Open access peer-reviewed chapter

Perspective Chapter: The Role of Modifiable Factors, Particularly Nutritional Factors, on Age-Related Sarcopenia

Written By

Nafiseh Shokri-Mashhadi

Submitted: 23 February 2022 Reviewed: 17 May 2022 Published: 21 June 2023

DOI: 10.5772/intechopen.105433

From the Edited Volume

Geriatric Medicine and Healthy Aging

Edited by Élvio Rúbio Gouveia, Bruna Raquel Gouveia, Adilson Marques and Andreas Ihle

Chapter metrics overview

65 Chapter Downloads

View Full Metrics

Abstract

Advances in medicine result in an increase in the age of global population. The percentage of people over 60 years will approximately be duplicated up to 22 between 2015 and 2050, which is associated with a notable rise in age-related complications such as sarcopenia and frailty. The age-related sarcopenia is defined by low muscle strength, and it is considered severe if low muscle strength, low muscle mass, and low physical performance are detected.This condition is associated with poor quality of life, risk of falls, fractures, and higher healthcare costs. Despite the growing interest regarding the treatment of this phenomenon, the lack of adequate knowledge underlying the multifactorial parthenogenesis of age-related sarcopenia hinders the diagnosis of effective therapeutic approaches. In this respect, one of the major solutions would be to recognize the effect of modifiable factors on muscle health during the lifetime. Previous observations indicated that dietary and nutritional factors, beyond other environmental agents across the life course are related to muscle mass and function in the elderly. With respect to the fundamental role of nutrients with antioxidants properties in maintaining many aspects of health, this chapter aims to discuss the association between components of sarcopenia and nutritional status in older adults, and their potential effect on prevention and treatment of age-related sarcopenia.

Keywords

  • age-related sarcopenia
  • nutritional factors
  • prevention
  • treatment
  • dietary quality

1. Introduction

1.1 Definition of sarcopenia

The average age of populations is increasing because of numerous factors, including advances in medical care and decreasing birth rate [1, 2]. The percentage of people over 60 years will approximately be duplicated up to 22 between 2015 and 2050, which is associated with a notable rise in age-related complications such as sarcopenia and frailty [3]. Definitions of age-related sarcopenia have evolved over time in an attempt to better characterize sarcopenia. The name for this phenomenon derives from the Greek term sarx (flesh) and penia (loss) [4]. Early definitions of sarcopenia were based exclusively on an age-related reduction in muscle mass [5]. However, the two-dimensional nature of these conditions (muscle mass loss and muscle strength loss) suggests that both its quantitative and qualitative range should be evaluated [6]. Therefore, the European Working Group on Sarcopenia in Older People (EWGSOP) described sarcopenia as an age-associated decline in muscle mass and strength with functional impairment [7].

Beyond the loss of muscle tissue that occurs over a lifetime, this condition is also associated with the conversion of type II fibers to type I fibers, which results in impairment of muscle quality and muscle function [8].

Categorizing sarcopenia into pre-sarcopenia, sarcopenia, and severe sarcopenia has also been defined by the EWGSOP that suggested the pre-sarcopenia stage as low muscle mass with no impact on muscle strength or physical performance, whereas the sarcopenia stage distinguished as low muscle mass with either low muscle strength or low physical performance and severe sarcopenia as the presence of all three criteria [9]. While interest in sarcopenia has risen in recent years, contention still exists over most components of the disease, with a universally accepted definition still lacking.

1.2 Diagnosis of sarcopenia

Various approaches can be used to assess muscle mass. Current assessment tools include body imaging techniques, bioelectric impedance analysis, anthropometric parameters, and biochemical markers [10].

Computed tomography (CT) and magnetic resonance imaging are able to effectively distinguish fat from other soft tissues, which makes these presently the gold standard method for the assessment of body composition. However, limited access, the high cost, and the risk of radiation inhibit the use of these techniques in clinical practice [11]. Therefore, dual-energy X-ray absorptiometry (DXA) is the most popular method for correctly evaluating body composition and widely used to assess muscle mass in research studies due to speed of measurement and relatively low per patient scan cost with typically low radiation [12]. Though, seeking for inexpensive, easy-to-use, and derived measures methods such as phase angle causes the application of bioimpedance technology. Nevertheless, using the estimation of body composition and muscle mass through anthropometric measurements, such as mid-upper arm circumference, calf circumference, and skinfold thickness, may allow us initially assess sarcopenia in situations that imaging equipment is typically unavailable in primary care settings [13].

The defining cutoff point for the identification of muscle loss depends upon the measurement technique chosen and the availability of reference studies. Low muscle mass is usually distinguished by a skeletal muscle mass index ranging from 7.23 kg/m2 to 8.87 kg/m2 in men and 5.45 kg/m2 to 6.42 kg/m2 in women [14]. Moreover, it is generally accepted that low physical performance is defined as a gait speed of less than 0.8 m/sec [15] and low muscle strength is usually defined by handgrip strength of less than 30 kg for men and less than 20 kg for women [7]. Nevertheless, the quadriceps strength cut-off points of 18.0 kg for older men and 16.0 kg for older women proposed as a muscle strength measurement for sarcopenia diagnosis in older Asian people [16].

Because of this diversity in the cutoffs of the sarcopenia’s characterization, EWGSOP has recommended that more research is urgently needed in order to obtain accurate reference values for different nations and countries around the world [17].

1.3 Pathophysiology of and risk factors for sarcopenia

The pathophysiology of sarcopenia is multifactorial. Several underlying mechanisms have been linked to the development of sarcopenia, although not all have been fully elucidated [6]. Prevalence of sarcopenia is mostly associated with chronic inflammation, which may lead to a vicious cycle of intricate interactions among risk factors [18]. However, research on sarcopenia prevention and treatment is developing quickly because of insufficient evidence for the underlying cellular mechanisms of the progress and maintenance of sarcopenia. So, it seems that understanding the factors related to increasing sarcopenia risk may provide strategies for intervention and disease improvement.

Inflammation in aging is one of the main suggested factors of sarcopenia characterized by a chronic progressive increase in pro-inflammatory cytokines, and the reduced serum level of anti-inflammatory cytokines [17]. Decline in immune function, plays an important role in several age-related diseases, for example Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, atherosclerosis, and other complications [19, 20].

The majority of studies have demonstrated that inflammatory cytokines have an important effect on skeletal muscle wasting, leading to an imbalance between protein synthesis and catabolism [21]. It reasonably has shown that reduced rates of protein synthesis paralleled to increased protein breakdown in the skeletal muscle are associated with a variety of produced pro-inflammatory cytokines during the inflammatory response. Indeed, the effects of pro-inflammatory cytokines on muscle mass may be mediated by activating the transcription factor NF-κB in line with a production of ROS in the muscle of the elderly people [22, 23]. Additionally, we recently indicated that circulating levels of C-reactive protein (CRP) and hs-CRP are independently associated with impairment of muscle strength [24]. It is also suggested that low muscle strength is associated with the high levels of inflammatory cytokines [25] such as CRP [26]. These findings may suggest that the plasma concentration of some inflammatory molecules is related to the aspects of muscle decline and functional impairment.

Another suggested factor is Insulin resistance (IR) which is defined by reduced peripheral glucose utilization in skeletal muscle, majority of whole-body insulin-stimulated glucose disposal, that develops with age [27]. Various studies demonstrated that IR is related markedly to the different diagnostic components of sarcopenia [28]. Data on the prevalence of sarcopenia in Korean elderly men aged more than 65 years recommended that higher IR and lower vitamin D levels are independently associated with the presence of sarcopenia in community-dwelling elderly men [29]. In another study conducted by Gorshunova et al., low muscle mass and muscle strength were significantly related to increased indices of IR, probably as a result of energy homeostasis disorders and the deterioration of glucose in the skeletal muscles [30]. One of the anticipated mechanisms of insulin resistance in elderly people is a reduction in the size of type II fibers which may reduce mitochondrial activity and result in IR in muscle [31, 32].

On the other hand, recent investigations illustrated that serum level of negative regulator of muscle growth, such as myostatin could increase with advancing age [33] and may play an important role in the resistance of insulin in muscles [34]. Moreover, aging skeletal muscle inflammation through activation of the classical signaling pathway also has impact on insulin uptake [35]. Furthermore, the effect of accumulation of intramyocellular lipid has been systematically evaluated and reported a well-established association between accumulation of intramyocellular lipid and muscle IR [30]. Finally, it is understood that strategies for identifying improvements and insulin sensitivity treatments can propose possible preventive measures against sarcopenia.

Aging is also related to changes in a several hormones status, including testosterone, estrogen, growth hormone, insulin-like growth factor 1, and corticosteroids [36], and the clinical significance of these deficiencies is variable with age [37]. It is previously supposed that the age-dependent decline in GH and IGF-1 levels is related to the pathogenesis of sarcopenia [38]. Moreover, in a recent observational cohort study, low baseline serum IGF-1 levels correlated with lower handgrip strength and worse physical performance [39]. Nevertheless, the impact of long-term GH therapy in the treatment of sarcopenia in elderly individuals with low GH/IGF-1 levels is still unclear. Cortisol is also the most potent immunosuppressive agent which can be stimulated by inflammation and therefore can be related to the development of sarcopenia and its components; muscle strength, muscle mass, and physical function. We could speculate from studies that the systemic overproduction of glucocorticoids during aging is associated with an increase of sarcopenia risk. However, further longitudinal studies are required to confirm these relationships.

In terms of modifiable risk factors, the relationship between adult lifestyle and sarcopenia has been highlighted in order to provide strategies for prevention of and improvement in age-related sarcopenia. In this concern, mental state, smoking, low body mass index (BMI), nutritional status, and physical activities have been introduced as the most potential changeable factors that could be applied in future strategies to prevent or delay the progression of sarcopenia [40, 41, 42]. The strong association between alteration in body composition in lifetime, namely fat-free mass, skeletal muscle mass, and BMI with prevalence and incidence of sarcopenia has been shown [43]. In addition, various previous reviews of studies revealed the relationship between physical inactivity and losses of muscle mass and strength [40], while resistance training was reported to have a beneficial effect on the physical performance measures in most studies [44, 45]. Regarding mental health, several studies found that sarcopenia is associated with cognitive decline and depression which could be due to some of the predisposing factors underlying sarcopenia, such as oxidative stress, inflammation, and insulin resistance [46]. Among them, one of the important modifiable factors in maintaining healthy status, in helping recovery from acute conditions, and in prevention of chronic diseases across lifespan is optimal nutritional status [47]. On the other hand, poor nutritional status is associated with several adverse consequences in community-dwelling older individuals, such as inflammation, cachexia, altered gut integrity, and muscle dysfunction [48, 49]. In addition, it is shown that the quality of the diet along with the lifetime has a close relationship with the sarcopenia [50]. It has been demonstrated that individuals consuming less energy will lose more muscle tissue. Therefore, avoiding under-nutrition is required to prevent muscle loss [47, 51]. Many nutrients have also been linked with the development of sarcopenia [47]. In this regard, the Korea National Health and Nutrition Examination Survey (KNHANES) cohort has revealed a lower energy, protein, and carbohydrate intake among sarcopenic older adults [52]. Given that evaluating the role of dietary nutrient intake in the treatment and development of sarcopenia would be valuable. Following we summarized studies in which the role of various nutritional factors on age-related sarcopenia was evaluated.

Advertisement

2. Role of nutritional factors in prevention and treatment of sarcopenia

2.1 Role of dietary protein in prevention and treatment of sarcopenia

The scientific literature indicated that the amount of daily protein intake is related to prevention of muscle functional decline and sarcopenia treatment, as older adults have an increased need for dietary protein to stimulate their muscle protein synthesis [53, 54]. Furthermore, Sovianne et al., showed a significant difference in protein intake (gram/day) among sarcopenic and non-sarcopenic older adults [55]. It is supposed that lower energy requirements and reduced appetite decrease total energy consumption in aging, which can significantly reduce dietary intake of protein [56]. It proposed that dietary protein supplementation more than recommended dietary intake (above 0.8 g/kg/d) may have beneficial effect on sarcopenia [57, 58]. However, other recent studies have shown that muscle loss cannot be entirely stopped, even when daily protein intake is maintained at a high level [59]. So, the role of protein supplementation on age-related sarcopenia is somewhat controversial and might be affected by the nutritional status of individuals [60].

It is also worth noting that the type of protein ingested and timing of protein intake throughout the day could determine the amount of skeletal muscle mass [61]. In these regards,nutritional supplementation including 20 g whey protein and 800 IU vitamin D in previous randomized double blind research leads to further losses of intermuscular fat (p = .049) and increased normal muscle density (p = .018) after the 6-month intervention [62]. These results were also confirmed by another study [63]. However, a recent meta-analysis of eight studies (n = 557) conducted by Tieland et al., showed no significant positive effects of protein or amino acid supplementation on lean body mass, muscle strength, or handgrip. Nevertheless, it seems that there were various heterogeneities in included trials in the mentioned study, such as duration of treatment and type of supplementation, and dosage which could affect the result interpretation [64]. So, it can be concluded that this general lack of effect of protein supplements directed us to assess positive associations between protein intake and muscle mass and function through cohort studies.

The anabolic effects of some amino acids on muscle mass also have been investigated in several studies. Creatine, as one of the most important amino acids located primarily in muscle tissue, accelerates muscle ATP regeneration throughout the increased energy demand [65, 66]. In this aera, the increased muscle mass and muscle strength with exercises and an additional effect of creatine were found in clinical trials [67]. Moreover, increased physical performance with exercises and an interactive effect of creatine were observed in some studies [68]. Results of a previous meta-analyses specified that creatine supplementation combined with resistance training could have a positive effect on aging muscle mass and upper body strength compared to resistance training alone [69]. In agreement with these findings, the recent meta-analyses also showed that creatine supplementation leads to greater increases in muscle mass (SMD: 1.37 kg [95% CI = 0.97–1.76]) and leg muscle strength (SMD: 0.24 kg [95% CI = 0.05–0.43]) in participants with a mean age of 57–70 years [70]. The molecular mechanisms underlying the improved protein synthesis and muscle strength following creatine administration might be correlated with an increase in skeletal muscle phosphocreatine content and enhanced muscle glycogen storage through exercise [67]. Despite these promising results, it is worth to mention that the vast majority of these studies measured the impact of combined exercise interventions and creatine supplementation in the sarcopenic populations suffering from malnutrition [69].

2.2 Role of n-3 fatty acids in prevention and treatment of sarcopenia

Many nutrients also have anabolic effects on aging musculoskeletal health. There is growing evidence for an association between n-3 fatty acids intake alone or in combination of other nutrients and components of sarcopenia, including muscle mass, muscle strength, and physical performance [71, 72]. In this regard, the investigation of possible relationship between circulating n-3 FA levels and sarcopenia among 125 participants in Asian older adults indicated that subjects with low muscle strength had 32.4% lower serum n-3 concentrations (P = 0.030) than controls [73]. In addition, omega-3 fatty acids intake was lower in elderly sarcopenic patients than elderly subjects without sarcopenia [2.6 ± 1.0 vs. 3.0 ± 1.2 kcal/day, p = 0.046] [74].

Furthermore, the recent results of the Maastricht Sarcopenia Study also showed that sarcopenic older adults had a 10–18% lower intake of five nutrients (n-3 fatty acids, vitamin B6, folic acid, vitamin E, magnesium) compared with non-sarcopenic older adults (P < .05) [55]. Similarly, prolonged supplementation with omega-3 fatty acids has been examined in older adults in order to improve the muscle protein synthetic response [75, 76], and importantly, the combined supplements providing high-quality proteins, leucine, vitamin D, and omega-3 fatty acids all together appear to be most favorable effects in the prevention of sarcopenia, while also being safe [77]. The positive effects of omega-3 fatty acids supplementation on muscle mass and function have also been identified by a systematic review and meta-analysis [72]. In addition, some studies specified that vitamin D supplementation combined with n-3 fatty acids, in particular EPA + DHA, may have favorable effect on physical function, muscle mass, and strength [77]. It is supposed that the anti-inflammatory effects of omega-3 fatty acids have an important role in the reduction in sarcopenia risk [78]. Nevertheless, the exact mechanisms by which n-3 fatty acids apply their beneficial effects on components of sarcopenia remain to be elucidated.

2.3 Role of vitamin D in prevention and treatment of sarcopenia

Among several nutrients, there has been increasing interest in the implications of vitamin D, either as single supplements or in combination with other supplements, for improving the physical function of older adults due to high prevalence of vitamin D deficiency [79]. A positive correlation between serum 25(OH)D concentration and muscle function has been shown [80]. The previous systematic reviews aimed at examining the benefits of vitamin D supplementation on sarcopenia in aging indicated the importance of considering baseline serum 25(OH)D concentrations in the response to supplementation [81]. While, concerning this issue, the recent meta-analysis of RCTs (2017) confirmed a slight improvement in the physical performance test following supplementation, no overall increase in handgrip strength was detected [82]. Nevertheless, this finding strengthens that vitamin D with a range of 800–1000 IU/day, but not necessarily at higher doses, has beneficial effect on muscle strength [77].

2.4 Role of gut microbiota in prevention and treatment of sarcopenia

Epidemiologic studies point out that altered gut microbiota structure according to diet, taking drugs, and other environmental factors across the lifespan result in different microbiota patterns, composition, and function in the elderly [83, 84]. The gut microbiota has the essential function maintaining some aspects of health [85]. Changing microbiota patterns are associated with significant changes in metabolic and physiologic regulation, and markers of inflammation that could result in age-related adverse health consequences [86, 87]. Recent researchers have postulated that gut microbiota composition may have a great relationship with age-related alterations in skeletal muscle function [87]. In this concern, experimental studies revealed that changes in the gut composition via probiotic/prebiotic administration could influence muscle function and inflammatory status [88, 89, 90]. Our recent meta-analysis emphasized that probiotic supplementation for more than 12 weeks has positive impact on the muscle strength. However, the clear mechanism underlying the positive effect of probiotic administration on muscle strength was not identified. The possible explanation for these findings may be related to reduced levels of IGF-1 (insulin-like growth factor 1) during lifetime [39, 91]. The beneficial probiotic effects on circulating inflammatory biomarkers could be another description [92].

2.5 Role of dietary quality in prevention of sarcopenia

Research on dietary quality and dietary patterns has recently been undertaken to better understand the effects of diet as a whole and its impact on aging health complication such as sarcopenia. Results of recent cross-sectional study in 250 menopausal women 45 years old or older found that Mediterranean dietary pattern has a favorable role in the prevention of sarcopenia [93]. Additionally, adherence to dietary pattern including “vegetables-fruits” was associated with lower odds of prevalent sarcopenia in Chinese older men [94]. Furthermore, it is shown that subjects with higher consumption of “snacks-drinks-milk products” score had lower odds of sarcopenia (OR = 0.41, 95% CI: 0.24–0.70, Ptrend < 0.001) [94]. Interestingly, findings of recent cross-sectional study revealed that adherence to the Western dietary pattern, characterized by a high intake of sugar, soy, and fast foods, was not linked to sarcopenia (OR = 0.51; 95% CI: 0.21–1.24; Ptrend = 0.13) [95]. The link between healthier diets and physical performance among older adults has been proposed [41]. However, there is not enough research evidence available in various communities to inform about the definitive decision on the specific food pattern for the prevention or treatment of sarcopenia.

Advertisement

3. Conclusion

Age-related sarcopenia is a phenomenon with significant disability among the elderly. This condition is one of the most important public health problems among the healthy community. While there are the various notable bodies of research in order to define sarcopenia, the diversity in the cutoffs of the sarcopenia’s characterization revealed the needs for more research to define accurate reference values in different nations and countries around the world. On the other hand, one of the main controversial topics in evaluating the influence of nutrition on age-related sarcopenia is related to various definitions of sarcopenia in different nations and countries. In addition, due to the lack of a valid biomarker for the detection of sarcopenia, the exact mechanism underlying beneficial effect of numerous nutritional factors on components of sarcopenia remains unknown. Furthermore, there is not enough research evidence available in various communities to inform about the definitive decision on the food for the prevention or treatment of sarcopenia. The other limitation of included studies in this review was connected to absence of information about dietary intakes and serum concentrations of many micronutrients in older adults. So, considering the biochemical levels and dietary intakes of micronutrients in future studies is recommended. Finally, further studies are needed to investigate the interaction effect of modifiable risk factors, in particularly nutrition over time based in the near future.

Overall, present findings from the scientific literature specified that the combined nutritional supplements, such as vitamin D, n-3 fatty acids, and creatine along with resistance training could have better improvement in aging muscle mass and upper body strength compared with each alone. This effect particularly was shown in subjects with low serum vitamin D concentrations. In addition, it seems that the beneficial effect of probiotic/prebiotic administration also depends on changed gut microbiota composition. It is worth to mention that adherence to healthy dietary pattern with high quality including vegetables and fruits may lead to lower odds of sarcopenia.

Advertisement

Conflict of interest

None.

References

  1. 1. Burch JB et al. Advances in geroscience: Impact on healthspan and chronic disease. Journals of Gerontology Series A: Biomedical Sciences and Medical Sciences. 2014;69(Suppl_1):S1-S3
  2. 2. Nargund G. Declining birth rate in Developed Countries: A radical policy re-think is required. Facts, Views & Vision in ObGyn. 2009;1(3):191-193
  3. 3. Castrejón-Pérez RC et al. Frailty, diabetes, and the convergence of chronic disease in an age-related condition: A population-based nationwide cross-sectional analysis of the Mexican nutrition and health survey. Aging Clinical and Experimental Research. 2018;30(8):935-941
  4. 4. Rosenberg IH. Sarcopenia: Origins and clinical relevance. The Journal of Nutrition. 1997;127(5):990S-991S
  5. 5. Rosenberg IH. Summary comments. The American Journal of Clinical Nutrition. 1989;50(5):1231-1233
  6. 6. Reiss J et al. Consequences of applying the new EWGSOP2 guideline instead of the former EWGSOP guideline for sarcopenia case finding in older patients. Age and Ageing. 2019;48(5):719-724
  7. 7. Cruz-Jentoft AJ et al. Sarcopenia: European consensus on definition and diagnosisReport of the European Working Group on Sarcopenia in Older PeopleA. J. Cruz-Gentoft et al. Age and Ageing. 2010;39(4):412-423
  8. 8. Lang T et al. Sarcopenia: Etiology, clinical consequences, intervention, and assessment. Osteoporosis International. 2010;21(4):543-559
  9. 9. Cruz-Jentoft A. European Working Group on Sarcopenia in Older People: Sarcopenia: European consensus on definition and diagnosis. Report of the European Working Group on Sarcopenia in Older People. Age and Ageing. 2010;39:412-423
  10. 10. Tosato M et al. Measurement of muscle mass in sarcopenia: From imaging to biochemical markers. Aging Clinical and Experimental Research. 2017;29(1):19-27
  11. 11. Ida S et al. Sarcopenia is a predictor of postoperative respiratory complications in patients with esophageal cancer. Annals of Surgical Oncology. 2015;22(13):4432-4437
  12. 12. Heymsfield SB et al. Assessing skeletal muscle mass: Historical overview and state of the art. Journal of Cachexia, Sarcopenia and Muscle. 2014;5(1):9-18
  13. 13. Malafarina V et al. Sarcopenia in the elderly: Diagnosis, physiopathology and treatment. Maturitas. 2012;71(2):109-114
  14. 14. Baumgartner RN et al. Predictors of skeletal muscle mass in elderly men and women. Mechanisms of Ageing and Development. 1999;107(2):123-136
  15. 15. Newman AB et al. Strength and muscle quality in a well-functioning cohort of older adults: The Health, Aging and Body Composition Study. Journal of the American Geriatrics Society. 2003;51(3):323-330
  16. 16. Assantachai P et al. Cut-off points of quadriceps strength, declines and relationships of sarcopenia-related variables among T hai community-dwelling older adults. Geriatrics & Gerontology International. 2014;14:61-68
  17. 17. Wilson D et al. Frailty and sarcopenia: The potential role of an aged immune system. Ageing Research Reviews. 2017;36:1-10
  18. 18. Ferrucci L, Fabbri E. Inflammageing: Chronic inflammation in ageing, cardiovascular disease, and frailty. Nature Reviews Cardiology. 2018;15(9):1
  19. 19. Ilia S. Stop Aging Disease! ICAD 2014. Aging and Disease. 2015;6(2):76
  20. 20. Deleidi M, Jäggle M, Rubino G. Immune aging, dysmetabolism, and inflammation in neurological diseases. Frontiers in Neuroscience. 2015;9:172
  21. 21. Londhe P, Guttridge DC. Inflammation induced loss of skeletal muscle. Bone. 2015;80:131-142
  22. 22. Bar-Shai M et al. The effect of hindlimb immobilization on acid phosphatase, metalloproteinases and nuclear factor-κB in muscles of young and old rats. Mechanisms of Ageing and Development. 2005;126(2):289-297
  23. 23. Lin H et al. Whole blood gene expression and interleukin-6 levels. Genomics. 2014;104(6):490-495
  24. 24. Paolisso G et al. Advancing age and insulin resistance: Role of plasma tumor necrosis factor-α. American Journal of Physiology-Endocrinology And Metabolism. 1998;275(2):E294-E299
  25. 25. Shokri-Mashhadi N et al. Association of circulating C-reactive protein and high-sensitivity C-reactive protein with components of sarcopenia: A systematic review and meta-analysis of observational studies. Experimental Gerontology. 2021;150:111330
  26. 26. Hida T et al. Sarcopenia and physical function are associated with inflammation and arteriosclerosis in community-dwelling people: The Yakumo study. Modern Rheumatology. 2018;28(2):345-350
  27. 27. Ryan AS. Insulin resistance with aging. Sports Medicine. 2000;30(5):327-346
  28. 28. Cleasby ME, Jamieson PM, Atherton PJ. Insulin resistance and sarcopenia: Mechanistic links between common co-morbidities. Journal of Endocrinology. 2016;229(2):R67-R81
  29. 29. Kim S-H, Kwon HS, Hwang H-J. White blood cell counts, insulin resistance, vitamin D levels and sarcopenia in Korean elderly men. Scandinavian Journal of Clinical and Laboratory Investigation. 2017;77(3):228-233
  30. 30. Gorshunova N, Medvedev N. Significance of insulin resistance in the pathogenesis of sarcopenia and chronic heart failure in elderly hypertensive patients. Advances in Gerontology—Uspekhi Gerontologii. 2016;29(2):324-328
  31. 31. Johannsen DL et al. Ectopic lipid accumulation and reduced glucose tolerance in elderly adults are accompanied by altered skeletal muscle mitochondrial activity. The Journal of Clinical Endocrinology & Metabolism. 2012;97(1):242-250
  32. 32. Stuart CA et al. Slow-twitch fiber proportion in skeletal muscle correlates with insulin responsiveness. The Journal of Clinical Endocrinology & Metabolism. 2013;98(5):2027-2036
  33. 33. Yarasheski K et al. Serum myostatin-immunoreactive protein is increased in 60-92 year old women and men with muscle wasting. Journal of Nutrition Health and Aging. 2002;6(5):343-348
  34. 34. Bonala S et al. Myostatin induces insulin resistance via Casitas B-lineage lymphoma b (Cblb)-mediated degradation of insulin receptor substrate 1 (IRS1) in response to high calorie diet intake. The Journal of Biological Chemistry. 2016;291(27):14392
  35. 35. Dagdeviren S et al. IL-10 prevents aging-associated inflammation and insulin resistance in skeletal muscle. The FASEB Journal. 2016;31(2):701-710
  36. 36. Vitale G, Cesari M, Mari D. Aging of the endocrine system and its potential impact on sarcopenia. European Journal of Internal Medicine. 2016;35:10-15
  37. 37. Chahal H, Drake W. The endocrine system and ageing. The Journal of Pathology: A Journal of the Pathological Society of Great Britain and Ireland. 2007;211(2):173-180
  38. 38. Sattler FR. Growth hormone in the aging male. Best Practice & Research Clinical Endocrinology & Metabolism. 2013;27(4):541-555
  39. 39. Van Nieuwpoort C et al. The relationship between serum IGF-1, handgrip strength, physical performance and falls in elderly men and women. European Journal of Endocrinology. 2018;179(2):EJE-18-0076
  40. 40. Bosaeus I, Rothenberg E. Nutrition and physical activity for the prevention and treatment of age-related sarcopenia. Proceedings of the Nutrition Society. 2015;75(2):174-180
  41. 41. Bloom I et al. Diet quality and sarcopenia in older adults: A systematic review. Nutrients. 2018;10(3):308
  42. 42. Hayashi T et al. Association between sarcopenia and depressive mood in urban-dwelling older adults: A cross-sectional study. Geriatrics & Gerontology International. 2019;19(6):508-512
  43. 43. Gruber ES et al. Sarcopenia and sarcopenic obesity are independent adverse prognostic factors in resectable pancreatic ductal adenocarcinoma. PLoS One. 2019;14(5):e0215915
  44. 44. Fiatarone MA et al. The Boston FICSIT study: The effects of resistance training and nutritional supplementation on physical frailty in the oldest old. Journal of the American Geriatrics Society. 1993;41(3):333-337
  45. 45. De Jong N et al. Dietary supplements and physical exercise affecting bone and body composition in frail elderly persons. American Journal of Public Health. 2000;90(6):947
  46. 46. Chang K-V et al. Association between sarcopenia and cognitive impairment: A systematic review and meta-analysis. Journal of the American Medical Directors Association. 2016;17(12):1164. e7-1164. e15
  47. 47. Laviano A, Gori C, Rianda S. Sarcopenia and nutrition. Advances in Food and Nutrition Research. 2014;71:101-136
  48. 48. Walston JD. Sarcopenia in older adults. Current Opinion in Rheumatology. 2012;24(6):623
  49. 49. Agarwal E et al. Malnutrition in the elderly: A narrative review. Maturitas. 2013;76(4):296-302
  50. 50. Eglseer D, Eminovic S, Lohrmann C. Association between sarcopenia and nutritional status in older adults: A systematic literature review. Journal of Gerontological Nursing. 2016;42(7):33-41
  51. 51. Morley JE et al. Nutritional recommendations for the management of sarcopenia. Journal of the American Medical Directors Association. 2010;11(6):391-396
  52. 52. Seo MH et al. The association between daily calcium intake and sarcopenia in older, non-obese Korean adults: The fourth Korea National Health and Nutrition Examination Survey (KNHANES IV) 2009. Endocrine Journal. 2013;60(5):679-686
  53. 53. Bauer J et al. Evidence-based recommendations for optimal dietary protein intake in older people: A position paper from the PROT-AGE study group. Journal of the American Medical Directors Association. 2013;14(8):542-559
  54. 54. Paddon-Jones D et al. Role of dietary protein in the sarcopenia of aging. The American Journal of Clinical Nutrition. 2008;87(5):1562S-1566S
  55. 55. ter Borg S et al. Differences in nutrient intake and biochemical nutrient status between sarcopenic and nonsarcopenic older adults—results from the Maastricht Sarcopenia Study. Journal of the American Medical Directors Association. 2016;17(5):393-401
  56. 56. Pasiakos SM et al. Acute Energy Deprivation Affects Skeletal Muscle Protein Synthesis and Associated Intracellular Signaling Proteins in Physically Active Adults, 2. The Journal of Nutrition. 2010;140(4):745-751
  57. 57. Joint WHO. FAO/UNU expert consultation. Protein and amino acid requirements in human nutrition. World Health Organanization Technical Report Series. Vol. 935. 2007. pp. 1-265
  58. 58. Volpi E et al. Is the optimal level of protein intake for older adults greater than the recommended dietary allowance? Journals of Gerontology Series A: Biomedical Sciences and Medical Sciences. 2013;68(6):677-681
  59. 59. Trappe T et al. Influence of concurrent exercise or nutrition countermeasures on thigh and calf muscle size and function during 60 days of bed rest in women. Acta Physiologica. 2007;191(2):147-159
  60. 60. Beasley JM, Shikany JM, Thomson CA. The role of dietary protein intake in the prevention of sarcopenia of aging. Nutrition in Clinical Practice. 2013;28(6):684-690
  61. 61. van Loon LJ. Leucine as a pharmaconutrient in health and disease. Current Opinion in Clinical Nutrition & Metabolic Care. 2012;15(1):71-77
  62. 62. Englund DA et al. Nutritional supplementation with physical activity improves muscle composition in mobility-limited older adults, the VIVE2 study: A randomized, double-blind, placebo-controlled trial. The Journals of Gerontology: Series A. 2017;73(1):95-101
  63. 63. Wall BT, Van Loon LJ. Nutritional strategies to attenuate muscle disuse atrophy. Nutrition Reviews. 2013;71(4):195-208
  64. 64. Tieland M et al. The impact of dietary protein or amino acid supplementation on muscle mass and strength in elderly people: Individual participant data and meta-analysis of rct’s. The Journal of Nutrition, Health & Aging. 2017;21(9):994-1001
  65. 65. Greenhaff PL. The creatine-phosphocreatine system: There’s more than one song in its repertoire. The Journal of Physiology. 2001;537(Pt 3):657
  66. 66. Riesberg LA et al. Beyond muscles: The untapped potential of creatine. International Immunopharmacology. 2016;37:31-42
  67. 67. Tarnopolsky MA, Safdar A. The potential benefits of creatine and conjugated linoleic acid as adjuncts to resistance training in older adults. Applied Physiology, Nutrition, and Metabolism. 2008;33(1):213-227
  68. 68. Beaudart C et al. Nutrition and physical activity in the prevention and treatment of sarcopenia: Systematic review. Osteoporosis International. 2017;28(6):1817-1833
  69. 69. Candow DG, Chilibeck PD, Forbes SC. Creatine supplementation and aging musculoskeletal health. Endocrine. 2014;45(3):354-361
  70. 70. Chilibeck PD et al. Effect of creatine supplementation during resistance training on lean tissue mass and muscular strength in older adults: A meta-analysis. Open access Journal of Sports Medicine. 2017;8:213
  71. 71. Santiago EC et al. Comparison of calorie and nutrient intake among elderly with and without sarcopenia: A systematic review and meta-analysis. Nutrition Reviews. 2021;79(12):1338-1352
  72. 72. Bird JK et al. The effect of long chain omega-3 polyunsaturated fatty acids on muscle mass and function in sarcopenia: A scoping systematic review and meta-analysis. Clinical Nutrition ESPEN. 2021;46:73-86
  73. 73. Jang I-Y et al. Lower Serum n-3 Fatty Acid Level in Older Adults with Sarcopenia. Nutrients. 2020;12(10):2959
  74. 74. Okamura T et al. Reduced dietary omega-3 fatty acids intake is associated with sarcopenia in elderly patients with type 2 diabetes: A cross-sectional study of KAMOGAWA-DM cohort study. Journal of Clinical Biochemistry and Nutrition. 2020;66(3):19-85
  75. 75. Hutchins-Wiese H et al. The impact of supplemental n-3 long chain polyunsaturated fatty acids and dietary antioxidants on physical performance in postmenopausal women. The Journal of Nutrition, Health & Aging. 2013;17(1):76-80
  76. 76. Smith GI et al. Fish oil–derived n− 3 PUFA therapy increases muscle mass and function in healthy older adults. The American Journal of Clinical Nutrition. 2015;102(1):115-122
  77. 77. Tessier A-J, Chevalier S. An update on protein, leucine, omega-3 fatty acids, and vitamin D in the prevention and treatment of sarcopenia and functional decline. Nutrients. 2018;10(8):1099
  78. 78. Gray SR, Mittendorfer B. Fish oil-derived n-3 polyunsaturated fatty acids for the prevention and treatment of sarcopenia. Current Opinion in Clinical Nutrition & Metabolic Care. 2018;21(2):104-109
  79. 79. Lips P, van Schoor N. Worldwide Vitamin D Status, in Vitamin D (Third Edition). Amsterdam, Netherlands: Elsevier; 2011. pp. 947-963
  80. 80. Okazaki R et al. Assessment criteria for vitamin D deficiency/insufficiency in Japan: Proposal by an expert panel supported by the Research Program of Intractable Diseases, Ministry of Health, Labour and Welfare, Japan, the Japanese Society for Bone and Mineral Research and the Japan Endocrine Society [Opinion]. Journal of Bone and Mineral Metabolism. 2017;35(1):1-5
  81. 81. Annweiler C et al. Vitamin D-related changes in physical performance: A systematic review. The Journal of Nutrition, Health & Aging. 2009;13(10):893-898
  82. 82. Rosendahl-Riise H et al. Vitamin D supplementation and its influence on muscle strength and mobility in community-dwelling older persons: A systematic review and meta-analysis. Journal of Human Nutrition and Dietetics. 2017;30(1):3-15
  83. 83. Lakshminarayanan B et al. Compositional dynamics of the human intestinal microbiota with aging: Implications for health. The Journal of Nutrition, Health & Aging. 2014;18(9):773-786
  84. 84. Siddharth J et al. Aging and sarcopenia associate with specific interactions between gut microbes, serum biomarkers and host physiology in rats. Aging (Albany NY). 2017;9(7):1698
  85. 85. Tiihonen K, Ouwehand AC, Rautonen N. Human intestinal microbiota and healthy ageing. Ageing Research Reviews. 2010;9(2):107-116
  86. 86. Ticinesi A et al. Aging gut microbiota at the cross-road between nutrition, physical frailty, and sarcopenia: Is there a gut–muscle axis? Nutrients. 2017;9(12):1303
  87. 87. Ticinesi A et al. Gut microbiota, muscle mass and function in aging: A focus on physical frailty and sarcopenia. Nutrients. 2019;11(7):1633
  88. 88. Iolascon G et al. Pharmacological therapy of sarcopenia: Past, present and future. Clinical Cases in Mineral & Bone Metabolism. 2018;15(3):407-415
  89. 89. Ni Y et al. Lactobacillus and Bifidobacterium improves physiological function and cognitive ability in aged mice by the regulation of gut microbiota. Molecular Nutrition & Food Research. 2019;63(22):1900603
  90. 90. Lahiri S et al. The gut microbiota influences skeletal muscle mass and function in mice. Science Translational Medicine. 2019;11(502):eaan5662
  91. 91. Barbieri M et al. Chronic inflammation and the effect of IGF-I on muscle strength and power in older persons. American Journal of Physiology-Endocrinology and Metabolism. 2003;284(3):E481-E487
  92. 92. Kazemi A et al. Effect of probiotic and synbiotic supplementation on inflammatory markers in health and disease status: A systematic review and meta-analysis of clinical trials. Clinical Nutrition. 2020;39(3):789-819
  93. 93. Mohseni R et al. Relationship between major dietary patterns and sarcopenia among menopausal women. Aging Clinical and Experimental Research. 2017;29(6):1241-1248
  94. 94. Chan R, Leung J, Woo J. A prospective cohort study to examine the association between dietary patterns and sarcopenia in Chinese community-dwelling older people in Hong Kong. Journal of the American Medical Directors Association. 2016;17(4):336-342
  95. 95. Hashemi R et al. Diet and its relationship to sarcopenia in community dwelling Iranian elderly: A cross sectional study. Nutrition. 2015;31(1):97-104

Written By

Nafiseh Shokri-Mashhadi

Submitted: 23 February 2022 Reviewed: 17 May 2022 Published: 21 June 2023