Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Mar 1.
Published in final edited form as: Heart Fail Rev. 2017 Mar;22(2):133–139. doi: 10.1007/s10741-016-9592-1

EXERCISE CAPACITY, PHYSICAL ACTIVITY AND MORBIDITY

Danielle L Brunjes 1, Peter J Kennel 1, P Christian Schulze 1
PMCID: PMC5352495  NIHMSID: NIHMS843990  PMID: 28091824

INTRODUCTION

By 2050, 20% of the US population will be over the age of 65 years, with muscle weakness and atrophy as the key characteristic traits for the aging adult (1). A similar trend can be seen in patients with heart failure (HF) with an accelerated level of muscle dysfunction (2). The absence of physical activity greatly increases muscle loss and decreases quality of life in both the aging and HF populations (3, 4). The purpose of this review is to provide an overview of the effects of aging and HF on skeletal muscle function and how exercise training can improve long-term outcomes associated with skeletal muscle dysfunction.

Effects of Aging on Skeletal Muscle

Sarcopenia, a geriatric syndrome (5), is an age-related loss of muscle mass associated with senescence and begins in the fourth decade of adult life, with an approximate loss of muscle mass of 0.4% per year while living a sedentary lifestyle (6). Sarcopenia has to be distinguished from cachexia, which describes the syndrome of loss of skeletal muscle with and underlying pathological correlate such as cancer cachexia or cardiac cachexia. Sarcopenia implies quantitative as well as qualitative changes in skeletal muscle with strength lost most rapidly, indicating derangements in skeletal muscle composition and functioning. In the aging muscle, changes in muscle fiber composition such as a change in the myosin heavy chain ratio can be observed with an increase of a co-expression of myosin heavy chain (MHC) I and MHC IIA in muscle fibers (7). Furthermore, metabolic derangements have been described in the sarcopenic muscle and have been linked to mitochondrial dysfunction (8). A recent study by Porter et al., comparing mitochondrial respiratory capacity in young versus elderly adults, found that mitochondrial respiratory capacity and coupling control declines with age. This indicates a decreased ATP production capacity in the aging muscle of elderly adults (9). However, while a large body of research supports the importance of mitochondrial senescence in sarcopenia, contradictory theories emphasize on the detrimental effects of inactivity for muscle homeostasis, attributing changes in muscle energy capacity to a sedentary lifestyle of the elderly (10). Recently, Johnson et al. compared the effects of endurance training on muscular mitochondrial health in the elderly (11). Interpreting changes of mitochondrial function on a transcriptional level, they concluded that in sarcopenia there is a decrease of mitochondrial genes and pathways involved in oxidative phosphorylation, which was compensated by endurance training in the older cohort compared to the sedentary cohort. Interestingly, however, physical activity had a more profound effect on the transcriptional profile in the younger cohort than in the elderly (11).

Skeletal muscle, which contributes 30–50% of the total body mass, influences a plethora of other organs and physiological processes (12) and muscle mass as well as strength can be correlated with longevity (13, 14). Skeletal muscle tissue is pivotal for insulin-mediated glucose turnover and has signaling function through cytokine secretion. Since insulin is a major anabolic stimulus for skeletal muscle homeostasis, age-related insulin resistance can contribute to the development of sarcopenia (15, 16). Of note, physically active older adults have better insulin sensitivity which coincides with a reduced rate of sarcopenia (17).

Another proposed mechanism contributing to sarcopenia are systemic inflammatory processes (18, 19). In a large study including over 900 elderly study participants, Schaap et al. could correlate higher circulating pro-inflammatory cytokine levels with loss of muscle strength (20). Proposed pathomechanisms describe the inhibitory effect of circulating cytokines such as TNFα and interleukins on insulin signaling (21, 22) and through the PI3K-Akt signaling pathway (23). It is important to note that physically active older adults have decreased levels of chronic inflammation compared to sedentary controls (24).

Effects of Heart Failure on Skeletal Muscle

While mortality from coronary artery disease, sudden cardiac death and other cardiovascular illnesses has declined, those that survive become predisposed to develop heart failure (HF) (25). Exercise intolerance is a common complaint expressed by HF patients and functional decline compounds HF risks (26, 27). As well-established in the literature (28), peak exercise capacity is not the only determinant of exercise performance in HF patients. Reduced cardiac output, inflammation, and insulin resistance are all contributing factors leading to skeletal muscle abnormalities seen in advanced HF (28). Patients with chronic HF subsequently have reduced physical activity, leading to low-grade systemic inflammation. Thus, these mechanisms promote further loss of muscle mass and exacerbation of local inflammation (29, 30). Skeletal muscle undergoes extensive changes in patients with HF, independent of tissue perfusion or hemodynamic instability, contributing to intrinsic muscle weakening and fatigability as well as to functional declines (3).

Phenotypic and metabolic changes in skeletal muscle of HF patients have been well described (3, 31). Changes in fiber composition include reduced type I oxidative fibers (slow twitch) with increases in less aerobic type II fibers (fast twitch) and altered myofibrillar protein isoform expression (with shifts in distribution from MHC I slow oxidative towards MHC IIA fast oxidative and MHC IIB fast glycolytic forms) (32). Histomorphometric analyses demonstrate decreased fiber sizes (both type I and II) as well as reduced number and altered structure of mitochondria (33, 34). Decreased levels of mitochondrial creatine kinase and downregulation of the sarcoplasmatic reticulum Ca2+-ATPase-1 (SERCA-1) are also seen, signifying a shift away from aerobic metabolism (35, 36). While progressive muscle wasting develops in HF, metabolic and functional derangements typically precede the loss of muscle mass, with subsequent muscle atrophy then exacerbating functional decline. HF skeletal myopathy is omnipresent, independent from disuse, refuting the hypothesis that skeletal muscle changes arise from disuse rather than direct HF effects (37).

Muscle protein breakdown is regulated by several cellular systems, including the ATP-dependent ubiquitin (Ub)-proteasome system (UPS). The UPS regulates skeletal muscle protein breakdown in a number of disease models including diabetes mellitus, cancer, renal failure, starvation (38) and HF (39). Through transcriptional profiling, a set of ~120 response genes, or atrogenes, has previously been identified in atrophying muscles (38). Dysregulation of atrogenes occurs in muscle atrophy, including genes required for ATP production and transcripts for extracellular matrix proteins. Further, protein quality control mechanisms in the cell are closely controlled by cellular autophagy and dysregulation of autophagy has been linked to abnormal protein turnover and overall cellular dysfunction (40) in the failing myocardium (41) and skeletal muscle (42). Suppression of PGC-1α, a central regulator of metabolism and mitochondrial biogenesis, was found in the setting of muscle atrophy (43). Induction of PGC-1α prevents muscle atrophy and transcription of atrogenes (44).

These changes in skeletal muscle lead to progressive muscle wasting and atrophy, causing further impairments in the exercise capacity of HF patients. Severe exercise intolerance is primarily due to reductions in peak cardiac output (CO) and peripheral arterial-venous oxygen difference (A-VO2 Diff) (26, 45), as well as a proposed impairment of skeletal muscle oxidative metabolism (46). HF therapeutic advances have centered on central cardiac pathophysiology, i.e., improving ventricular contractility, improving cardiac pre- and afterload and reducing remodeling and arrhythmias (47), which only yield marginal improvements in functional capacity. In contrast, therapies oriented towards skeletal muscle functional improvement (e.g. exercise training) have led to greater functional gains in patients with HF (31, 48).

Comparison of Skeletal Muscle Changes in HF and Normal Aging

Clinically, both sarcopenia and cachexia overlap and in principal, similar factors have been described to lead to the induction of muscle mass loss, with changes in muscle fiber composition along with mitochondrial derangements, insulin-resistance (or reduced sensitivity to other hormones) and chronic inflammation among them. The pathophysiological substrate of sarcopenia and cachexia is similar: Circulating proinflammatory cytokines, such as TNFα, IL-1 and others have been found to be pivotal especially in cachectic muscle wasting. The ill-fated combination of disease-related bed rest, decreased food intake and systemic inflammation caused by the underlying pathology such as a neoplasm or a chronic proinflammatory state in severe HF are thought to be the major underlying processes in cachexia.

However, in recent years, sophisticated studies focusing on protein turnover and anabolic-catabolic balances have unveiled pathomechanistic differences between these two syndromes. While cachexia is characterized by massive protein degradation, in sarcopenia, mainly a decrease in the rate of protein synthesis can be observed (49). In cachexia, protein degrading machineries such as the ubiquitin-proteasome system along with the suppression of the muscle anabolic PI3K-Akt-system, have been identified to be important mechanisms in this complex system of skeletal muscle turnover (50).

Age-related sarcopenia can also be linked to a decrease in regenerative capacity of injured muscle tissue. Satellite cells, which are characterized as myogenic stem cells are reduced and have a decreased regenerative potential (50, 51) in the aging muscle. Hence, repair mechanisms for cumulative muscle tissue injury decrease in the aging muscle with the myogenic stem cells likely converting from activatable quiescentic state to a senescence state (52).

Clinical Testing Methodology for Exercise Capacity

Regardless of how muscle mass is lost, sarcopenia or cachexia, there are a variety of methods used for measuring physical activity and exercise capacity. The gold standard method for measuring exercise capacity is the treadmill or bike exercise testing in association with air-gas-exchange, also known as cardiopulmonary exercise testing (CPET). This is considered to be an optimal gauge of functional capacity (53). Often older adults and HF subjects may perform incremental biking exercise on an electronically braked cycle ergometer as previously described by the American Thoracic Society (54), instead of on a treadmill due to issues with balance and stability on a treadmill. However, treadmill testing is recommended for optimal peak VO2 levels (55). Peak oxygen consumption (VO2peak), VO2 at anaerobic threshold, VO2/VCO2, and respiratory exchange ratio (RER) are measured using online computer-assisted open-circuit spirometry. Rate of perceived exertion, blood pressure and heart rate should be measured throughout exercise. Beyond a CPET, the six minute walk test is a commonly used clinical tool, which allows for the assessment of the functional capacity in HF patients in a “real-life” setting. Using standard methodology (56), patients will be asked to walk as quickly as possible on a 25 meter course for 6 minutes.

For the specific assessment of skeletal muscle function and fatigue, measures of isolated muscle group strength and fatigability can be analyzed using a dynamometer. The Biodex System 4 Pro can be used for measuring isometric and isokinetic measurements on the extremities. Isometric data will be collected to measure peak torque to body weight (PKTQ/BW). Isokinetic data will be collected to measure PKTQ/BW, work to body weight (WK/BW), average power, fatigability, time to peak torque, acceleration time and deceleration time. Muscle strength can be further analyzed through the use of hand grip strength (57), repeated three times on both dominant and non-dominant hands.

Obtaining skeletal muscle tissue provides researchers with information about local changes in skeletal muscle of older adults or patients with HF. This procedure should be done at least one week after the cardiopulmonary stress test or other strenuous activity. Aspirin should be held for two days (the day prior to the muscle biopsy and the day of the muscle biopsy) and study subjects should not be taking any blood thinning medication. The patients will lie comfortably in the supine position for biopsy of the vastus lateralis muscle. One fragment of the biopsied muscle should be frozen in liquid nitrogen cooled isopentane for histomorphological and enzymatic analysis. A second fragment can be cut and placed in cryotubes containing RNA preservation solution (RNAlater) and immediately frozen in liquid nitrogen (58).

In addition to muscle biopsies, a whole body dual-energy x-ray absorptiometry (DXA) scan characterizes the different compartments of soft tissue in the body. Analysis for specific compartments is only possible with an imaging method, which is beneficial because as stated earlier, both aged and patients with advanced HF develop metabolic abnormalities that include muscle wasting (1, 59).

Clinical Implications of Skeletal Muscle Dysfunction

Similarly to cachexia, sarcopenia, as a geriatric syndrome, but not attributable to an underlying disease, has important clinical implications. Sarcopenia has been defined as “a syndrome characterized by progressive and generalized loss of skeletal muscle mass and strength with a risk of adverse outcomes such as physical disability, poor quality of life and death” (60). Clinically relevant sarcopenia has been associated with mobility impairment (61) and increased mortality, especially in female patients (62) and in the very old, where it has been shown that sarcopenia is associated with mortality, independently of age or other clinical factors (63). Mobility impairment is a major risk in this context implying an increased risk of falls and fractures with hospitalization and immobilization. Sarcopenia has been termed a “societal burden” (64), reflecting on the consequential individual and socio-economical risk of physical disability, nursing home admission, depression, hospitalization and death (64).

Cachexia is a serious complication of many diseases and is associated with poor outcome. The diagnosis of cachexia against the background of an underlying disease is associated with a high mortality (65, 66). For example, cardiac cachexia mortality rates doubled in the cachectic cohort when compared to non-cachectic patients with the same underlying condition (67). After cachexia is diagnosed, yearly mortality rates are reported to range from 10% to 15% per year for COPD and 20% to 30% per year in HF (68). It is believed that several pathomechanisms occur in the cachectic patient that lead to the observed increase of mortality independently of the primary condition (69). Fatal consequences are cardiac arrhythmia, thromboembolism, compromised immune system and multi organ failure due to complex systemic derangements (69).

Unfortunately. treatment options for muscle wasting are limited with aerobic exercise being the best proven and most effective therapeutic approach (70). Many studies have been undertaken to find a pharmacological approach to tackle wasting syndromes, including appetite stimulants, anti-inflammatory agents or anabolic drugs. Promising candidates are the appetite stimulant megestrol acetate, anabolics such as the selective androgen receptor modulator enobosarm and the hormone ghrelin (71). Currently, larger clinical trials are under way to assess for the usefulness of these approaches to treat muscle wasting. Nutritional supplementation, another often studied approach to countervail muscle wasting, shows inconclusive results in many studies with a large variety of supplements (72).

Effects of Exercise Training

It is important to note that lower exercise capacity in heart failure and aging are associated with worse outcomes (73). Exercise training has been associated with reductions in both mortality and hospitalizations for HF patients with New York Heart Association class III or IV symptoms (74). Regardless of advanced age or disease state, exercise training is recommend for all age groups and patients with CVD, including HF by both the American Heart Association, American College of Cardiology Foundation and American College of Sports Medicine among other organizations (75). The Physical Activity Federal Guidelines recommend a minimum of 150 minutes per week of moderate aerobic exercise, but there are substantial evidence that shows benefits can be attained at much lower levels of weekly aerobic exercise (75, 76). There is a 14% reduction in mortality with just 15 minutes of exercise per day (77) and exercise training provides important improvements in health–related quality of life (78). Aerobic exercise training provides many beneficial improvements to the cardiovascular system, such as reduced resting HR (79), improved vascular endothelial function and parasympathetic tone, and myocardial improvements that result in improved tolerance for reperfusion injuries and ischemia (80). These improvements not only affect the cardiovascular system, but also the musculoskeletal system which aides in decreasing morbidity and increasing quality of life.

Continuous moderate intensity training has been the standard program choice for the elderly and patients with HF, but recent studies have demonstrated that high intensity interval training (HIIT) can be performed safely and provides greater improvements in VO2 peak and quality of life (81, 82). Wisloff et. al. showed that compared to moderate intensity training, patients with stable HF had a 46% increase in VO2max compared to a 14% increase in patients exercising at a moderate intensity (83). While HIIT training may show the greatest improvements for older adults and HF patients, not everyone is able to deal with its high demands and moderate aerobic intensity may be a better alternative with proven benefits (84).

Resistance training has become more popular over the last decade for both the elderly and patients with stable HF due to a decreased cardiac load requirement making resistance training well tolerated (84). Furthermore, the benefits of aerobic exercise training may not be enough to increase muscle mass and combat sarcopenia and cardiac cachexia (85). The use of resistance training in individual muscle groups has shown to improve. In a study by Esposito et al., knee extension training for 8 weeks in patients with HF showed total body improvements oxygen transport and metabolism (86).

SUMMARY and CONCLUSIONS

Sarcopenia and a decrease in quality of life are often associated with aging and HF, which can be attributed to the absence of physical activity. Exercise intolerance is a common problem for both the aging and HF population, and it is important to note that poor functional capacity increases HF risk. Throughout this review we have examined the skeletal physiological changes associated with aging and HF, as well as provide an overview of how exercise training can improve outcomes associated with skeletal muscle dysfunction. The benefits of exercise training are widely accepted for all ages and various disease states, and exercise programs and interventions have become the standard of care for patients with stable HF. Unfortunately, while the benefits of exercise training is shown in numerous studies for both aging adults and HF patients, the greatest setback is compliance.

References

  • 1.Miljkovic N, Lim JY, Miljkovic I, Frontera WR. Aging of skeletal muscle fibers. Annals of rehabilitation medicine. 2015;39(2):155–62. doi: 10.5535/arm.2015.39.2.155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Brassard P, Maltais F, Noel M, Doyon JF, LeBlanc P, Allaire J, et al. Skeletal muscle endurance and muscle metabolism in patients with chronic heart failure. The Canadian journal of cardiology. 2006;22(5):387–92. doi: 10.1016/s0828-282x(06)70923-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Duscha BD, Schulze PC, Robbins JL, Forman DE. Implications of chronic heart failure on peripheral vasculature and skeletal muscle before and after exercise training. Heart failure reviews. 2008;13(1):21–37. doi: 10.1007/s10741-007-9056-8. [DOI] [PubMed] [Google Scholar]
  • 4.Montero-Fernandez N, Serra-Rexach JA. Role of exercise on sarcopenia in the elderly. European journal of physical and rehabilitation medicine. 2013;49(1):131–43. [PubMed] [Google Scholar]
  • 5.Rolland Y, Abellan van Kan G, Gillette-Guyonnet S, Vellas B. Cachexia versus sarcopenia. Curr Opin Clin Nutr Metab Care. 2011;14(1):15–21. doi: 10.1097/MCO.0b013e328340c2c2. [DOI] [PubMed] [Google Scholar]
  • 6.Mitchell WK, Williams J, Atherton P, Larvin M, Lund J, Narici M. Sarcopenia, dynapenia, and the impact of advancing age on human skeletal muscle size and strength; a quantitative review. Front Physiol. 2012;3:260. doi: 10.3389/fphys.2012.00260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Andersen JL. Muscle fibre type adaptation in the elderly human muscle. Scand J Med Sci Sports. 2003;13(1):40–7. doi: 10.1034/j.1600-0838.2003.00299.x. [DOI] [PubMed] [Google Scholar]
  • 8.Boffoli D, Scacco SC, Vergari R, Solarino G, Santacroce G, Papa S. Decline with age of the respiratory chain activity in human skeletal muscle. Biochim Biophys Acta. 1994;1226(1):73–82. doi: 10.1016/0925-4439(94)90061-2. [DOI] [PubMed] [Google Scholar]
  • 9.Porter C, Hurren NM, Cotter MV, Bhattarai N, Reidy PT, Dillon EL, et al. Mitochondrial respiratory capacity and coupling control decline with age in human skeletal muscle. Am J Physiol Endocrinol Metab. 2015;309(3):E224–32. doi: 10.1152/ajpendo.00125.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Rasmussen UF, Krustrup P, Kjaer M, Rasmussen HN. Human skeletal muscle mitochondrial metabolism in youth and senescence: no signs of functional changes in ATP formation and mitochondrial oxidative capacity. Pflugers Arch. 2003;446(2):270–8. doi: 10.1007/s00424-003-1022-2. [DOI] [PubMed] [Google Scholar]
  • 11.Johnson ML, Lanza IR, Short DK, Asmann YW, Nair KS. Chronically endurance-trained individuals preserve skeletal muscle mitochondrial gene expression with age but differences within age groups remain. Physiol Rep. 2014;2(12) doi: 10.14814/phy2.12239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Demontis F, Piccirillo R, Goldberg AL, Perrimon N. The influence of skeletal muscle on systemic aging and lifespan. Aging Cell. 2013;12(6):943–9. doi: 10.1111/acel.12126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ruiz JR, Sui X, Lobelo F, Morrow JR, Jr, Jackson AW, Sjostrom M, et al. Association between muscular strength and mortality in men: prospective cohort study. BMJ. 2008;337:a439. doi: 10.1136/bmj.a439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Srikanthan P, Karlamangla AS. Muscle mass index as a predictor of longevity in older adults. Am J Med. 2014;127(6):547–53. doi: 10.1016/j.amjmed.2014.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Sharples AP, Hughes DC, Deane CS, Saini A, Selman C, Stewart CE. Longevity and skeletal muscle mass: the role of IGF signalling, the sirtuins, dietary restriction and protein intake. Aging Cell. 2015;14(4):511–23. doi: 10.1111/acel.12342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Rasmussen BB, Fujita S, Wolfe RR, Mittendorfer B, Roy M, Rowe VL, et al. Insulin resistance of muscle protein metabolism in aging. FASEB J. 2006;20(6):768–9. doi: 10.1096/fj.05-4607fje. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ryan AS. Exercise in aging: its important role in mortality, obesity and insulin resistance. Aging health. 2010;6(5):551–63. doi: 10.2217/ahe.10.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Perkisas S, Vandewoude M. Where frailty meets diabetes. Diabetes Metab Res Rev. 2015 doi: 10.1002/dmrr.2743. [DOI] [PubMed] [Google Scholar]
  • 19.Curtis E, Litwic A, Cooper C, Dennison E. Determinants of Muscle and Bone Aging. J Cell Physiol. 2015;230(11):2618–25. doi: 10.1002/jcp.25001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Schaap LA, Pluijm SM, Deeg DJ, Visser M. Inflammatory markers and loss of muscle mass (sarcopenia) and strength. Am J Med. 2006;119(6):526, e9–17. doi: 10.1016/j.amjmed.2005.10.049. [DOI] [PubMed] [Google Scholar]
  • 21.Budui SL, Rossi AP, Zamboni M. The pathogenetic bases of sarcopenia. Clin Cases Miner Bone Metab. 2015;12(1):22–6. doi: 10.11138/ccmbm/2015.12.1.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Borst SE. The role of TNF-alpha in insulin resistance. Endocrine. 2004;23(2–3):177–82. doi: 10.1385/ENDO:23:2-3:177. [DOI] [PubMed] [Google Scholar]
  • 23.Frost RA, Lang CH. Protein kinase B/Akt: a nexus of growth factor and cytokine signaling in determining muscle mass. J Appl Physiol (1985) 2007;103(1):378–87. doi: 10.1152/japplphysiol.00089.2007. [DOI] [PubMed] [Google Scholar]
  • 24.Santos-Parker JR, LaRocca TJ, Seals DR. Aerobic exercise and other healthy lifestyle factors that influence vascular aging. Advances in physiology education. 2014;38(4):296–307. doi: 10.1152/advan.00088.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Barker WH, Mullooly JP, Getchell W. Changing incidence and survival for heart failure in a well-defined older population, 1970–1974 and 1990–1994. Circulation. 2006;113(6):799–805. doi: 10.1161/CIRCULATIONAHA.104.492033. [DOI] [PubMed] [Google Scholar]
  • 26.Haykowsky MJ, Brubaker PH, Stewart KP, Morgan TM, Eggebeen J, Kitzman DW. Effect of endurance training on the determinants of peak exercise oxygen consumption in elderly patients with stable compensated heart failure and preserved ejection fraction. Journal of the American College of Cardiology. 2012;60(2):120–8. doi: 10.1016/j.jacc.2012.02.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Mancini DM, Walter G, Reichek N, Lenkinski R, McCully KK, Mullen JL, et al. Contribution of skeletal muscle atrophy to exercise intolerance and altered muscle metabolism in heart failure. Circulation. 1992;85(4):1364–73. doi: 10.1161/01.cir.85.4.1364. [DOI] [PubMed] [Google Scholar]
  • 28.Zizola C, Schulze PC. Metabolic and structural impairment of skeletal muscle in heart failure. Heart failure reviews. 2013;18(5):623–30. doi: 10.1007/s10741-012-9353-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ferrari R, Bachetti T, Confortini R, Opasich C, Febo O, Corti A, et al. Tumor necrosis factor soluble receptors in patients with various degrees of congestive heart failure. Circulation. 1995;92(6):1479–86. doi: 10.1161/01.cir.92.6.1479. [DOI] [PubMed] [Google Scholar]
  • 30.Testa M, Yeh M, Lee P, Fanelli R, Loperfido F, Berman JW, et al. Circulating levels of cytokines and their endogenous modulators in patients with mild to severe congestive heart failure due to coronary artery disease or hypertension. Journal of the American College of Cardiology. 1996;28(4):964–71. doi: 10.1016/s0735-1097(96)00268-9. [DOI] [PubMed] [Google Scholar]
  • 31.Clark AL, Poole-Wilson PA, Coats AJ. Exercise limitation in chronic heart failure: central role of the periphery. Journal of the American College of Cardiology. 1996;28(5):1092–102. doi: 10.1016/S0735-1097(96)00323-3. [DOI] [PubMed] [Google Scholar]
  • 32.Sullivan MJ, Duscha BD, Klitgaard H, Kraus WE, Cobb FR, Saltin B. Altered expression of myosin heavy chain in human skeletal muscle in chronic heart failure. Medicine and science in sports and exercise. 1997;29(7):860–6. doi: 10.1097/00005768-199707000-00004. [DOI] [PubMed] [Google Scholar]
  • 33.Drexler H, Riede U, Munzel T, Konig H, Funke E, Just H. Alterations of skeletal muscle in chronic heart failure. Circulation. 1992;85(5):1751–9. doi: 10.1161/01.cir.85.5.1751. [DOI] [PubMed] [Google Scholar]
  • 34.Hambrecht R, Fiehn E, Yu J, Niebauer J, Weigl C, Hilbrich L, et al. Effects of endurance training on mitochondrial ultrastructure and fiber type distribution in skeletal muscle of patients with stable chronic heart failure. Journal of the American College of Cardiology. 1997;29(5):1067–73. doi: 10.1016/s0735-1097(97)00015-6. [DOI] [PubMed] [Google Scholar]
  • 35.Simonini A, Chang K, Yue P, Long CS, Massie BM. Expression of skeletal muscle sarcoplasmic reticulum calcium-ATPase is reduced in rats with postinfarction heart failure. Heart. 1999;81(3):303–7. doi: 10.1136/hrt.81.3.303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Spangenburg EE, Lees SJ, Otis JS, Musch TI, Talmadge RJ, Williams JH. Effects of moderate heart failure and functional overload on rat plantaris muscle. J Appl Physiol (1985) 2002;92(1):18–24. doi: 10.1152/jappl.2002.92.1.18. [DOI] [PubMed] [Google Scholar]
  • 37.Simonini A, Long CS, Dudley GA, Yue P, McElhinny J, Massie BM. Heart failure in rats causes changes in skeletal muscle morphology and gene expression that are not explained by reduced activity. Circulation research. 1996;79(1):128–36. doi: 10.1161/01.res.79.1.128. [DOI] [PubMed] [Google Scholar]
  • 38.Lecker SH, Jagoe RT, Gilbert A, Gomes M, Baracos V, Bailey J, et al. Multiple types of skeletal muscle atrophy involve a common program of changes in gene expression. FASEB J. 2004;18(1):39–51. doi: 10.1096/fj.03-0610com. [DOI] [PubMed] [Google Scholar]
  • 39.Schulze PC, Fang J, Kassik KA, Gannon J, Cupesi M, MacGillivray C, et al. Transgenic overexpression of locally acting insulin-like growth factor-1 inhibits ubiquitin-mediated muscle atrophy in chronic left-ventricular dysfunction. Circulation research. 2005;97(5):418–26. doi: 10.1161/01.RES.0000179580.72375.c2. [DOI] [PubMed] [Google Scholar]
  • 40.Wang EY, Biala AK, Gordon JW, Kirshenbaum LA. Autophagy in the heart: too much of a good thing? Journal of cardiovascular pharmacology. 2012;60(2):110–7. doi: 10.1097/FJC.0b013e31824cc427. [DOI] [PubMed] [Google Scholar]
  • 41.Rifki OF, Hill JA. Cardiac autophagy: good with the bad. Journal of cardiovascular pharmacology. 2012;60(3):248–52. doi: 10.1097/FJC.0b013e3182646cb1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Carnio S, LoVerso F, Baraibar MA, Longa E, Khan MM, Maffei M, et al. Autophagy impairment in muscle induces neuromuscular junction degeneration and precocious aging. Cell reports. 2014;8(5):1509–21. doi: 10.1016/j.celrep.2014.07.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Sacheck JM, Hyatt JP, Raffaello A, Jagoe RT, Roy RR, Edgerton VR, et al. Rapid disuse and denervation atrophy involve transcriptional changes similar to those of muscle wasting during systemic diseases. FASEB J. 2007;21(1):140–55. doi: 10.1096/fj.06-6604com. [DOI] [PubMed] [Google Scholar]
  • 44.Sandri M, Lin J, Handschin C, Yang W, Arany ZP, Lecker SH, et al. PGC-1alpha protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(44):16260–5. doi: 10.1073/pnas.0607795103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Maeder MT, Thompson BR, Brunner-La Rocca HP, Kaye DM. Hemodynamic basis of exercise limitation in patients with heart failure and normal ejection fraction. Journal of the American College of Cardiology. 2010;56(11):855–63. doi: 10.1016/j.jacc.2010.04.040. [DOI] [PubMed] [Google Scholar]
  • 46.Bhella PS, Prasad A, Heinicke K, Hastings JL, Arbab-Zadeh A, Adams-Huet B, et al. Abnormal haemodynamic response to exercise in heart failure with preserved ejection fraction. European journal of heart failure. 2011;13(12):1296–304. doi: 10.1093/eurjhf/hfr133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jessup M, Brozena S. Heart failure. The New England journal of medicine. 2003;348(20):2007–18. doi: 10.1056/NEJMra021498. [DOI] [PubMed] [Google Scholar]
  • 48.Pina IL, Apstein CS, Balady GJ, Belardinelli R, Chaitman BR, Duscha BD, et al. Exercise and heart failure: A statement from the American Heart Association Committee on exercise, rehabilitation, and prevention. Circulation. 2003;107(8):1210–25. doi: 10.1161/01.cir.0000055013.92097.40. [DOI] [PubMed] [Google Scholar]
  • 49.Argiles JM, Busquets S, Felipe A, Lopez-Soriano FJ. Molecular mechanisms involved in muscle wasting in cancer and ageing: cachexia versus sarcopenia. Int J Biochem Cell Biol. 2005;37(5):1084–104. doi: 10.1016/j.biocel.2004.10.003. [DOI] [PubMed] [Google Scholar]
  • 50.Bowen TS, Schuler G, Adams V. Skeletal muscle wasting in cachexia and sarcopenia: molecular pathophysiology and impact of exercise training. J Cachexia Sarcopenia Muscle. 2015;6(3):197–207. doi: 10.1002/jcsm.12043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Alway SE, Myers MJ, Mohamed JS. Regulation of satellite cell function in sarcopenia. Front Aging Neurosci. 2014;6:246. doi: 10.3389/fnagi.2014.00246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Sousa-Victor P, Gutarra S, Garcia-Prat L, Rodriguez-Ubreva J, Ortet L, Ruiz-Bonilla V, et al. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature. 2014;506(7488):316–21. doi: 10.1038/nature13013. [DOI] [PubMed] [Google Scholar]
  • 53.Templeton DL, Kelly AS, Steinberger J, Dengel DR. Lower relative bone mineral content in obese adolescents: role of non-weight bearing exercise. Pediatric exercise science. 2010;22(4):557–68. doi: 10.1123/pes.22.4.557. [DOI] [PubMed] [Google Scholar]
  • 54.Society AT. Standards for the diagnosis and care of patients with chronic obstructive pulmonary disease. American Thoracic Society. American journal of respiratory and critical care medicine. 1995;152(5 Pt 2):S77–121. [PubMed] [Google Scholar]
  • 55.Kim S, Yamabe H, Yokoyama M. Hemodynamic characteristics during treadmill and bicycle exercise in chronic heart failure: mechanism for different responses of peak oxygen uptake. Japanese circulation journal. 1999;63(12):965–70. doi: 10.1253/jcj.63.965. [DOI] [PubMed] [Google Scholar]
  • 56.Bittner V, Weiner DH, Yusuf S, Rogers WJ, McIntyre KM, Bangdiwala SI, et al. Prediction of mortality and morbidity with a 6-minute walk test in patients with left ventricular dysfunction. SOLVD Investigators. Jama. 1993;270(14):1702–7. [PubMed] [Google Scholar]
  • 57.Chung CJ, Wu C, Jones M, Kato TS, Dam TT, Givens RC, et al. Reduced handgrip strength as a marker of frailty predicts clinical outcomes in patients with heart failure undergoing ventricular assist device placement. Journal of cardiac failure. 2014;20(5):310–5. doi: 10.1016/j.cardfail.2014.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Forman DE, Daniels KM, Cahalin LP, Zavin A, Allsup K, Cao P, et al. Analysis of Skeletal Muscle Gene Expression Patterns and the Impact of Functional Capacity in Patients with Systolic Heart Failure. Journal of cardiac failure. 2014 doi: 10.1016/j.cardfail.2014.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Zavin A, Daniels K, Arena R, Allsup K, Lazzari A, Joseph J, et al. Adiposity facilitates increased strength capacity in heart failure patients with reduced ejection fraction. International journal of cardiology. 2013;167(6):2468–71. doi: 10.1016/j.ijcard.2012.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, et al. Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing. 2010;39(4):412–23. doi: 10.1093/ageing/afq034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.McLean RR, Shardell MD, Alley DE, Cawthon PM, Fragala MS, Harris TB, et al. Criteria for clinically relevant weakness and low lean mass and their longitudinal association with incident mobility impairment and mortality: the foundation for the National Institutes of Health (FNIH) sarcopenia project. J Gerontol A Biol Sci Med Sci. 2014;69(5):576–83. doi: 10.1093/gerona/glu012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Batsis JA, Mackenzie TA, Barre LK, Lopez-Jimenez F, Bartels SJ. Sarcopenia, sarcopenic obesity and mortality in older adults: results from the National Health and Nutrition Examination Survey III. Eur J Clin Nutr. 2014;68(9):1001–7. doi: 10.1038/ejcn.2014.117. [DOI] [PubMed] [Google Scholar]
  • 63.Landi F, Cruz-Jentoft AJ, Liperoti R, Russo A, Giovannini S, Tosato M, et al. Sarcopenia and mortality risk in frail older persons aged 80 years and older: results from ilSIRENTE study. Age Ageing. 2013;42(2):203–9. doi: 10.1093/ageing/afs194. [DOI] [PubMed] [Google Scholar]
  • 64.Clark BC, Manini TM. Functional consequences of sarcopenia and dynapenia in the elderly. Curr Opin Clin Nutr Metab Care. 2010;13(3):271–6. doi: 10.1097/MCO.0b013e328337819e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Farkas J, von Haehling S, Kalantar-Zadeh K, Morley JE, Anker SD, Lainscak M. Cachexia as a major public health problem: frequent, costly, and deadly. J Cachexia Sarcopenia Muscle. 2013;4(3):173–8. doi: 10.1007/s13539-013-0105-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Anker SD, Ponikowski P, Varney S, Chua TP, Clark AL, Webb-Peploe KM, et al. Wasting as independent risk factor for mortality in chronic heart failure. Lancet. 1997;349(9058):1050–3. doi: 10.1016/S0140-6736(96)07015-8. [DOI] [PubMed] [Google Scholar]
  • 67.von Haehling S, Lainscak M, Springer J, Anker SD. Cardiac cachexia: a systematic overview. Pharmacol Ther. 2009;121(3):227–52. doi: 10.1016/j.pharmthera.2008.09.009. [DOI] [PubMed] [Google Scholar]
  • 68.von Haehling S, Anker SD. Cachexia as a major underestimated and unmet medical need: facts and numbers. J Cachexia Sarcopenia Muscle. 2010;1(1):1–5. doi: 10.1007/s13539-010-0002-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kalantar-Zadeh K, Rhee C, Sim JJ, Stenvinkel P, Anker SD, Kovesdy CP. Why cachexia kills: examining the causality of poor outcomes in wasting conditions. J Cachexia Sarcopenia Muscle. 2013;4(2):89–94. doi: 10.1007/s13539-013-0111-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Alves CR, da Cunha TF, da Paixao NA, Brum PC. Aerobic exercise training as therapy for cardiac and cancer cachexia. Life Sci. 2015;125:9–14. doi: 10.1016/j.lfs.2014.11.029. [DOI] [PubMed] [Google Scholar]
  • 71.von Haehling S, Anker SD. Treatment of cachexia: an overview of recent developments. J Am Med Dir Assoc. 2014;15(12):866–72. doi: 10.1016/j.jamda.2014.09.007. [DOI] [PubMed] [Google Scholar]
  • 72.Cruz-Jentoft AJ, Landi F, Schneider SM, Zuniga C, Arai H, Boirie Y, et al. Prevalence of and interventions for sarcopenia in ageing adults: a systematic review. Report of the International Sarcopenia Initiative (EWGSOP and IWGS) Age Ageing. 2014;43(6):748–59. doi: 10.1093/ageing/afu115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Forman DE, Clare R, Kitzman DW, Ellis SJ, Fleg JL, Chiara T, et al. Relationship of age and exercise performance in patients with heart failure: the HF-ACTION study. American heart journal. 2009;158(4 Suppl):S6–S15. doi: 10.1016/j.ahj.2009.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.O’Connor CM, Whellan DJ, Lee KL, Keteyian SJ, Cooper LS, Ellis SJ, et al. Efficacy and safety of exercise training in patients with chronic heart failure: HF-ACTION randomized controlled trial. Jama. 2009;301(14):1439–50. doi: 10.1001/jama.2009.454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lavie CJ, Arena R, Swift DL, Johannsen NM, Sui X, Lee DC, et al. Exercise and the cardiovascular system: clinical science and cardiovascular outcomes. Circulation research. 2015;117(2):207–19. doi: 10.1161/CIRCRESAHA.117.305205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Lee DC, Pate RR, Lavie CJ, Sui X, Church TS, Blair SN. Leisure-time running reduces all-cause and cardiovascular mortality risk. Journal of the American College of Cardiology. 2014;64(5):472–81. doi: 10.1016/j.jacc.2014.04.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Wen CP, Wai JP, Tsai MK, Yang YC, Cheng TY, Lee MC, et al. Minimum amount of physical activity for reduced mortality and extended life expectancy: a prospective cohort study. Lancet. 2011;378(9798):1244–53. doi: 10.1016/S0140-6736(11)60749-6. [DOI] [PubMed] [Google Scholar]
  • 78.Taylor RS, Sagar VA, Davies EJ, Briscoe S, Coats AJ, Dalal H, et al. Exercise-based rehabilitation for heart failure. The Cochrane database of systematic reviews. 2014;4:CD003331. doi: 10.1002/14651858.CD003331.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Bohm M, Swedberg K, Komajda M, Borer JS, Ford I, Dubost-Brama A, et al. Heart rate as a risk factor in chronic heart failure (SHIFT): the association between heart rate and outcomes in a randomised placebo-controlled trial. Lancet. 2010;376(9744):886–94. doi: 10.1016/S0140-6736(10)61259-7. [DOI] [PubMed] [Google Scholar]
  • 80.Gielen S, Laughlin MH, O’Conner C, Duncker DJ. Exercise training in patients with heart disease: review of beneficial effects and clinical recommendations. Progress in cardiovascular diseases. 2015;57(4):347–55. doi: 10.1016/j.pcad.2014.10.001. [DOI] [PubMed] [Google Scholar]
  • 81.Raymond MJ, Bramley-Tzerefos RE, Jeffs KJ, Winter A, Holland AE. Systematic review of high-intensity progressive resistance strength training of the lower limb compared with other intensities of strength training in older adults. Archives of physical medicine and rehabilitation. 2013;94(8):1458–72. doi: 10.1016/j.apmr.2013.02.022. [DOI] [PubMed] [Google Scholar]
  • 82.Arena R, Myers J, Forman DE, Lavie CJ, Guazzi M. Should high-intensity-aerobic interval training become the clinical standard in heart failure? Heart failure reviews. 2013;18(1):95–105. doi: 10.1007/s10741-012-9333-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Wisloff U, Stoylen A, Loennechen JP, Bruvold M, Rognmo O, Haram PM, et al. Superior cardiovascular effect of aerobic interval training versus moderate continuous training in heart failure patients: a randomized study. Circulation. 2007;115(24):3086–94. doi: 10.1161/CIRCULATIONAHA.106.675041. [DOI] [PubMed] [Google Scholar]
  • 84.Hirai DM, Musch TI, Poole DC. Exercise training in chronic heart failure: improving skeletal muscle O2 transport and utilization. Am J Physiol Heart Circ Physiol. 2015 doi: 10.1152/ajpheart.00469.2015. ajpheart 00469 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Coats AJ. Clinical utility of exercise training in chronic systolic heart failure. Nature reviews Cardiology. 2011;8(7):380–92. doi: 10.1038/nrcardio.2011.47. [DOI] [PubMed] [Google Scholar]
  • 86.Esposito F, Reese V, Shabetai R, Wagner PD, Richardson RS. Isolated quadriceps training increases maximal exercise capacity in chronic heart failure: the role of skeletal muscle convective and diffusive oxygen transport. Journal of the American College of Cardiology. 2011;58(13):1353–62. doi: 10.1016/j.jacc.2011.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES