation levels, one of the major forms of epigenetic modification; inflammation induced by various chemicals in cigarettes; and smoking-induced complications such as cardiovascular disease, lung disease, and cancer that ultimately contribute to the development of frailty.60,61 A good diet throughout the lifespan is critical for the maintenance of skeletal muscle, and poor dietary patterns have been implicated in the development of frailty through their effects on systemic inflammation, oxidative stress, and the composition of the gut microbiota.62,63 Physical inactivity and sedentary behaviour also cause oxidative damage and chronic inflammation, mitochondrial dysfunction, decreased autophagy, and impaired IGF-1/mTOR signalling pathway, which are implicated in worsening frailty levels.64,65 Exercise and nutritional therapy both improve the aforementioned drivers of frailty, respectively, but their combination would be more desirable.235 In the following section, we further provide recent promising treatments for frailty and sarcopenia, including exercise and nutritional therapy.

5. Treatment strategies for frailty and sarcopenia

Patients living with the clinical syndromes of frailty and sarcopenia endure an immense burden, with no particularly effective method available to slow their progression other than exercise training. Since disability is difficult to reverse in the elderly and is extremely devastating for individuals and societies, new strategies should be developed to preserve adequate levels of functional capacity and independence in late life and in chronic illness. The combination of exercise, nutrition, and pharmacological therapy may counteract the development and progression of sarcopenia and frailty.236 Here we describe the treatment strategies for sarcopenia and frailty in association with pathophysiological mechanisms (Figure 3).

Treatment strategies for frailty and sarcopenia. CAD, coronary artery disease; GDF-15, Growth differentiation factor-15; HF, heart failure.
Figure 3

Treatment strategies for frailty and sarcopenia. CAD, coronary artery disease; GDF-15, Growth differentiation factor-15; HF, heart failure.

5.1 Exercise

Physical exercise is now the most effective therapy for slowing the progress of sarcopenia and frailty. Precisely organized and supervised exercise training interventions aim at preventing muscle atrophy, promoting muscle growth, and ultimately, maintaining muscle functions during ageing. Beckwée et al.66 reviewed the currently available exercise interventions for the prevention and treatment of sarcopenia using the method of a systematic umbrella review and provided high-quality evidence for the beneficial and significant effect of resistance training on functional capacity, muscle mass, and muscle strength. Given the fact that sarcopenia is omnipresent and is affecting all skeletal muscles in the body with the most functional consequences in the limbs, the authors recommend training that activates the large muscle groups in a total body approach.66 Beneficial effects of resistance exercise training are explained through different physiological mechanisms and signalling pathways: vasodilation, antithrombotic effects, inhibition of oxidative stress, anti-inflammatory effects, mTORC1 activation, augmented mitochondrial biogenesis, elevated IGF-1/myostatin ratio, stimulation of peroxisomes, and enhanced insulin sensitivity.237,238 The molecular biological changes with exercise interventions confirm that the skeletal muscle phenotype is subject to considerable adaptation and plasticity depending on use. Low-intensity training improves mitochondrial function and increases oxygen consumption, whereas high-intensity exercise leads to muscle proliferation and augmentation of contractile proteins.239 Exercise training stimulates transcriptional up-regulation of the genes involved in Ca2+ signalling via the AMPK pathway and influences skeletal myocytes' energy status.239

With regard to HF, data from multiple exercise intervention trials have shown that supervised exercise training is safe and useful to improve exercise capacity and quality of life in patients with HF, although it failed to reduce mortality and hospitalization rates.67 In addition, albeit low-intensity resistance training (≤50% 1-repetition maximum) is sufficient to promote strength gains, high-intensity resistance training (i.e. 80% 1-repetition maximum) is recommended to obtain maximal strength gains, however, different approaches may be necessary for elderly geriatric patients and in patients with HF.

The Sarcopenia and Physical fRailty IN older people: multi-componenT Treatment strategies (SPRINTT) project conducted a randomized controlled trial (RCT) to test a multicomponent intervention (MCI) specifically designed to prevent mobility disability in high-risk older people.240 SPRINTT was a phase III, multicentre RCT aimed at comparing the efficacy of MCI, which encompasses long-term structured physical activity, nutritional counselling, and communication technology intervention vs. a standardized healthy ageing lifestyle education programme, designed to prevent adverse outcomes and disability in more than 1500 frail elderly subjects. The primary endpoint of the SPIRINTT trial was mobility disability, defined as the inability to walk 400 m in <15 min, without sitting, help of another person, or the use of a walker. Secondary endpoints included changes in muscle mass and strength, persistent mobility disability, injury and falls, disability in activities of daily living, nutritional status, cognitive function, mood, the use of healthcare resources, cost-effectiveness analyses, quality of life, and the mortality rate. Recently published results of the SPRINTT trial demonstrated that the multicomponent intervention is associated with a reduction in the incidence of mobility disability in frail sarcopenic elderly subjects and an SPPB score of 3–7.68 These results imply that physical frailty and sarcopenia can be targeted with multicomponent interventions in order to preserve mobility and functional capacity in vulnerable elderly subjects.68 Naturally, there are limitations frequently faced when treating subjects with frailty, as subjects very often do not accept or cannot follow exercise interventions. Another limitation would be that despite the overwhelming proof of the health benefits of regular exercise, there is limited knowledge regarding the proper prescription of exercise regimens as well as predictors of individual responses to exercise interventions in the population of frail elderly. This primarily emanates from broad interindividual differences in the biological responses to exercise.241

5.2 Nutrition

Malnutrition and anorexia are per se associated with adverse clinical outcomes. Malnutrition includes all forms of undernutrition (wasting, stunting, underweight), vitamin and mineral deficiencies, obesity, and resulting diet-related non-communicable diseases, whereas anorexia is common in elderly subjects and independently associated with sarcopenia.242,243 Energy expenditure is reduced as a protective mechanism in malnutrition whereas it remains inappropriately high in sarcopenia and muscle wasting.69 Also, increased resting energy expenditure (REE) has been shown in HF,70 where a negative balance between energy demand and consumption promotes catabolism and leads to malnutrition, whilst REE in HF patients with muscle wasting is reported to be lower than in those without muscle wasting.244 Dietary deficiencies in micro- (‘nutraceuticals’) and macronutrients (energy storage and substrates) contribute to the progression of a catabolic state in sarcopenia, frailty, and HF. Certainly, deficiency in micronutrients (vitamins and minerals, e.g. vitamin D or iron) impairs muscle physiology.245,246 Vitamin D deficiency is common in patients with HF and not only exacerbates the pathogenesis of HF by enhancing renin-angiotensin system activity, altering calcium influx, and causing inflammation and endothelial dysfunction but is also associated with reduced muscle mass and impaired physical performance in the elderly with or without HF.247 Thus, vitamin D supplementation could be an attractive strategy for the management of sarcopenia in the setting of HF.248

There is emerging evidence on the pathophysiology of skeletal muscle and nutritional interventions. Adequate nutrition improves muscle function, contributes to muscle anabolism, and regulates muscle protein synthesis, glucose and insulin homeostasis, and neuromuscular and microvascular function.249 Moreover, nutritional interventions regulate nutrient sensing, improve mitochondrial function, and cross-talk between muscle and the immune system.249 While both exercise and nutritional interventions serve as anabolic stimuli to skeletal myocytes, when these strategies are combined, additive effects on muscle mass and function are observed, especially using protein supplantation and resistance training.250

Dietary protein is one of the most important components for frail elderly and HF patients with regard to muscle structure and function as well as to anabolic–catabolic balance.71 Upon protein intake, essential amino acids (e.g. leucine) exert strong anabolic effects via activation of muscle intracellular signalling pathways that culminate in increased efficiency of mRNA translation and muscle protein synthesis.251 Therefore, high protein intake is generally recommended in elderly patients with sarcopenia and patients with HF, but special attention needs to consider chronic kidney disease, which frequently coexists with these conditions.72 Additionally, omega-3 polyunsaturated fatty acids could be an alternative anti-sarcopenic therapeutic agent, due to their anti-inflammatory properties.252 Furthermore, SPRINTT trial has shown that multicomponent intervention, based on physical activity and nutritional counselling has various beneficial effects in preserving mobility in older people.68

As such, nutritional interventions for patients with sarcopenia and frailty could prevent or ameliorate adverse events associated with these conditions but should be taken into account with the patient's current body weight and comorbidities. Despite being central in treating frailty, nutritional interventions have several important limitations. Common challenges and limitations of dietary interventions comprise the complex nature of food and nutrient interactions, lack of blinding of the treatments, low compliance, high susceptibility to confounding variables (e.g. ethnicity, genotype, and physiological state), diverse dietary behaviours as well as food culture.253

5.3 Drug therapy

The management of sarcopenia is primarily focused on physical exercise for muscle strengthening, supported by nutritional interventions. There are no pharmacologic agents approved worldwide for the treatment of sarcopenia. However, treating the underlying comorbidities, such as HF, cancer, chronic obstructive pulmonary disease, chronic kidney disease, or dementia may exert beneficial effects on sarcopenia via multiple biological pathways and cross-talks. Some of the potential pharmacological interventions that had been widely studied pre-clinically are hormonal treatment and myostatin inhibition. Hormonal treatment embraces testosterone application, GH, ghrelin, insulin, and thyroid hormones.236 Due to its well-known metabolic, anabolic, and peripheral effects, testosterone replacement therapy has been considered as a potential anti-sarcopenic therapy. Randomized controlled trials have demonstrated that testosterone therapy could moderately improve functional capacity and muscle function in sarcopenic HF patients.73 However, given the various androgen side effects, including the risk of prostatic hyperplasia, large-scale trials are needed to confirm the efficacy and safety of testosterone application. GH replacement therapy could pre-clinically ameliorate muscle wasting through anabolic, anti-inflammatory, and antioxidant effects.74 However, the clinical effectiveness of GH has not been confirmed yet. Due to its beneficial effects on appetite and gastric motility, ghrelin may be a promising therapeutic target for sarcopenia. Its effectiveness has been shown in animal models as well as in a clinical study, which demonstrated that ghrelin administration could improve functional capacity, muscle strength, and mass as well as left ventricular ejection fraction in sarcopenic HF patients.75 Insulin stimulates muscle protein synthesis by increasing amino acid delivery and intramuscular blood flow.76 However, a large-scale clinical study displayed that insulin therapy might be associated with worse outcomes in patients with chronic HF and diabetes.254 Thyroid hormone is an essential metabolic regulator, whose major target is skeletal myocytes. It has been shown that overt and latent thyroid dysfunction has been associated with muscle wasting and reduced muscle function.255 Over the past decade, myostatin has been a primary pharmacological target in sarcopenia. Myostatin is an extracellular cytokine and a member of the TGF-β family, and it is highly expressed in skeletal myocytes.256 The results of several trials with myostatin inhibitors were disappointing, and there was only limited therapeutical effectiveness. However, a study with bimagrumab in sarcopenic elderly subjects has shown beneficial effects and improved functional capacity and independence.77

Currently, a new star in the field is certainly GDF-15, which is important in muscle pathophysiology and has been described as a global regulator of stress. Contemporary evidence suggests that GDF-15 is associated with decreased muscle mass and performance and increased inflammation.257 Therefore, GDF-15 neutralization is a transformative therapeutic approach with effects on restoring muscle function and physical performance. Anti-GDF-15 treatment in experimental models increased muscle mass by increasing appetite and food intake and markedly improved muscle function and physical performance.78 Clinical studies with industrial engagement and knowledge transfer in the field presented recently a novel drug BIO101, a small molecule and a leading drug candidate, administered orally, and its effectiveness in treating sarcopenia.79 This candidate molecule showed promising results for the improvement of mobility disability in the gait speed from the 400 m walking test. The primary endpoint was the ability to complete a 400 m walk test in less than 15 min, and the secondary endpoint: gait speed of 4 m from SPPB, hand grip strength test, and patient-reported outcome.

6. Conclusions

With the ageing of the population, the prevalence of frailty and sarcopenia, causing a variety of adverse outcomes, is on the rise. The biological mechanisms of these geriatric syndromes are multifactorial and overlap considerably with the pathophysiology of cardiac disease, particularly HF, and their frequent concomitant and complex interactions clearly deteriorate prognosis. Whilst exercise training, nutritional therapies, and disease-specific therapies are certainly the primary treatments for such a complex condition, further research into therapies devised from pathophysiologies, such as drugs that enhance the regenerative capacity of SCs and regulate autophagy, anabolic hormone replacement therapy, myostatin inhibitors, and anti-GDF-15 therapy, is warranted to improve prognosis.

Data availability

No new data were generated or analysed in support of this research.

References

1

Fried
LP
,
Tangen
CM
,
Walston
J
,
Newman
AB
,
Hirsch
C
,
Gottdiener
J
,
Seeman
T
,
Tracy
R
,
Kop
WJ
,
Burke
G
,
McBurnie
MA
;
Cardiovascular Health Study Collaborative Research Group
.
Frailty in older adults: evidence for a phenotype
.
J Gerontol A Biol Sci Med Sci
2001
;
56
:
M146
M156
.

2

He
W
,
Goodkind
D
,
Kowal
PR
.
An Aging World: 2015
.
Washington, DC
:
United States Census Bureau
;
2016
.

3

O’Caoimh
R
,
Sezgin
D
,
O’Donovan
MR
,
Molloy
DW
,
Clegg
A
,
Rockwood
K
,
Liew
A
.
Prevalence of frailty in 62 countries across the world: a systematic review and meta-analysis of population-level studies
.
Age Ageing
2021
;
50
:
96
104
.

4

Ryall
JG
,
Schertzer
JD
,
Lynch
GS
.
Cellular and molecular mechanisms underlying age-related skeletal muscle wasting and weakness
.
Biogerontology
2008
;
9
:
213
228
.

5

Bauer
J
,
Morley
JE
,
Schols
AMWJ
,
Ferrucci
L
,
Cruz-Jentoft
AJ
,
Dent
E
,
Baracos
VE
,
Crawford
JA
,
Doehner
W
,
Heymsfield
SB
,
Jatoi
A
,
Kalantar-Zadeh
K
,
Lainscak
M
,
Landi
F
,
Laviano
A
,
Mancuso
M
,
Muscaritoli
M
,
Prado
CM
,
Strasser
F
,
von Haehling
S
,
Coats
AJS
,
Anker
SD
.
Sarcopenia: a time for action. An SCWD position paper
.
J Cachexia Sarcopenia Muscle
2019
;
10
:
956
961
.

6

Matsue
Y
,
Kamiya
K
,
Saito
H
,
Saito
K
,
Ogasahara
Y
,
Maekawa
E
,
Konishi
M
,
Kitai
T
,
Iwata
K
,
Jujo
K
,
Wada
H
,
Kasai
T
,
Nagamatsu
H
,
Ozawa
T
,
Izawa
K
,
Yamamoto
S
,
Aizawa
N
,
Yonezawa
R
,
Oka
K
,
Momomura
SI
,
Kagiyama
N
.
Prevalence and prognostic impact of the coexistence of multiple frailty domains in elderly patients with heart failure: the FRAGILE-HF cohort study
.
Eur J Heart Fail
2020
;
22
:
2112
2119
.

7

Bielecka-Dabrowa
A
,
Ebner
N
,
Dos Santos
MR
,
Ishida
J
,
Hasenfuss
G
,
von Haehling
S
.
Cachexia, muscle wasting, and frailty in cardiovascular disease
.
Eur J Heart Fail
2020
;
22
:
2314
2326
.

8

Casas-Herrero
Á
,
Sáez de Asteasu
ML
,
Antón-Rodrigo
I
,
Sánchez-Sánchez
JL
,
Montero-Odasso
M
,
Marín-Epelde
I
,
Ramón-Espinoza
F
,
Zambom-Ferraresi
F
,
Petidier-Torregrosa
R
,
Elexpuru-Estomba
J
,
Álvarez-Bustos
A
,
Galbete
A
,
Martínez-Velilla
N
,
Izquierdo
M
.
Effects of Vivifrail multicomponent intervention on functional capacity: a multicentre, randomized controlled trial
.
J Cachexia Sarcopenia Muscle
2022
;
13
:
884
893
.

9

Vitale
C
,
Jankowska
E
,
Hill
L
,
Piepoli
M
,
Doehner
W
,
Anker
SD
,
Lainscak
M
,
Jaarsma
T
,
Ponikowski
P
,
Rosano
GMC
,
Seferovic
P
,
Coats
AJ
.
Heart Failure Association/European Society of Cardiology position paper on frailty in patients with heart failure
.
Eur J Heart Fail
2019
;
21
:
1299
1305
.

10

Yamada
S
,
Adachi
T
,
Izawa
H
,
Murohara
T
,
Kondo
T
.
Prognostic score based on physical frailty in patients with heart failure: a multicenter prospective cohort study (FLAGSHIP)
.
J Cachexia Sarcopenia Muscle
2021
;
12
:
1995
2006
.

11

Rolfson
DB
,
Majumdar
SR
,
Tsuyuki
RT
,
Tahir
A
,
Rockwood
K
.
Validity and reliability of the Edmonton Frail Scale
.
Age Ageing
2006
;
35
:
526
529
.

12

Rockwood
K
,
Song
X
,
MacKnight
C
,
Bergman
H
,
Hogan
DB
,
McDowell
I
,
Mitnitski
A
.
A global clinical measure of fitness and frailty in elderly people
.
CMAJ
2005
;
173
:
489
495
.

13

Damluji
AA
,
Chung
SE
,
Xue
QL
,
Hasan
RK
,
Moscucci
M
,
Forman
DE
,
Bandeen-Roche
K
,
Batchelor
W
,
Walston
JD
,
Resar
JR
,
Gerstenblith
G
.
Frailty and cardiovascular outcomes in the National Health and Aging Trends Study
.
Eur Heart J
2021
;
42
:
3856
3865
.

14

Khan
H
,
Kalogeropoulos
AP
,
Georgiopoulou
VV
,
Newman
AB
,
Harris
TB
,
Rodondi
N
,
Bauer
DC
,
Kritchevsky
SB
,
Butler
J
.
Frailty and risk for heart failure in older adults: the health, aging, and body composition study
.
Am Heart J
2013
;
166
:
887
894
.

15

Jun
JE
,
Choi
MS
,
Park
SW
,
Kim
G
,
Jin
S-M
,
Kim
K
,
Hwang
Y-C
,
Ahn
KJ
,
Chung
HY
,
Jeong
I-K
,
Kang
M
,
Kim
JH
.
Low skeletal muscle mass is associated with the presence, incidence, and progression of coronary artery calcification
.
Can J Cardiol
2021
;
37
:
1480
1488
.

16

Ko
B-J
,
Chang
Y
,
Jung
H-S
,
Yun
KE
,
Kim
C-W
,
Park
HS
,
Chung
EC
,
Shin
H
,
Ryu
S
.
Relationship between low relative muscle mass and coronary artery calcification in healthy adults
.
Arterioscler Thromb Vasc Biol
2016
;
36
:
1016
1021
.

17

Radak
Z
,
Bori
Z
,
Koltai
E
,
Fatouros
IG
,
Jamurtas
AZ
,
Douroudos
II
,
Terzis
G
,
Nikolaidis
MG
,
Chatzinikolaou
A
,
Sovatzidis
A
,
Kumagai
S
,
Naito
H
,
Boldogh
I
.
Age-dependent changes in 8-oxoguanine-DNA glycosylase activity are modulated by adaptive responses to physical exercise in human skeletal muscle
.
Free Radic Biol Med
2011
;
51
:
417
423
.

18

Sánchez-Flores
M
,
Marcos-Pérez
D
,
Lorenzo-López
L
,
Maseda
A
,
Millán-Calenti
JC
,
Bonassi
S
,
Pásaro
E
,
Laffon
B
,
Valdiglesias
V
.
Frailty syndrome and genomic instability in older adults: suitability of the cytome micronucleus assay as a diagnostic tool
.
J Gerontol A Biol Sci Med Sci
2018
;
73
:
864
872
.

19

van der Harst
P
,
van der Steege
G
,
de Boer
RA
,
Voors
AA
,
Hall
AS
,
Mulder
MJ
,
van Gilst
WH
,
van Veldhuisen
DJ
;
MERIT-HF Study Group
.
Telomere length of circulating leukocytes is decreased in patients with chronic heart failure
.
J Am Coll Cardiol
2007
;
49
:
1459
1464
.

20

Lorenzi
M
,
Bonassi
S
,
Lorenzi
T
,
Giovannini
S
,
Bernabei
R
,
Onder
G
.
A review of telomere length in sarcopenia and frailty
.
Biogerontology
2018
;
19
:
209
221
.

21

Bernabeu-Wittel
M
,
Gómez-Díaz
R
,
González-Molina
Á
,
Vidal-Serrano
S
,
Díez-Manglano
J
,
Salgado
F
,
Soto-Martín
M
,
Ollero-Baturone
M
;
On Behalf Of The Proteo Researchers
.
Oxidative stress, telomere shortening, and apoptosis associated to sarcopenia and frailty in patients with multimorbidity
.
J Clin Med
2020
;
9
:
2669
.

22

Bountziouka
V
,
Nelson
CP
,
Codd
V
,
Wang
Q
,
Musicha
C
,
Allara
E
,
Kaptoge
S
,
Di Angelantonio
E
,
Butterworth
AS
,
Thompson
JR
,
Curtis
EM
,
Wood
AM
,
Danesh
JN
,
Harvey
NC
,
Cooper
C
,
Samani
NJ
.
Association of shorter leucocyte telomere length with risk of frailty
.
J Cachexia Sarcopenia Muscle
2022
;
13
:
1741
1751
.

23

Gensous
N
,
Bacalini
MG
,
Franceschi
C
,
Meskers
CGM
,
Maier
AB
,
Garagnani
P
.
Age-related DNA methylation changes: potential impact on skeletal muscle aging in humans
.
Front Physiol
2019
;
10
:
996
.

24

He
L
,
Khanal
P
,
Morse
CI
,
Williams
A
,
Thomis
M
.
Differentially methylated gene patterns between age-matched sarcopenic and non-sarcopenic women
.
J Cachexia Sarcopenia Muscle
2019
;
10
:
1295
1306
.

25

Gehrig
SM
,
van der Poel
C
,
Sayer
TA
,
Schertzer
JD
,
Henstridge
DC
,
Church
JE
,
Lamon
S
,
Russell
AP
,
Davies
KE
,
Febbraio
MA
,
Lynch
GS
.
Hsp72 preserves muscle function and slows progression of severe muscular dystrophy
.
Nature
2012
;
484
:
394
398
.

26

Milne
KJ
,
Noble
EG
.
Exercise-induced elevation of HSP70 is intensity dependent
.
J Appl Physiol (Bethesda, Md: 1985)
2002
;
93
:
561
568
.

27

Mangner
N
,
Weikert
B
,
Bowen
TS
,
Sandri
M
,
Höllriegel
R
,
Erbs
S
,
Hambrecht
R
,
Schuler
G
,
Linke
A
,
Gielen
S
,
Adams
V
.
Skeletal muscle alterations in chronic heart failure: differential effects on quadriceps and diaphragm
.
J Cachexia Sarcopenia Muscle
2015
;
6
:
381
390
.

28

Adams
V
,
Linke
A
,
Wisloff
U
,
Döring
C
,
Erbs
S
,
Kränkel
N
,
Witt
CC
,
Labeit
S
,
Müller-Werdan
U
,
Schuler
G
,
Hambrecht
R
.
Myocardial expression of Murf-1 and MAFbx after induction of chronic heart failure: effect on myocardial contractility
.
Cardiovasc Res
2007
;
73
:
120
129
.

29

Whitman
SA
,
Wacker
MJ
,
Richmond
SR
,
Godard
MP
.
Contributions of the ubiquitin-proteasome pathway and apoptosis to human skeletal muscle wasting with age
.
Pflugers Archiv
2005
;
450
:
437
446
.

30

Edström
E
,
Altun
M
,
Hägglund
M
,
Ulfhake
B
.
Atrogin-1/MAFbx and MuRF1 are downregulated in aging-related loss of skeletal muscle
.
J Gerontol A Biol Sci Med Sci
2006
;
61
:
663
674
.

31

Clavel
S
,
Coldefy
AS
,
Kurkdjian
E
,
Salles
J
,
Margaritis
I
,
Derijard
B
.
Atrophy-related ubiquitin ligases, atrogin-1 and MuRF1 are up-regulated in aged rat tibialis anterior muscle
.
Mech Ageing Dev
2006
;
127
:
794
801
.

32

Drummond
MJ
,
Addison
O
,
Brunker
L
,
Hopkins
PN
,
McClain
DA
,
LaStayo
PC
,
Marcus
RL
.
Downregulation of E3 ubiquitin ligases and mitophagy-related genes in skeletal muscle of physically inactive, frail older women: a cross-sectional comparison
.
J Gerontol A Biol Sci Med Sci
2014
;
69
:
1040
1048
.

33

Potes
Y
,
de Luxán-Delgado
B
,
Rodriguez-González
S
,
Guimarães
MRM
,
Solano
JJ
,
Fernández-Fernández
M
,
Bermúdez
M
,
Boga
JA
,
Vega-Naredo
I
,
Coto-Montes
A
.
Overweight in elderly people induces impaired autophagy in skeletal muscle
.
Free Radic Biol Med
2017
;
110
:
31
41
.

34

Aucello
M
,
Dobrowolny
G
,
Musarò
A
.
Localized accumulation of oxidative stress causes muscle atrophy through activation of an autophagic pathway
.
Autophagy
2009
;
5
:
527
529
.

35

Nakai
A
,
Yamaguchi
O
,
Takeda
T
,
Higuchi
Y
,
Hikoso
S
,
Taniike
M
,
Omiya
S
,
Mizote
I
,
Matsumura
Y
,
Asahi
M
,
Nishida
K
,
Hori
M
,
Mizushima
N
,
Otsu
K
.
The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress
.
Nat Med
2007
;
13
:
619
624
.

36

Kanamori
H
,
Takemura
G
,
Maruyama
R
,
Goto
K
,
Tsujimoto
A
,
Ogino
A
,
Li
L
,
Kawamura
I
,
Takeyama
T
,
Kawaguchi
T
,
Nagashima
K
,
Fujiwara
T
,
Fujiwara
H
,
Seishima
M
,
Minatoguchi
S
.
Functional significance and morphological characterization of starvation-induced autophagy in the adult heart
.
Am J Pathol
2009
;
174
:
1705
1714
.

37

Kontoleon
PE
,
Anastasiou-Nana
MI
,
Papapetrou
PD
,
Alexopoulos
G
,
Ktenas
V
,
Rapti
AC
,
Tsagalou
EP
,
Nanas
JN
.
Hormonal profile in patients with congestive heart failure
.
Int J Cardiol
2003
;
87
:
179
183
.

38

Bian
A
,
Ma
Y
,
Zhou
X
,
Guo
Y
,
Wang
W
,
Zhang
Y
,
Wang
X
.
Association between sarcopenia and levels of growth hormone and insulin-like growth factor-1 in the elderly
.
BMC Musculoskelet Disord
2020
;
21
:
214
.

39

Cappola
AR
,
Xue
Q-L
,
Fried
LP
.
Multiple hormonal deficiencies in anabolic hormones are found in frail older women: the Women’s Health and Aging studies
.
J Gerontol A Biol Sci Med Sci
2009
;
64
:
243
248
.

40

Gangitano
E
,
Barbaro
G
,
Susi
M
,
Rossetti
R
,
Spoltore
ME
,
Masi
D
,
Tozzi
R
,
Mariani
S
,
Gnessi
L
,
Lubrano
C
.
Growth hormone secretory capacity is associated with cardiac morphology and function in overweight and obese patients: a controlled, cross-sectional study
.
Cells
2022
;
11
:
2420
.

41

Amorim
JA
,
Coppotelli
G
,
Rolo
AP
,
Palmeira
CM
,
Ross
JM
,
Sinclair
DA
.
Mitochondrial and metabolic dysfunction in ageing and age-related diseases
.
Nat Rev Endocrinol
2022
;
18
:
243
258
.

42

Rosca
MG
,
Vazquez
EJ
,
Kerner
J
,
Parland
W
,
Chandler
MP
,
Stanley
W
,
Sabbah
HN
,
Hoppel
CL
.
Cardiac mitochondria in heart failure: decrease in respirasomes and oxidative phosphorylation
.
Cardiovasc Res
2008
;
80
:
30
39
.

43

Harman
D
.
Aging: a theory based on free radical and radiation chemistry
.
J Gerontol
1956
;
11
:
298
300
.

44

Keith
M
,
Geranmayegan
A
,
Sole
MJ
,
Kurian
R
,
Robinson
A
,
Omran
AS
,
Jeejeebhoy
KN
.
Increased oxidative stress in patients with congestive heart failure
.
J Am Coll Cardiol
1998
;
31
:
1352
1356
.

45

Klimiuk
A
,
Zalewska
A
,
Sawicki
R
,
Knapp
M
,
Maciejczyk
M
.
Salivary oxidative stress increases with the progression of chronic heart failure
.
J Clin Med
2020
;
9
:
769
.

46

Shirakawa
R
,
Yokota
T
,
Nakajima
T
,
Takada
S
,
Yamane
M
,
Furihata
T
,
Maekawa
S
,
Nambu
H
,
Katayama
T
,
Fukushima
A
,
Saito
A
,
Ishimori
N
,
Dela
F
,
Kinugawa
S
,
Anzai
T
.
Mitochondrial reactive oxygen species generation in blood cells is associated with disease severity and exercise intolerance in heart failure patients
.
Sci Rep
2019
;
9
:
14709
.

47

Al-Mehdi
A-B
,
Pastukh
VM
,
Swiger
BM
,
Reed
DJ
,
Patel
MR
,
Bardwell
GC
,
Pastukh
VV
,
Alexeyev
MF
,
Gillespie
MN
.
Perinuclear mitochondrial clustering creates an oxidant-rich nuclear domain required for hypoxia-induced transcription
.
Sci Signal
2012
;
5
:
ra47
.

48

Schulz
TJ
,
Zarse
K
,
Voigt
A
,
Urban
N
,
Birringer
M
,
Ristow
M
.
Glucose restriction extends Caenorhabditis elegans life span by inducing mitochondrial respiration and increasing oxidative stress
.
Cell Metab
2007
;
6
:
280
293
.

49

Zhang
J
,
Khvorostov
I
,
Hong
JS
,
Oktay
Y
,
Vergnes
L
,
Nuebel
E
,
Wahjudi
PN
,
Setoguchi
K
,
Wang
G
,
Do
A
,
Jung
H-J
,
McCaffery
JM
,
Kurland
IJ
,
Reue
K
,
Lee
WN
,
Koehler
CM
,
Teitell
MA
.
UCP2 regulates energy metabolism and differentiation potential of human pluripotent stem cells
.
EMBO J
2011
;
30
:
4860
4873
.

50

Melzer
D
,
Frayling
TM
,
Murray
A
,
Hurst
AJ
,
Harries
LW
,
Song
H
,
Khaw
K
,
Luben
R
,
Surtees
PG
,
Bandinelli
SS
,
Corsi
A-M
,
Ferrucci
L
,
Guralnik
JM
,
Wallace
RB
,
Hattersley
AT
,
Pharoah
PD
.
A common variant of the p16(INK4a) genetic region is associated with physical function in older people
.
Mech Ageing Dev
2007
;
128
:
370
377
.

51

McPherson
R
,
Pertsemlidis
A
,
Kavaslar
N
,
Stewart
A
,
Roberts
R
,
Cox
DR
,
Hinds
DA
,
Pennacchio
LA
,
Tybjaerg-Hansen
A
,
Folsom
AR
,
Boerwinkle
E
,
Hobbs
HH
,
Cohen
JC
.
A common allele on chromosome 9 associated with coronary heart disease
.
Science (New York, NY)
2007
;
316
:
1488
1491
.

52

Song
P
,
An
J
,
Zou
M-H
.
Immune clearance of senescent cells to combat ageing and chronic diseases
.
Cells
2020
;
9
:
671
.

53

Fry
CS
,
Lee
JD
,
Mula
J
,
Kirby
TJ
,
Jackson
JR
,
Liu
F
,
Yang
L
,
Mendias
CL
,
Dupont-Versteegden
EE
,
McCarthy
JJ
,
Peterson
CA
.
Inducible depletion of satellite cells in adult, sedentary mice impairs muscle regenerative capacity without affecting sarcopenia
.
Nat Med
2015
;
21
:
76
80
.

54

Chen
Z
,
Li
L
,
Wu
W
,
Liu
Z
,
Huang
Y
,
Yang
L
,
Luo
Q
,
Chen
J
,
Hou
Y
,
Song
G
.
Exercise protects proliferative muscle satellite cells against exhaustion via the Igfbp7-Akt-mTOR axis
.
Theranostics
2020
;
10
:
6448
6466
.

55

Ding
C
,
Parameswaran
V
,
Udayan
R
,
Burgess
J
,
Jones
G
.
Circulating levels of inflammatory markers predict change in bone mineral density and resorption in older adults: a longitudinal study
.
J Clin Endocrinol Metab
2008
;
93
:
1952
1958
.

56

Visser
M
,
Pahor
M
,
Taaffe
DR
,
Goodpaster
BH
,
Simonsick
EM
,
Newman
AB
,
Nevitt
M
,
Harris
TB
.
Relationship of interleukin-6 and tumor necrosis factor-alpha with muscle mass and muscle strength in elderly men and women: the Health ABC study
.
J Gerontol A Biol Sci Med Sci
2002
;
57
:
M326
M332
.

57

Markousis-Mavrogenis
G
,
Tromp
J
,
Ouwerkerk
W
,
Devalaraja
M
,
Anker
SD
,
Cleland
JG
,
Dickstein
K
,
Filippatos
GS
,
van der Harst
P
,
Lang
CC
,
Metra
M
,
Ng
LL
,
Ponikowski
P
,
Samani
NJ
,
Zannad
F
,
Zwinderman
AH
,
Hillege
HL
,
van Veldhuisen
DJ
,
Kakkar
R
,
Voors
AA
,
van der Meer
P
.
The clinical significance of interleukin-6 in heart failure: results from the BIOSTAT-CHF study
.
Eur J Heart Fail
2019
;
21
:
965
973
.

58

Shirazi
LF
,
Bissett
J
,
Romeo
F
,
Mehta
JL
.
Role of inflammation in heart failure
.
Curr Atheroscler Rep
2017
;
19
:
27
.

59

Morales
MG
,
Vazquez
Y
,
Acuña
MJ
,
Rivera
JC
,
Simon
F
,
Salas
JD
,
Alvarez Ruf
J
,
Brandan
E
,
Cabello-Verrugio
C
.
Angiotensin II-induced pro-fibrotic effects require p38MAPK activity and transforming growth factor beta 1 expression in skeletal muscle cells
.
Int J Biochem Cell Biol
2012
;
44
:
1993
2002
.

60

Gao
X
,
Zhang
Y
,
Saum
KU
,
Schöttker
B
,
Breitling
LP
,
Brenner
H
.
Tobacco smoking and smoking-related DNA methylation are associated with the development of frailty among older adults
.
Epigenetics
2017
;
12
:
149
156
.

61

Gonçalves
RB
,
Coletta
RD
,
Silvério
KG
,
Benevides
L
,
Casati
MZ
,
da Silva
JS
,
Nociti
FH
Jr
.
Impact of smoking on inflammation: overview of molecular mechanisms
.
Inflamm Res
2011
;
60
:
409
424
.

62

Ni Lochlainn
M
,
Cox
NJ
,
Wilson
T
,
Hayhoe
RPG
,
Ramsay
SE
,
Granic
A
,
Isanejad
M
,
Roberts
HC
,
Wilson
D
,
Welch
C
,
Hurst
C
,
Atkins
JL
,
Mendonça
N
,
Horner
K
,
Tuttiett
ER
,
Morgan
Y
,
Heslop
P
,
Williams
EA
,
Steves
CJ
,
Greig
C
,
Draper
J
,
Corish
CA
,
Welch
A
,
Witham
MD
,
Sayer
AA
,
Robinson
S
.
Nutrition and frailty: opportunities for prevention and treatment
.
Nutrients
2021
;
13
:
2349
.

63

Giron
M
,
Thomas
M
,
Dardevet
D
,
Chassard
C
,
Savary-Auzeloux
I
.
Gut microbes and muscle function: can probiotics make our muscles stronger?
J Cachexia Sarcopenia Muscle
2022
;
13
:
1460
1476
.

64

Angulo
J
,
El Assar
M
,
Álvarez-Bustos
A
,
Rodríguez-Mañas
L
.
Physical activity and exercise: strategies to manage frailty
.
Redox Biol
2020
;
35
:
101513
.

65

Kehler
DS
,
Theou
O
.
The impact of physical activity and sedentary behaviors on frailty levels
.
Mech Ageing Dev
2019
;
180
:
29
41
.

66

Beckwée
D
,
Delaere
A
,
Aelbrecht
S
,
Baert
V
,
Beaudart
C
,
Bruyere
O
,
de Saint-Hubert
M
,
Bautmans
I
.
Exercise interventions for the prevention and treatment of sarcopenia. A systematic umbrella review
.
J Nutr Health Aging
2019
;
23
:
494
502
.

67

von Haehling
S
,
Arzt
M
,
Doehner
W
,
Edelmann
F
,
Evertz
R
,
Ebner
N
,
Herrmann-Lingen
C
,
Garfias Macedo
T
,
Koziolek
M
,
Noutsias
M
,
Schulze
PC
,
Wachter
R
,
Hasenfuß
G
,
Laufs
U
.
Improving exercise capacity and quality of life using non-invasive heart failure treatments: evidence from clinical trials
.
Eur J Heart Fail
2021
;
23
:
92
113
.

68

Bernabei
R
,
Landi
F
,
Calvani
R
,
Cesari
M
,
Del Signore
S
,
Anker
SD
,
Bejuit
R
,
Bordes
P
,
Cherubini
A
,
Cruz-Jentoft
AJ
,
Di Bari
M
,
Friede
T
,
Gorostiaga Ayestarán
C
,
Goyeau
H
,
Jónsson
PV
,
Kashiwa
M
,
Lattanzio
F
,
Maggio
M
,
Mariotti
L
,
Miller
RR
,
Rodriguez-Mañas
L
,
Roller-Wirnsberger
R
,
Rýznarová
I
,
Scholpp
J
,
Schols
A
,
Sieber
CC
,
Sinclair
AJ
,
Skalska
A
,
Strandberg
T
,
Tchalla
A
,
Topinková
E
,
Tosato
M
,
Vellas
B
,
von Haehling
S
,
Pahor
M
,
Roubenoff
R
,
Marzetti
E
;
SPRINTT Consortium
.
Multicomponent intervention to prevent mobility disability in frail older adults: randomised controlled trial (SPRINTT project)
.
BMJ
2022
;
377
:
e068788
.

69

Wu
J
,
Huang
C
,
Xiao
H
,
Tang
Q
,
Cai
W
.
Weight loss and resting energy expenditure in male patients with newly diagnosed esophageal cancer
.
Nutrition (Burbank, Los Angeles County, Calif)
2013
;
29
:
1310
1314
.

70

Sandek
A
,
Doehner
W
,
Anker
SD
,
von Haehling
S
.
Nutrition in heart failure: an update
.
Curr Opin Clin Nutr Metab Care
2009
;
12
:
384
391
.

71

Beasley
JM
,
Shikany
JM
,
Thomson
CA
.
The role of dietary protein intake in the prevention of sarcopenia of aging
.
Nutr Clin Pract
2013
;
28
:
684
690
.

72

Rhee
CM
,
Ahmadi
SF
,
Kovesdy
CP
,
Kalantar-Zadeh
K
.
Low-protein diet for conservative management of chronic kidney disease: a systematic review and meta-analysis of controlled trials
.
J Cachexia Sarcopenia Muscle
2018
;
9
:
235
245
.

73

Caminiti
G
,
Volterrani
M
,
Iellamo
F
,
Marazzi
G
,
Massaro
R
,
Miceli
M
,
Mammi
C
,
Piepoli
M
,
Fini
M
,
Rosano
GM
.
Effect of long-acting testosterone treatment on functional exercise capacity, skeletal muscle performance, insulin resistance, and baroreflex sensitivity in elderly patients with chronic heart failure a double-blind, placebo-controlled, randomized study
.
J Am Coll Cardiol
2009
;
54
:
919
927
.

74

Brioche
T
,
Kireev
RA
,
Cuesta
S
,
Gratas-Delamarche
A
,
Tresguerres
JA
,
Gomez-Cabrera
MC
,
Viña
J
.
Growth hormone replacement therapy prevents sarcopenia by a dual mechanism: improvement of protein balance and of antioxidant defenses
.
J Gerontol A Biol Sci Med Sci
2014
;
69
:
1186
1198
.

75

Nagaya
N
,
Moriya
J
,
Yasumura
Y
,
Uematsu
M
,
Ono
F
,
Shimizu
W
,
Ueno
K
,
Kitakaze
M
,
Miyatake
K
,
Kangawa
K
.
Effects of ghrelin administration on left ventricular function, exercise capacity, and muscle wasting in patients with chronic heart failure
.
Circulation
2004
;
110
:
3674
3679
.

76

Fujita
S
,
Rasmussen
BB
,
Cadenas
JG
,
Grady
JJ
,
Volpi
E
.
Effect of insulin on human skeletal muscle protein synthesis is modulated by insulin-induced changes in muscle blood flow and amino acid availability
.
Am J Physiol Endocrinol Metab
2006
;
291
:
E745
E754
.

77

Rooks
D
,
Praestgaard
J
,
Hariry
S
,
Laurent
D
,
Petricoul
O
,
Perry
RG
,
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
.

78

Wu
Z
,
Bennett
D
,
Brosnan
J
,
Calle
RA
,
Collins
S
,
Esquejo
R
,
Joaquim
S
,
Joyce
A
,
Kim
H
,
LaCarubba
B
,
Lin
L
,
Kim-Muller
JY
,
Peloquin
M
,
Pettersen
B
,
Qiao
S
,
Rossulek
M
,
Weber
G
,
Zhang
B
,
Birnbaum
M
,
Breen
D
.
2O growth differentiation factor 15 (GDF-15) neutralization reverses cancer cachexia, restores physical performance and mitigates emesis associated with platinum-based chemotherapy
.
Ann Oncol
2020
;
31
:
S245
.

79

Ebner
N
,
Anker
SD
,
von Haehling
S
.
Recent developments in the field of cachexia, sarcopenia, and muscle wasting: highlights from the 11th Cachexia Conference
.
J Cachexia Sarcopenia Muscle
2019
;
10
:
218
225
.

80

Sutton
JL
,
Gould
RL
,
Daley
S
,
Coulson
MC
,
Ward
EV
,
Butler
AM
,
Nunn
SP
,
Howard
RJ
.
Psychometric properties of multicomponent tools designed to assess frailty in older adults: a systematic review
.
BMC Geriatr
2016
;
16
:
55
.

81

Li
M
,
Harris
I
,
Lu
ZK
.
Differences in proxy-reported and patient-reported outcomes: assessing health and functional status among medicare beneficiaries
.
BMC Med Res Methodol
2015
;
15
:
62
.

82

Eeles
EMP
,
White
SV
,
O’Mahony
SM
,
Bayer
AJ
,
Hubbard
RE
.
The impact of frailty and delirium on mortality in older inpatients
.
Age Ageing
2012
;
41
:
412
416
.

83

Bagshaw
SM
,
Stelfox
HT
,
Johnson
JA
,
McDermid
RC
,
Rolfson
DB
,
Tsuyuki
RT
,
Ibrahim
Q
,
Majumdar
SR
.
Long-term association between frailty and health-related quality of life among survivors of critical illness: a prospective multicenter cohort study
.
Crit Care Med
2015
;
43
:
973
982
.

84

Cruz-Jentoft
AJ
,
Bahat
G
,
Bauer
J
,
Boirie
Y
,
Bruyère
O
,
Cederholm
T
,
Cooper
C
,
Landi
F
,
Rolland
Y
,
Sayer
AA
,
Schneider
SM
,
Sieber
CC
,
Topinkova
E
,
Vandewoude
M
,
Visser
M
,
Zamboni
M
;
Writing Group for the European Working Group on Sarcopenia in Older People 2 (EWGSOP2), and the Extended Group for EWGSOP2
.
Sarcopenia: revised European consensus on definition and diagnosis
.
Age Ageing
2019
;
48
:
16
31
.

85

Wang
DXM
,
Yao
J
,
Zirek
Y
,
Reijnierse
EM
,
Maier
AB
.
Muscle mass, strength, and physical performance predicting activities of daily living: a meta-analysis
.
J Cachexia Sarcopenia Muscle
2020
;
11
:
3
25
.

86

Kitamura
A
,
Seino
S
,
Abe
T
,
Nofuji
Y
,
Yokoyama
Y
,
Amano
H
,
Nishi
M
,
Taniguchi
Y
,
Narita
M
,
Fujiwara
Y
,
Shinkai
S
.
Sarcopenia: prevalence, associated factors, and the risk of mortality and disability in Japanese older adults
.
J Cachexia Sarcopenia Muscle
2021
;
12
:
30
38
.

87

Fülster
S
,
Tacke
M
,
Sandek
A
,
Ebner
N
,
Tschöpe
C
,
Doehner
W
,
Anker
SD
,
von Haehling
S
.
Muscle wasting in patients with chronic heart failure: results from the studies investigating co-morbidities aggravating heart failure (SICA-HF)
.
Eur Heart J
2013
;
34
:
512
519
.

88

Bekfani
T
,
Pellicori
P
,
Morris
DA
,
Ebner
N
,
Valentova
M
,
Steinbeck
L
,
Wachter
R
,
Elsner
S
,
Sliziuk
V
,
Schefold
JC
,
Sandek
A
,
Doehner
W
,
Cleland
JG
,
Lainscak
M
,
Anker
SD
,
von Haehling
S
.
Sarcopenia in patients with heart failure with preserved ejection fraction: impact on muscle strength, exercise capacity and quality of life
.
Int J Cardiol
2016
;
222
:
41
46
.

89

Hajahmadi
M
,
Shemshadi
S
,
Khalilipur
E
,
Amin
A
,
Taghavi
S
,
Maleki
M
,
Malek
H
,
Naderi
N
.
Muscle wasting in young patients with dilated cardiomyopathy
.
J Cachexia Sarcopenia Muscle
2017
;
8
:
542
548
.

90

Saitoh
M
,
Dos Santos
MR
,
Emami
A
,
Ishida
J
,
Ebner
N
,
Valentova
M
,
Bekfani
T
,
Sandek
A
,
Lainscak
M
,
Doehner
W
,
Anker
SD
,
von Haehling
S
.
Anorexia, functional capacity, and clinical outcome in patients with chronic heart failure: results from the Studies Investigating Co-morbidities Aggravating Heart Failure (SICA-HF)
.
ESC Heart Fail
2017
;
4
:
448
457
.

91

da Fonseca
GWP
,
Garfias Macedo
T
,
Ebner
N
,
Dos Santos
MR
,
de Souza
FR
,
Mady
C
,
Takayama
L
,
Pereira
RMR
,
Doehner
W
,
Anker
SD
,
Negrão
CE
,
Alves
M
,
von Haehling
S
.
Muscle mass, muscle strength, and functional capacity in patients with heart failure of Chagas disease and other aetiologies
.
ESC Heart Fail
2020
;
7
:
3086
3094
.

92

Loncar
G
,
Garfias-Veitl
T
,
Valentova
M
,
Vatic
M
,
Lainscak
M
,
Obradović
D
,
Dschietzig
TB
,
Doehner
W
,
Jankowska
EA
,
Anker
SD
,
von Haehling
S
.
Bone status in men with heart failure: results from the Studies Investigating Co-morbidities Aggravating Heart Failure
.
Eur J Heart Fail
2023
;
25
:
714
723
.

93

Dos Santos
MR
,
Saitoh
M
,
Ebner
N
,
Valentova
M
,
Konishi
M
,
Ishida
J
,
Emami
A
,
Springer
J
,
Sandek
A
,
Doehner
W
,
Anker
SD
,
von Haehling
S
.
Sarcopenia and endothelial function in patients with chronic heart failure: results from the Studies Investigating Comorbidities Aggravating Heart Failure (SICA-HF)
.
J Am Med Dir Assoc
2017
;
18
:
240
245
.

94

von Haehling
S
,
Garfias Macedo
T
,
Valentova
M
,
Anker
MS
,
Ebner
N
,
Bekfani
T
,
Haarmann
H
,
Schefold
JC
,
Lainscak
M
,
Cleland
JGF
,
Doehner
W
,
Hasenfuss
G
,
Anker
SD
.
Muscle wasting as an independent predictor of survival in patients with chronic heart failure
.
J Cachexia Sarcopenia Muscle
2020
;
11
:
1242
1249
.

95

Anker
SD
,
Morley
JE
,
von Haehling
S
.
Welcome to the ICD-10 code for sarcopenia
.
J Cachexia Sarcopenia Muscle
2016
;
7
:
512
514
.

96

Kim
KM
,
Jang
HC
,
Lim
S
.
Differences among skeletal muscle mass indices derived from height-, weight-, and body mass index-adjusted models in assessing sarcopenia
.
Korean J Intern Med
2016
;
31
:
643
650
.

97

Baumgartner
RN
,
Koehler
KM
,
Gallagher
D
,
Romero
L
,
Heymsfield
SB
,
Ross
RR
,
Garry
PJ
,
Lindeman
RD
.
Epidemiology of sarcopenia among the elderly in New Mexico
.
Am J Epidemiol
1998
;
147
:
755
763
.

98

Janssen
I
,
Heymsfield
SB
,
Ross
R
.
Low relative skeletal muscle mass (sarcopenia) in older persons is associated with functional impairment and physical disability
.
J Am Geriatr Soc
2002
;
50
:
889
896
.

99

Studenski
SA
,
Peters
KW
,
Alley
DE
,
Cawthon
PM
,
McLean
RR
,
Harris
TB
,
Ferrucci
L
,
Guralnik
JM
,
Fragala
MS
,
Kenny
AM
,
Kiel
DP
,
Kritchevsky
SB
,
Shardell
MD
,
Dam
T-TL
,
Vassileva
MT
.
The FNIH sarcopenia project: rationale, study description, conference recommendations, and final estimates
.
J Gerontol A Biol Sci Med Sci
2014
;
69
:
547
558
.

100

Newman
AB
,
Kupelian
V
,
Visser
M
,
Simonsick
E
,
Goodpaster
B
,
Nevitt
M
,
Kritchevsky
SB
,
Tylavsky
FA
,
Rubin
SM
,
Harris
TB
.
Sarcopenia: alternative definitions and associations with lower extremity function
.
J Am Geriatr Soc
2003
;
51
:
1602
1609
.

101

da Fonseca
GWP
,
von Haehling
S
.
The fatter, the better in old age: the current understanding of a difficult relationship
.
Curr Opin Clin Nutr Metab Care
2022
;
25
:
1
6
.

102

Fonseca
GWPD
,
Dos Santos
MR
,
de Souza
FR
,
Takayama
L
,
Rodrigues Pereira
RM
,
Negrão
CE
,
Alves
MNN
.
Discriminating sarcopenia in overweight/obese male patients with heart failure: the influence of body mass index
.
ESC Heart Fail
2020
;
7
:
84
91
.

103

Marzetti
E
,
Calvani
R
,
Cesari
M
,
Tosato
M
,
Cherubini
A
,
Di Bari
M
,
Pahor
M
,
Savera
G
,
Collamati
A
,
D’Angelo
E
,
Bernabei
R
,
Landi
F
.
Operationalization of the physical frailty & sarcopenia syndrome: rationale and clinical implementation
.
Transl Med UniSa
2015
;
13
:
29
32
.

104

Fozouni
L
,
Wang
CW
,
Lai
JC
.
Sex differences in the association between frailty and sarcopenia in patients with cirrhosis
.
Clin Transl Gastroenterol
2019
;
10
:
e00102
.

105

Feng
H
,
Wang
X
,
Mao
L
,
Yu
Z
,
Cui
B
,
Lin
L
,
Hui
Y
,
Zhao
X
,
Xu
X
,
Fan
X
,
Wang
B
,
Yu
Q
,
Jiang
K
,
Sun
C
.
Relationship between sarcopenia/myosteatosis and frailty in hospitalized patients with cirrhosis: a sex-stratified analysis
.
Ther Adv Chronic Dis
2021
;
12
:
20406223211026996
.

106

Veronese
N
,
Cereda
E
,
Stubbs
B
,
Solmi
M
,
Luchini
C
,
Manzato
E
,
Sergi
G
,
Manu
P
,
Harris
T
,
Fontana
L
,
Strandberg
T
,
Amieva
H
,
Dumurgier
J
,
Elbaz
A
,
Tzourio
C
,
Eicholzer
M
,
Rohrmann
S
,
Moretti
C
,
D’Ascenzo
F
,
Quadri
G
,
Polidoro
A
,
Lourenço
RA
,
Moreira
VG
,
Sanchis
J
,
Scotti
V
,
Maggi
S
,
Correll
CU
.
Risk of cardiovascular disease morbidity and mortality in frail and pre-frail older adults: results from a meta-analysis and exploratory meta-regression analysis
.
Ageing Res Rev
2017
;
35
:
63
73
.

107

Shrauner
W
,
Lord
EM
,
Nguyen
XT
,
Song
RJ
,
Galloway
A
,
Gagnon
DR
,
Driver
JA
,
Gaziano
JM
,
Wilson
PWF
,
Djousse
L
,
Cho
K
,
Orkaby
AR
.
Frailty and cardiovascular mortality in more than 3 million US veterans
.
Eur Heart J
2022
;
43
:
818
826
.

108

Sze
S
,
Pellicori
P
,
Zhang
J
,
Weston
J
,
Clark
AL
.
Identification of frailty in chronic heart failure
.
JACC Heart Fail
2019
;
7
:
291
302
.

109

Vidán
MT
,
Blaya-Novakova
V
,
Sánchez
E
,
Ortiz
J
,
Serra-Rexach
JA
,
Bueno
H
.
Prevalence and prognostic impact of frailty and its components in non-dependent elderly patients with heart failure
.
Eur J Heart Fail
2016
;
18
:
869
875
.

110

Damluji
AA
,
Chung
SE
,
Xue
QL
,
Hasan
RK
,
Walston
JD
,
Forman
DE
,
Bandeen-Roche
K
,
Moscucci
M
,
Batchelor
W
,
Resar
JR
,
Gerstenblith
G
.
Physical frailty phenotype and the development of geriatric syndromes in older adults with coronary heart disease
.
Am J Med
2021
;
134
:
662
671.e1
.

111

Hong
S
,
Chang
Y
,
Jung
HS
,
Yun
KE
,
Shin
H
,
Ryu
S
.
Relative muscle mass and the risk of incident type 2 diabetes: a cohort study
.
PLoS One
2017
;
12
:
e0188650
.

112

Chang
K-V
,
Hsu
T-H
,
Wu
W-T
,
Huang
K-C
,
Han
D-S
.
Association between sarcopenia and cognitive impairment: a systematic review and meta-analysis
.
J Am Med Dir Assoc
2016
;
17
:
1164.e7
1164.e15
.

113

Chang
K-V
,
Hsu
T-H
,
Wu
W-T
,
Huang
K-C
,
Han
D-S
.
Is sarcopenia associated with depression? A systematic review and meta-analysis of observational studies
.
Age Ageing
2017
;
46
:
738
746
.

114

Maeda
D
,
Matsue
Y
,
Kagiyama
N
,
Jujo
K
,
Saito
K
,
Kamiya
K
,
Saito
H
,
Ogasahara
Y
,
Maekawa
E
,
Konishi
M
,
Kitai
T
,
Iwata
K
,
Wada
H
,
Hiki
M
,
Dotare
T
,
Sunayama
T
,
Kasai
T
,
Nagamatsu
H
,
Ozawa
T
,
Izawa
K
,
Yamamoto
S
,
Aizawa
N
,
Wakaume
K
,
Oka
K
,
Momomura
SI
,
Minamino
T
.
Sex differences in the prevalence and prognostic impact of physical frailty and sarcopenia among older patients with heart failure
.
Nutr Metab Cardiovasc Dis
2022
;
32
:
365
372
.

115

Hamada
T
,
Kubo
T
,
Kawai
K
,
Nakaoka
Y
,
Yabe
T
,
Furuno
T
,
Yamada
E
,
Kitaoka
H
;
Kochi YOSACOI study
.
Clinical characteristics and frailty status in heart failure with preserved vs. reduced ejection fraction
.
ESC Heart Fail
2022
;
9
:
1853
1863
.

116

Nadruz
W
Jr,
Kitzman
D
,
Windham
BG
,
Kucharska-Newton
A
,
Butler
K
,
Palta
P
,
Griswold
ME
,
Wagenknecht
LE
,
Heiss
G
,
Solomon
SD
,
Skali
H
,
Shah
AM
.
Cardiovascular dysfunction and frailty among older adults in the community: the ARIC study
.
J Gerontol A Biol Sci Med Sci
2017
;
72
:
958
964
.

117

Yang
X
,
Woo
J
,
Ting Lui
L
,
Li
Q
,
Fai Cheng
K
,
Fan
Y
,
Yau
F
,
Lee
APW
,
Lee
JSW
,
Fung
E
.
Cardiac manifestations of sarcopenia
.
J Nutr Health Aging
2020
;
24
:
478
484
.

118

Bellumkonda
L
,
Tyrrell
D
,
Hummel
SL
,
Goldstein
DR
.
Pathophysiology of heart failure and frailty: a common inflammatory origin?
Aging Cell
2017
;
16
:
444
450
.

119

López-Otín
C
,
Blasco
MA
,
Partridge
L
,
Serrano
M
,
Kroemer
G
.
The hallmarks of aging
.
Cell
2013
;
153
:
1194
1217
.

120

Di Ciaula
A
,
Portincasa
P
.
The environment as a determinant of successful aging or frailty
.
Mech Ageing Dev
2020
;
188
:
111244
.

121

Cho
J
,
Choi
Y
,
Sajgalik
P
,
No
M-H
,
Lee
S-H
,
Kim
S
,
Heo
J-W
,
Cho
E-J
,
Chang
E
,
Kang
J-H
,
Kwak
H-B
,
Park
D-H
.
Exercise as a therapeutic strategy for sarcopenia in heart failure: insights into underlying mechanisms
.
Cells
2020
;
9
:
2284
.

122

Kumar
AA
,
Kelly
DP
,
Chirinos
JA
.
Mitochondrial dysfunction in heart failure with preserved ejection fraction
.
Circulation
2019
;
139
:
1435
1450
.

123

De Meyer
GRY
,
De Keulenaer
GW
,
Martinet
W
.
Role of autophagy in heart failure associated with aging
.
Heart Fail Rev
2010
;
15
:
423
430
.

124

Cittadini
A
,
Marra
AM
,
Arcopinto
M
,
Bobbio
E
,
Salzano
A
,
Sirico
D
,
Napoli
R
,
Colao
A
,
Longobardi
S
,
Baliga
RR
,
Bossone
E
,
Saccà
L
.
Growth hormone replacement delays the progression of chronic heart failure combined with growth hormone deficiency: an extension of a randomized controlled single-blind study
.
JACC Heart Fail
2013
;
1
:
325
330
.

125

Chocron
ES
,
Munkácsy
E
,
Pickering
AM
.
Cause or casualty: the role of mitochondrial DNA in aging and age-associated disease
.
Biochim Biophys Acta Mol Basis Dis
2019
;
1865
:
285
297
.

126

Chabi
B
,
Ljubicic
V
,
Menzies
KJ
,
Huang
JH
,
Saleem
A
,
Hood
DA
.
Mitochondrial function and apoptotic susceptibility in aging skeletal muscle
.
Aging Cell
2008
;
7
:
2
12
.

127

Dai
DF
,
Chen
T
,
Szeto
H
,
Nieves-Cintrón
M
,
Kutyavin
V
,
Santana
LF
,
Rabinovitch
PS
.
Mitochondrial targeted antioxidant peptide ameliorates hypertensive cardiomyopathy
.
J Am Coll Cardiol
2011
;
58
:
73
82
.

128

Ji
CH
,
Kwon
YT
.
Crosstalk and interplay between the ubiquitin-proteasome system and autophagy
.
Mol Cells
2017
;
40
:
441
449
.

129

Bodine
SC
,
Latres
E
,
Baumhueter
S
,
Lai
VK
,
Nunez
L
,
Clarke
BA
,
Poueymirou
WT
,
Panaro
FJ
,
Na
E
,
Dharmarajan
K
,
Pan
ZQ
,
Valenzuela
DM
,
DeChiara
TM
,
Stitt
TN
,
Yancopoulos
GD
,
Glass
DJ
.
Identification of ubiquitin ligases required for skeletal muscle atrophy
.
Science (New York, NY)
2001
;
294
:
1704
1708
.

130

Gumucio
JP
,
Mendias
CL
.
Atrogin-1, MuRF-1, and sarcopenia
.
Endocrine
2013
;
43
:
12
21
.

131

Katano
S
,
Yano
T
,
Shimizu
M
,
Ohori
K
,
Kouzu
H
,
Koyama
M
,
Nagaoka
R
,
Inoue
T
,
Takamura
Y
,
Ishigo
T
,
Takashima
H
,
Katayose
M
,
Ohnishi
H
,
Miura
T
.
Does renin-angiotensin system inhibition have impacts on muscle mass and bone mineral density in heart failure patients?
ESC Heart Fail
2021
;
8
:
2617
2624
.

132

Emmerson
PJ
,
Duffin
KL
,
Chintharlapalli
S
,
Wu
X
.
GDF15 and growth control
.
Front Physiol
2018
;
9
:
1712
.

133

Alcazar
J
,
Frandsen
U
,
Prokhorova
T
,
Kamper
RS
,
Haddock
B
,
Aagaard
P
,
Suetta
C
.
Changes in systemic GDF15 across the adult lifespan and their impact on maximal muscle power: the Copenhagen Sarcopenia Study
.
J Cachexia Sarcopenia Muscle
2021
;
12
:
1418
1427
.

134

Hagström
E
,
Held
C
,
Stewart
RA
,
Aylward
PE
,
Budaj
A
,
Cannon
CP
,
Koenig
W
,
Krug-Gourley
S
,
Mohler
ER
3rd
,
Steg
PG
,
Tarka
E
,
Östlund
O
,
White
HD
,
Siegbahn
A
,
Wallentin
L
.
Growth differentiation factor 15 predicts all-cause morbidity and mortality in stable coronary heart disease
.
Clin Chem
2017
;
63
:
325
333
.

135

Anand
IS
,
Kempf
T
,
Rector
TS
,
Tapken
H
,
Allhoff
T
,
Jantzen
F
,
Kuskowski
M
,
Cohn
JN
,
Drexler
H
,
Wollert
KC
.
Serial measurement of growth-differentiation factor-15 in heart failure: relation to disease severity and prognosis in the Valsartan Heart Failure Trial
.
Circulation
2010
;
122
:
1387
1395
.

136

Wang
Y
,
Zhen
C
,
Wang
R
,
Wang
G
.
Growth-differentiation factor-15 predicts adverse cardiac events in patients with acute coronary syndrome: a meta-analysis
.
Am J Emerg Med
2019
;
37
:
1346
1352
.

137

Bettencourt
P
,
Ferreira-Coimbra
J
,
Rodrigues
P
,
Marques
P
,
Moreira
H
,
Pinto
MJ
,
Guimarães
JT
,
Lourenço
P
.
Towards a multi-marker prognostic strategy in acute heart failure: a role for GDF-15
.
ESC Heart Fail
2018
;
5
:
1017
1022
.

138

Albuquerque
B
,
Chen
X
,
Hirenallur-Shanthappa
D
,
Zhao
Y
,
Stansfield
JC
,
Zhang
BB
,
Sheikh
A
,
Wu
Z
.
Neutralization of GDF15 prevents anorexia and weight loss in the monocrotaline-induced cardiac cachexia rat model
.
Cells
2022
;
11
:
1073
.

139

Brambilla
F
,
Bellodi
L
,
Brunetta
M
,
Perna
G
.
Plasma concentrations of interleukin-1 beta, interleukin-6 and tumor necrosis factor-alpha in anorexia and bulimia nervosa
.
Psychoneuroendocrinology
1998
;
23
:
439
447
.

140

Guttridge
DC
,
Mayo
MW
,
Madrid
LV
,
Wang
C-Y
,
Baldwin
AS
Jr
.
NF-kappaB-induced loss of MyoD messenger RNA: possible role in muscle decay and cachexia
.
Science (New York, NY)
2000
;
289
:
2363
2366
.

141

Reid
MB
,
Li
Y-P
.
Tumor necrosis factor-alpha and muscle wasting: a cellular perspective
.
Respir Res
2001
;
2
:
269
272
.

142

da Fonseca
GWP
,
von Haehling
S
.
Sarcopaenia complicating heart failure
.
Eur Heart J Suppl
2019
;
21
:
L20
L23
.

143

von Haehling
S
,
Schefold
JC
,
Lainscak
M
,
Doehner
W
,
Anker
SD
.
Inflammatory biomarkers in heart failure revisited: much more than innocent bystanders
.
Heart Fail Clin
2009
;
5
:
549
560
.

144

Hoeijmakers
JHJ
.
DNA damage, aging, and cancer
.
N Engl J Med
2009
;
361
:
1475
1485
.

145

Lord
CJ
,
Ashworth
A
.
The DNA damage response and cancer therapy
.
Nature
2012
;
481
:
287
294
.

146

Hou
Y
,
Song
H
,
Croteau
DL
,
Akbari
M
,
Bohr
VA
.
Genome instability in Alzheimer disease
.
Mech Ageing Dev
2017
;
161
:
83
94
.

147

Capell
BC
,
Collins
FS
,
Nabel
EG
.
Mechanisms of cardiovascular disease in accelerated aging syndromes
.
Circ Res
2007
;
101
:
13
26
.

148

García-Prat
L
,
Martínez-Vicente
M
,
Perdiguero
E
,
Ortet
L
,
Rodríguez-Ubreva
J
,
Rebollo
E
,
Ruiz-Bonilla
V
,
Gutarra
S
,
Ballestar
E
,
Serrano
AL
,
Sandri
M
,
Muñoz-Cánoves
P
.
Autophagy maintains stemness by preventing senescence
.
Nature
2016
;
529
:
37
42
.

149

Varela
E
,
Blasco
MA
.
2009 Nobel Prize in physiology or medicine: telomeres and telomerase
.
Oncogene
2010
;
29
:
1561
1565
.

150

Wu
X
,
Amos
CI
,
Zhu
Y
,
Zhao
H
,
Grossman
BH
,
Shay
JW
,
Luo
S
,
Hong
WK
,
Spitz
MR
.
Telomere dysfunction: a potential cancer predisposition factor
.
J Natl Cancer Inst
2003
;
95
:
1211
1218
.

151

Adaikalakoteswari
A
,
Balasubramanyam
M
,
Mohan
V
.
Telomere shortening occurs in Asian Indian type 2 diabetic patients
.
Diabet Med
2005
;
22
:
1151
1156
.

152

Wu
C-T
,
Morris
JR
.
Genes, genetics, and epigenetics: a correspondence
.
Science (New York, NY)
2001
;
293
:
1103
1105
.

153

Brivio
P
,
Paladini
MS
,
Racagni
G
,
Riva
MA
,
Calabrese
F
,
Molteni
R
.
From healthy aging to frailty: in search of the underlying mechanisms
.
Curr Med Chem
2019
;
26
:
3685
3701
.

154

Talens
RP
,
Christensen
K
,
Putter
H
,
Willemsen
G
,
Christiansen
L
,
Kremer
D
,
Suchiman
HE
,
Slagboom
PE
,
Boomsma
DI
,
Heijmans
BT
.
Epigenetic variation during the adult lifespan: cross-sectional and longitudinal data on monozygotic twin pairs
.
Aging Cell
2012
;
11
:
694
703
.

155

Pal
S
,
Tyler
JK
.
Epigenetics and aging
.
Sci Adv
2016
;
2
:
e1600584
.

156

Horvath
S
.
DNA methylation age of human tissues and cell types
.
Genome Biol
2013
;
14
:
R115
.

157

Florath
I
,
Butterbach
K
,
Müller
H
,
Bewerunge-Hudler
M
,
Brenner
H
.
Cross-sectional and longitudinal changes in DNA methylation with age: an epigenome-wide analysis revealing over 60 novel age-associated CpG sites
.
Hum Mol Genet
2014
;
23
:
1186
1201
.

158

Jin
C
,
Li
J
,
Green
CD
,
Yu
X
,
Tang
X
,
Han
D
,
Xian
B
,
Wang
D
,
Huang
X
,
Cao
X
,
Yan
Z
,
Hou
L
,
Liu
J
,
Shukeir
N
,
Khaitovich
P
,
Chen
CD
,
Zhang
H
,
Jenuwein
T
,
Han
J-D
.
Histone demethylase UTX-1 regulates C. elegans life span by targeting the insulin/IGF-1 signaling pathway
.
Cell Metab
2011
;
14
:
161
172
.

159

Ugalde
AP
,
Español
Y
,
López-Otín
C
.
Micromanaging aging with miRNAs: new messages from the nuclear envelope
.
Nucleus (Austin, Tex)
2011
;
2
:
549
555
.

160

Zhang
Y
,
Huang
B
,
Wang
H-Y
,
Chang
A
,
Zheng
XFS
.
Emerging role of microRNAs in mTOR signaling
.
Cell Mol Life Sci
2017
;
74
:
2613
2625
.

161

Labbadia
J
,
Morimoto
RI
.
The biology of proteostasis in aging and disease
.
Annu Rev Biochem
2015
;
84
:
435
464
.

162

Powers
ET
,
Morimoto
RI
,
Dillin
A
,
Kelly
JW
,
Balch
WE
.
Biological and chemical approaches to diseases of proteostasis deficiency
.
Annu Rev Biochem
2009
;
78
:
959
991
.

163

Hartl
FU
,
Bracher
A
,
Hayer-Hartl
M
.
Molecular chaperones in protein folding and proteostasis
.
Nature
2011
;
475
:
324
332
.

164

Calderwood
SK
,
Murshid
A
,
Prince
T
.
The shock of aging: molecular chaperones and the heat shock response in longevity and aging—a mini-review
.
Gerontology
2009
;
55
:
550
558
.

165

Westerheide
SD
,
Anckar
J
,
Stevens
SM
Jr
,
Sistonen
L
,
Morimoto
RI
.
Stress-inducible regulation of heat shock factor 1 by the deacetylase SIRT1
.
Science (New York, NY)
2009
;
323
:
1063
1066
.

166

Mohamed
JS
,
Wilson
JC
,
Myers
MJ
,
Sisson
KJ
,
Alway
SE
.
Dysregulation of SIRT-1 in aging mice increases skeletal muscle fatigue by a PARP-1-dependent mechanism
.
Aging
2014
;
6
:
820
834
.

167

Wuertz
K
,
Vo
N
,
Kletsas
D
,
Boos
N
.
Inflammatory and catabolic signalling in intervertebral discs: the roles of NF-κB and MAP kinases
.
Eur Cell Mater
2012
;
23
:
103
119
;
discussion 119–120
.

168

Schakman
O
,
Kalista
S
,
Barbé
C
,
Loumaye
A
,
Thissen
JP
.
Glucocorticoid-induced skeletal muscle atrophy
.
Int J Biochem Cell Biol
2013
;
45
:
2163
2172
.

169

Zhang
G
,
Li
Y-P
.
P38β MAPK upregulates atrogin1/MAFbx by specific phosphorylation of C/EBPβ
.
Skelet Muscle
2012
;
2
:
20
.

170

Ladner
KJ
,
Caligiuri
MA
,
Guttridge
DC
.
Tumor necrosis factor-regulated biphasic activation of NF-kappa B is required for cytokine-induced loss of skeletal muscle gene products
.
J Biol Chem
2003
;
278
:
2294
2303
.

171

Cai
D
,
Frantz
JD
,
Tawa
NE
Jr
,
Melendez
PA
,
Oh
B-C
,
Lidov
HGW
,
Hasselgren
P-O
,
Frontera
WR
,
Lee
J
,
Glass
DJ
,
Shoelson
SE
.
IKKβ/NF-κB activation causes severe muscle wasting in mice
.
Cell
2004
;
119
:
285
298
.

172

Greer
EL
,
Brunet
A
.
FOXO transcription factors at the interface between longevity and tumor suppression
.
Oncogene
2005
;
24
:
7410
7425
.

173

Waddell
DS
,
Baehr
LM
,
van den Brandt
J
,
Johnsen
SA
,
Reichardt
HM
,
Furlow
JD
,
Bodine
SC
.
The glucocorticoid receptor and FOXO1 synergistically activate the skeletal muscle atrophy-associated MuRF1 gene
.
Am J Physiol Endocrinol Metab
2008
;
295
:
E785
E797
.

174

Sandri
M
,
Sandri
C
,
Gilbert
A
,
Skurk
C
,
Calabria
E
,
Picard
A
,
Walsh
K
,
Schiaffino
S
,
Lecker
SH
,
Goldberg
AL
.
Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy
.
Cell
2004
;
117
:
399
412
.

175

Stitt
TN
,
Drujan
D
,
Clarke
BA
,
Panaro
F
,
Timofeyva
Y
,
Kline
WO
,
Gonzalez
M
,
Yancopoulos
GD
,
Glass
DJ
.
The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors
.
Mol Cell
2004
;
14
:
395
403
.

176

Allen
DL
,
Loh
AS
.
Posttranscriptional mechanisms involving microRNA-27a and b contribute to fast-specific and glucocorticoid-mediated myostatin expression in skeletal muscle
.
Am J Physiol Cell Physiol
2011
;
300
:
C124
C137
.

177

McPherron
AC
,
Lawler
AM
,
Lee
SJ
.
Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member
.
Nature
1997
;
387
:
83
90
.

178

Lee
SJ
,
McPherron
AC
.
Regulation of myostatin activity and muscle growth
.
Proc Natl Acad Sci U S A
2001
;
98
:
9306
9311
.

179

Shimizu
N
,
Yoshikawa
N
,
Ito
N
,
Maruyama
T
,
Suzuki
Y
,
Takeda
S
,
Nakae
J
,
Tagata
Y
,
Nishitani
S
,
Takehana
K
,
Sano
M
,
Fukuda
K
,
Suematsu
M
,
Morimoto
C
,
Tanaka
H
.
Crosstalk between glucocorticoid receptor and nutritional sensor mTOR in skeletal muscle
.
Cell Metab
2011
;
13
:
170
182
.

180

Wang
X-P
,
Huang
Z
,
Li
Y-L
,
Jin
K-Y
,
Dong
D-J
,
Wang
J-X
,
Zhao
X-F
.
Krüppel-like factor 15 integrated autophagy and gluconeogenesis to maintain glucose homeostasis under 20-hydroxyecdysone regulation
.
PLoS Genet
2022
;
18
:
e1010229
.

181

Fan
J
,
Kou
X
,
Jia
S
,
Yang
X
,
Yang
Y
,
Chen
N
.
Autophagy as a potential target for sarcopenia
.
J Cell Physiol
2016
;
231
:
1450
1459
.

182

Russ
DW
,
Boyd
IM
,
McCoy
KM
,
McCorkle
KW
.
Muscle-specificity of age-related changes in markers of autophagy and sphingolipid metabolism
.
Biogerontology
2015
;
16
:
747
759
.

183

White
Z
,
White
RB
,
McMahon
C
,
Grounds
MD
,
Shavlakadze
T
.
High mTORC1 signaling is maintained, while protein degradation pathways are perturbed in old murine skeletal muscles in the fasted state
.
Int J Biochem Cell Biol
2016
;
78
:
10
21
.

184

Campanario
S
,
Ramírez-Pardo
I
,
Hong
X
,
Isern
J
,
Muñoz-Cánoves
P
.
Assessing autophagy in muscle stem cells
.
Front Cell Dev Biol
2020
;
8
:
620409
.

185

Masiero
E
,
Agatea
L
,
Mammucari
C
,
Blaauw
B
,
Loro
E
,
Komatsu
M
,
Metzger
D
,
Reggiani
C
,
Schiaffino
S
,
Sandri
M
.
Autophagy is required to maintain muscle mass
.
Cell Metab
2009
;
10
:
507
515
.

186

Maurin
A-C
,
Parry
L
,
B’chir
W
,
Carraro
V
,
Coudy-Gandilhon
C
,
Chaouki
G
,
Chaveroux
C
,
Mordier
S
,
Martinie
B
,
Reinhardt
V
,
Jousse
C
,
Bruhat
A
,
Codogno
P
,
Averous
J
,
Fafournoux
P
.
GCN2 upregulates autophagy in response to short-term deprivation of a single essential amino acid
.
Autophagy Rep
2022
;
1
:
119
142
.

187

Mammucari
C
,
Milan
G
,
Romanello
V
,
Masiero
E
,
Rudolf
R
,
Del Piccolo
P
,
Burden
SJ
,
Di Lisi
R
,
Sandri
C
,
Zhao
J
,
Goldberg
AL
,
Schiaffino
S
,
Sandri
M
.
Foxo3 controls autophagy in skeletal muscle in vivo
.
Cell Metab
2007
;
6
:
458
471
.

188

Fulks
RM
,
Li
JB
,
Goldberg
AL
.
Effects of insulin, glucose, and amino acids on protein turnover in rat diaphragm
.
J Biol Chem
1975
;
250
:
290
298
.

189

Manning
BD
,
Cantley
LC
.
AKT/PKB signaling: navigating downstream
.
Cell
2007
;
129
:
1261
1274
.

190

McFarlane
C
,
Plummer
E
,
Thomas
M
,
Hennebry
A
,
Ashby
M
,
Ling
N
,
Smith
H
,
Sharma
M
,
Kambadur
R
.
Myostatin induces cachexia by activating the ubiquitin proteolytic system through an NF-kappaB-independent, FoxO1–dependent mechanism
.
J Cell Physiol
2006
;
209
:
501
514
.

191

Hirose
Y
,
Onishi
T
,
Miura
S
,
Hatazawa
Y
,
Kamei
Y
.
Vitamin D attenuates FOXO1-target atrophy gene expression in C2C12 muscle cells
.
J Nutr Sci Vitaminol
2018
;
64
:
229
232
.

192

Storer
TW
,
Basaria
S
,
Traustadottir
T
,
Harman
SM
,
Pencina
K
,
Li
Z
,
Travison
TG
,
Miciek
R
,
Tsitouras
P
,
Hally
K
,
Huang
G
,
Bhasin
S
.
Effects of testosterone supplementation for 3 years on muscle performance and physical function in older men
.
J Clin Endocrinol Metab
2017
;
102
:
583
593
.

193

Geraci
A
,
Calvani
R
,
Ferri
E
,
Marzetti
E
,
Arosio
B
,
Cesari
M
.
Sarcopenia and menopause: the role of estradiol
.
Front Endocrinol (Lausanne)
2021
;
12
:
682012
.

194

Lin
J
,
Wu
H
,
Tarr
PT
,
Zhang
C-Y
,
Wu
Z
,
Boss
O
,
Michael
LF
,
Puigserver
P
,
Isotani
E
,
Olson
EN
,
Lowell
BB
,
Bassel-Duby
R
,
Spiegelman
BM
.
Transcriptional co-activator PGC-1 alpha drives the formation of slow-twitch muscle fibres
.
Nature
2002
;
418
:
797
801
.

195

Anderson
R
,
Prolla
T
.
PGC-1alpha in aging and anti-aging interventions
.
Biochim Biophys Acta
2009
;
1790
:
1059
1066
.

196

Sandri
M
,
Lin
J
,
Handschin
C
,
Yang
W
,
Arany
ZP
,
Lecker
SH
,
Goldberg
AL
,
Spiegelman
BM
.
PGC-1alpha protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription
.
Proc Natl Acad Sci U S A
2006
;
103
:
16260
16265
.

197

Costford
SR
,
Bajpeyi
S
,
Pasarica
M
,
Albarado
DC
,
Thomas
SC
,
Xie
H
,
Church
TS
,
Jubrias
SA
,
Conley
KE
,
Smith
SR
.
Skeletal muscle NAMPT is induced by exercise in humans
.
Am J Physiol Endocrinol Metab
2010
;
298
:
E117
E126
.

198

Thirupathi
A
,
de Souza
CT
.
Multi-regulatory network of ROS: the interconnection of ROS, PGC-1 alpha, and AMPK-SIRT1 during exercise
.
J Physiol Biochem
2017
;
73
:
487
494
.

199

Garnier
A
,
Fortin
D
,
Deloménie
C
,
Momken
I
,
Veksler
V
,
Ventura-Clapier
R
.
Depressed mitochondrial transcription factors and oxidative capacity in rat failing cardiac and skeletal muscles
.
J Physiol (Lond)
2003
;
551
:
491
501
.

200

Rom
O
,
Reznick
AZ
.
The role of E3 ubiquitin-ligases MuRF-1 and MAFbx in loss of skeletal muscle mass
.
Free Radic Biol Med
2016
;
98
:
218
230
.

201

Mecocci
P
,
Fanó
G
,
Fulle
S
,
MacGarvey
U
,
Shinobu
L
,
Polidori
MC
,
Cherubini
A
,
Vecchiet
J
,
Senin
U
,
Beal
MF
.
Age-dependent increases in oxidative damage to DNA, lipids, and proteins in human skeletal muscle
.
Free Radic Biol Med
1999
;
26
:
303
308
.

202

Szentesi
P
,
Csernoch
L
,
Dux
L
,
Keller-Pintér
A
.
Changes in redox signaling in the skeletal muscle with aging
.
Oxid Med Cell Longev
2019
;
2019
:
4617801
.

203

Bouzid
MA
,
Filaire
E
,
McCall
A
,
Fabre
C
.
Radical oxygen species, exercise and aging: an update
.
Sports Med
2015
;
45
:
1245
1261
.

204

Viña
J
,
Borras
C
,
Gomez-Cabrera
MC
.
A free radical theory of frailty
.
Free Radic Biol Med
2018
;
124
:
358
363
.

205

Kuilman
T
,
Michaloglou
C
,
Mooi
WJ
,
Peeper
DS
.
The essence of senescence
.
Genes Dev
2010
;
24
:
2463
2479
.

206

Bodnar
AG
,
Ouellette
M
,
Frolkis
M
,
Holt
SE
,
Chiu
CP
,
Morin
GB
,
Harley
CB
,
Shay
JW
,
Lichtsteiner
S
,
Wright
WE
.
Extension of life-span by introduction of telomerase into normal human cells
.
Science (New York, NY)
1998
;
279
:
349
352
.

207

Serrano
M
,
Lin
AW
,
McCurrach
ME
,
Beach
D
,
Lowe
SW
.
Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a
.
Cell
1997
;
88
:
593
602
.

208

Scott
LJ
,
Mohlke
KL
,
Bonnycastle
LL
,
Willer
CJ
,
Li
Y
,
Duren
WL
,
Erdos
MR
,
Stringham
HM
,
Chines
PS
,
Jackson
AU
,
Prokunina-Olsson
L
,
Ding
CJ
,
Swift
AJ
,
Narisu
N
,
Hu
T
,
Pruim
R
,
Xiao
R
,
Li
XY
,
Conneely
KN
,
Riebow
NL
,
Sprau
AG
,
Tong
M
,
White
PP
,
Hetrick
KN
,
Barnhart
MW
,
Bark
CW
,
Goldstein
JL
,
Watkins
L
,
Xiang
F
,
Saramies
J
,
Buchanan
TA
,
Watanabe
RM
,
Valle
TT
,
Kinnunen
L
,
Abecasis
GR
,
Pugh
EW
,
Doheny
KF
,
Bergman
RN
,
Tuomilehto
J
,
Collins
FS
,
Boehnke
M
.
A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants
.
Science (New York, NY)
2007
;
316
:
1341
1345
.

209

Baker
DJ
,
Wijshake
T
,
Tchkonia
T
,
LeBrasseur
NK
,
Childs
BG
,
van de Sluis
B
,
Kirkland
JL
,
van Deursen
JM
.
Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders
.
Nature
2011
;
479
:
232
236
.

210

Hoenicke
L
,
Zender
L
.
Immune surveillance of senescent cells—biological significance in cancer- and non-cancer pathologies
.
Carcinogenesis
2012
;
33
:
1123
1126
.

211

Campisi
J
,
Robert
L
.
Cell senescence: role in aging and age-related diseases
.
Interdiscip Top Gerontol
2014
;
39
:
45
61
.

212

Calcinotto
A
,
Kohli
J
,
Zagato
E
,
Pellegrini
L
,
Demaria
M
,
Alimonti
A
.
Cellular senescence: aging, cancer, and injury
.
Physiol Rev
2019
;
99
:
1047
1078
.

213

LeBrasseur
NK
,
Tchkonia
T
,
Kirkland
JL
.
Cellular senescence and the biology of aging, disease, and frailty
.
Nestle Nutr Inst Workshop Ser
2015
;
83
:
11
18
.

214

Rhinn
M
,
Ritschka
B
,
Keyes
WM
.
Cellular senescence in development, regeneration and disease
.
Development (Cambridge, England)
2019
;
146
:
dev151837
.

215

Kuilman
T
,
Michaloglou
C
,
Vredeveld
LC
,
Douma
S
,
van Doorn
R
,
Desmet
CJ
,
Aarden
LA
,
Mooi
WJ
,
Peeper
DS
.
Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network
.
Cell
2008
;
133
:
1019
1031
.

216

da Silva
PFL
,
Ogrodnik
M
,
Kucheryavenko
O
,
Glibert
J
,
Miwa
S
,
Cameron
K
,
Ishaq
A
,
Saretzki
G
,
Nagaraja-Grellscheid
S
,
Nelson
G
,
von Zglinicki
T
.
The bystander effect contributes to the accumulation of senescent cells in vivo
.
Aging Cell
2019
;
18
:
e12848
.

217

Joanisse
S
,
Nederveen
JP
,
Baker
JM
,
Snijders
T
,
Iacono
C
,
Parise
G
.
Exercise conditioning in old mice improves skeletal muscle regeneration
.
FASEB J
2016
;
30
:
3256
3268
.

218

Cisterna
B
,
Giagnacovo
M
,
Costanzo
M
,
Fattoretti
P
,
Zancanaro
C
,
Pellicciari
C
,
Malatesta
M
.
Adapted physical exercise enhances activation and differentiation potential of satellite cells in the skeletal muscle of old mice
.
J Anat
2016
;
228
:
771
783
.

219

Franceschi
C
,
Bonafè
M
,
Valensin
S
,
Olivieri
F
,
De Luca
M
,
Ottaviani
E
,
De Benedictis
G
.
Inflamm-aging. An evolutionary perspective on immunosenescence
.
Ann N Y Acad Sci
2000
;
908
:
244
254
.

220

Saedi
AA
,
Feehan
J
,
Phu
S
,
Duque
G
.
Current and emerging biomarkers of frailty in the elderly
.
Clin Interv Aging
2019
;
14
:
389
398
.

221

Rea
IM
,
Gibson
DS
,
McGilligan
V
,
McNerlan
SE
,
Alexander
HD
,
Ross
OA
.
Age and age-related diseases: role of inflammation triggers and cytokines
.
Front Immunol
2018
;
9
:
586
.

222

Zhuang
P
,
Zhang
J
,
Wang
Y
,
Zhang
M
,
Song
L
,
Lu
Z
,
Zhang
L
,
Zhang
F
,
Wang
J
,
Zhang
Y
,
Wei
H
,
Li
H
.
Reversal of muscle atrophy by Zhimu and Huangbai herb pair via activation of IGF-1/Akt and autophagy signal in cancer cachexia
.
Support Care Cancer
2016
;
24
:
1189
1198
.

223

Chen
M-C
,
Hsu
W-L
,
Hwang
P-A
,
Chen
Y-L
,
Chou
T-C
.
Combined administration of fucoidan ameliorates tumor and chemotherapy-induced skeletal muscle atrophy in bladder cancer-bearing mice
.
Oncotarget
2016
;
7
:
51608
51618
.

224

Li
Y-P
,
Chen
Y
,
John
J
,
Moylan
J
,
Jin
B
,
Mann
DL
,
Reid
MB
.
TNF-alpha acts via p38 MAPK to stimulate expression of the ubiquitin ligase atrogin1/MAFbx in skeletal muscle
.
FASEB J
2005
;
19
:
362
370
.

225

Braun
TP
,
Zhu
X
,
Szumowski
M
,
Scott
GD
,
Grossberg
AJ
,
Levasseur
PR
,
Graham
K
,
Khan
S
,
Damaraju
S
,
Colmers
WF
,
Baracos
VE
,
Marks
DL
.
Central nervous system inflammation induces muscle atrophy via activation of the hypothalamic-pituitary-adrenal axis
.
J Exp Med
2011
;
208
:
2449
2463
.

226

Cella
PS
,
Marinello
PC
,
Borges
FH
,
Ribeiro
DF
,
Chimin
P
,
Testa
MTJ
,
Guirro
PB
,
Duarte
JA
,
Cecchini
R
,
Guarnier
FA
,
Deminice
R
.
Creatine supplementation in Walker-256 tumor-bearing rats prevents skeletal muscle atrophy by attenuating systemic inflammation and protein degradation signaling
.
Eur J Nutr
2020
;
59
:
661
669
.

227

Vaisman
N
,
Barak
Y
,
Hahn
T
,
Karov
Y
,
Malach
L
,
Barak
V
.
Defective in vitro granulopoiesis in patients with anorexia nervosa
.
Pediatr Res
1996
;
40
:
108
111
.

228

Bodnar
RJ
,
Pasternak
GW
,
Mann
PE
,
Paul
D
,
Warren
R
,
Donner
DB
.
Mediation of anorexia by human recombinant tumor necrosis factor through a peripheral action in the rat
.
Cancer Res
1989
;
49
:
6280
6284
.

229

Turrin
NP
,
Ilyin
SE
,
Gayle
DA
,
Plata-Salamán
CR
,
Ramos
EJ
,
Laviano
A
,
Das
UN
,
Inui
A
,
Meguid
MM
.
Interleukin-1beta system in anorectic catabolic tumor-bearing rats
.
Curr Opin Clin Nutr Metab Care
2004
;
7
:
419
426
.

230

Yang
Z-J
,
Blaha
V
,
Meguid
MM
,
Laviano
A
,
Oler
A
,
Zadak
Z
.
Interleukin-1alpha injection into ventromedial hypothalamic nucleus of normal rats depresses food intake and increases release of dopamine and serotonin
.
Pharmacol Biochem Behav
1999
;
62
:
61
65
.

231

Gilliam-Davis
S
,
Payne
VS
,
Kasper
SO
,
Tommasi
EN
,
Robbins
ME
,
Diz
DI
.
Long-term AT1 receptor blockade improves metabolic function and provides renoprotection in Fischer-344 rats
.
Am J Physiol Heart Circ Physiol
2007
;
293
:
H1327
H1333
.

232

Herbert
KE
,
Mistry
Y
,
Hastings
R
,
Poolman
T
,
Niklason
L
,
Williams
B
.
Angiotensin II-mediated oxidative DNA damage accelerates cellular senescence in cultured human vascular smooth muscle cells via telomere-dependent and independent pathways
.
Circ Res
2008
;
102
:
201
208
.

233

Feng
X
,
Wang
L
,
Li
Y
.
Change of telomere length in angiotensin II-induced human glomerular mesangial cell senescence and the protective role of losartan
.
Mol Med Rep
2011
;
4
:
255
260
.

234

Kojima
G
,
Iliffe
S
,
Walters
K
.
Smoking as a predictor of frailty: a systematic review
.
BMC Geriatr
2015
;
15
:
131
.

235

Wakabayashi
H
,
Sakuma
K
.
Rehabilitation nutrition for sarcopenia with disability: a combination of both rehabilitation and nutrition care management
.
J Cachexia Sarcopenia Muscle
2014
;
5
:
269
277
.

236

Yin
J
,
Lu
X
,
Qian
Z
,
Xu
W
,
Zhou
X
.
New insights into the pathogenesis and treatment of sarcopenia in chronic heart failure
.
Theranostics
2019
;
9
:
4019
4029
.

237

Phu
S
,
Boersma
D
,
Duque
G
.
Exercise and sarcopenia
.
J Clin Densitom
2015
;
18
:
488
492
.

238

Nascimento
CM
,
Ingles
M
,
Salvador-Pascual
A
,
Cominetti
MR
,
Gomez-Cabrera
MC
,
Viña
J
.
Sarcopenia, frailty and their prevention by exercise
.
Free Radic Biol Med
2019
;
132
:
42
49
.

239

Hoppeler
H
,
Baum
O
,
Lurman
G
,
Mueller
M
.
Molecular mechanisms of muscle plasticity with exercise
.
Compr Physiol
2011
;
1
:
1383
1412
.

240

Landi
F
,
Cesari
M
,
Calvani
R
,
Cherubini
A
,
Di Bari
M
,
Bejuit
R
,
Mshid
J
,
Andrieu
S
,
Sinclair
AJ
,
Sieber
CC
,
Vellas
B
,
Topinkova
E
,
Strandberg
T
,
Rodriguez-Manas
L
,
Lattanzio
F
,
Pahor
M
,
Roubenoff
R
,
Cruz-Jentoft
AJ
,
Bernabei
R
,
Marzetti
E
;
SPRINTT Consortium
.
The “Sarcopenia and Physical frailty IN older people: multi-component Treatment strategies” (SPRINTT) randomized controlled trial: design and methods
.
Aging Clin Exp Res
2017
;
29
:
89
100
.

241

Herold
F
,
Törpel
A
,
Hamacher
D
,
Budde
H
,
Zou
L
,
Strobach
T
,
Müller
NG
,
Gronwald
T
.
Causes and consequences of interindividual response variability: a call to apply a more rigorous research design in acute exercise-cognition studies
.
Front Physiol
2021
;
12
:
682891
.

242

UNICEF, WHO and the World Bank Group Joint Child Malnutrition Estimates
.
Levels and Trends in Child Malnutrition: Key Findings of the 2019 Edition of the Joint Child Malnutrition Estimates
.
Geneva
:
World Health Organization
;
2020
.

243

Landi
F
,
Liperoti
R
,
Russo
A
,
Giovannini
S
,
Tosato
M
,
Barillaro
C
,
Capoluongo
E
,
Bernabei
R
,
Onder
G
.
Association of anorexia with sarcopenia in a community-dwelling elderly population: results from the ilSIRENTE study
.
Eur J Nutr
2013
;
52
:
1261
1268
.

244

Tacke
M
,
Ebner
N
,
Boschmann
M
,
Jarius
A
,
Valentova
M
,
Fülster
S
,
Sandek
A
,
Schomburg
L
,
Anker
SD
,
Doehner
W
,
von Haehling
S
.
Resting energy expenditure and the effects of muscle wasting in patients with chronic heart failure: results from the Studies Investigating Comorbidities Aggravating Heart Failure (SICA-HF)
.
J Am Med Dir Assoc
2013
;
14
:
837
841
.

245

Pojednic
RM
,
Ceglia
L
.
The emerging biomolecular role of vitamin D in skeletal muscle
.
Exerc Sport Sci Rev
2014
;
42
:
76
81
.

246

Dziegala
M
,
Josiak
K
,
Kasztura
M
,
Kobak
K
,
von Haehling
S
,
Banasiak
W
,
Anker
SD
,
Ponikowski
P
,
Jankowska
E
.
Iron deficiency as energetic insult to skeletal muscle in chronic diseases
.
J Cachexia Sarcopenia Muscle
2018
;
9
:
802
815
.

247

Witham
MD
.
Vitamin D in chronic heart failure
.
Curr Heart Fail Rep
2011
;
8
:
123
130
.

248

Busa
V
,
Dardeir
A
,
Marudhai
S
,
Patel
M
,
Valaiyaduppu Subas
S
,
Ghani
MR
,
Cancarevic
I
.
Role of vitamin D supplementation in heart failure patients with vitamin D deficiency and its effects on clinical outcomes: a literature review
.
Cureus
2020
;
12
:
e10840
.

249

Prado
CM
,
Landi
F
,
Chew
STH
,
Atherton
PJ
,
Molinger
J
,
Ruck
T
,
Gonzalez
MC
.
Advances in muscle health and nutrition: a toolkit for healthcare professionals
.
Clin Nutr
2022
;
41
:
2244
2263
.

250

Nunes
EA
,
Colenso-Semple
L
,
McKellar
SR
,
Yau
T
,
Ali
MU
,
Fitzpatrick-Lewis
D
,
Sherifali
D
,
Gaudichon
C
,
Tomé
D
,
Atherton
PJ
,
Robles
MC
,
Naranjo-Modad
S
,
Braun
M
,
Landi
F
,
Phillips
SM
.
Systematic review and meta-analysis of protein intake to support muscle mass and function in healthy adults
.
J Cachexia Sarcopenia Muscle
2022
;
13
:
795
810
.

251

Atherton
PJ
,
Etheridge
T
,
Watt
PW
,
Wilkinson
D
,
Selby
A
,
Rankin
D
,
Smith
K
,
Rennie
MJ
.
Muscle full effect after oral protein: time-dependent concordance and discordance between human muscle protein synthesis and mTORC1 signaling
.
Am J Clin Nutr
2010
;
92
:
1080
1088
.

252

Dupont
J
,
Dedeyne
L
,
Dalle
S
,
Koppo
K
,
Gielen
E
.
The role of omega-3 in the prevention and treatment of sarcopenia
.
Aging Clin Exp Res
2019
;
31
:
825
836
.

253

Mirmiran
P
,
Bahadoran
Z
,
Gaeini
Z
.
Common limitations and challenges of dietary clinical trials for translation into clinical practices
.
Int J Endocrinol Metab
2021
;
19
:
e108170
.

254

Cosmi
F
,
Shen
L
,
Magnoli
M
,
Abraham
WT
,
Anand
IS
,
Cleland
JG
,
Cohn
JN
,
Cosmi
D
,
De Berardis
G
,
Dickstein
K
,
Franzosi
MG
,
Gullestad
L
,
Jhund
PS
,
Kjekshus
J
,
Køber
L
,
Lepore
V
,
Lucisano
G
,
Maggioni
AP
,
Masson
S
,
McMurray
JJV
,
Nicolucci
A
,
Petrarolo
V
,
Robusto
F
,
Staszewsky
L
,
Tavazzi
L
,
Teli
R
,
Tognoni
G
,
Wikstrand
J
,
Latini
R
.
Treatment with insulin is associated with worse outcome in patients with chronic heart failure and diabetes
.
Eur J Heart Fail
2018
;
20
:
888
895
.

255

Bloise
FF
,
Cordeiro
A
,
Ortiga-Carvalho
TM
.
Role of thyroid hormone in skeletal muscle physiology
.
J Endocrinol
2018
;
236
:
R57
R68
.

256

Elkina
Y
,
von Haehling
S
,
Anker
SD
,
Springer
J
.
The role of myostatin in muscle wasting: an overview
.
J Cachexia Sarcopenia Muscle
2011
;
2
:
143
151
.

257

Conte
M
,
Martucci
M
,
Mosconi
G
,
Chiariello
A
,
Cappuccilli
M
,
Totti
V
,
Santoro
A
,
Franceschi
C
,
Salvioli
S
.
GDF15 plasma level is inversely associated with level of physical activity and correlates with markers of inflammation and muscle weakness
.
Front Immunol
2020
;
11
:
915
.

Author notes

Conflict of interest: R.S. reports grants from Japan Heart Foundation/Bayer Yakuhin Research Grant Abroad, during the conduct of the study. S.D.A. reports grants and personal fees from Vifor Int, personal fees from Bayer, personal fees from Boehringer Ingelheim, personal fees from Servier, grants and personal fees from Abbott Vascular, personal fees from Cardiac Dimensions, personal fees from Actimed, personal fees from Astra Zeneca, personal fees from Amgen, personal fees from Bioventrix, personal fees from Janssen, personal fees from Respicardia, personal fees from V-Wave, personal fees from Brahms, personal fees from Cordio, personal fees from Occlutech, outside the submitted work. S.v.H. has been a paid consultant for and/or received honoraria payments from AstraZeneca, Bayer, Boehringer Ingelheim, BRAHMS, Chugai, Grünenthal, Helsinn, Hexal, Novartis, Pharmacosmos, Respicardia, Roche, Servier, Sorin, and Vifor. S.v.H. reports research support from Amgen, Boehringer Ingelheim, IMI, and the German Center for Cardiovascular Research (DZHK).

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/pages/standard-publication-reuse-rights)