Sarcopenia, frailty and type 2 diabetes mellitus (Review)

  • Authors:
    • Hiroki Nishikawa
    • Shinya Fukunishi
    • Akira Asai
    • Keisuke Yokohama
    • Hideko Ohama
    • Shuhei Nishiguchi
    • Kazuhide Higuchi
  • View Affiliations

  • Published online on: October 14, 2021     https://doi.org/10.3892/mmr.2021.12494
  • Article Number: 854
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Skeletal muscle is the largest and most energy‑consuming organ in the human body, which plays an important role in energy metabolism and glucose uptake. There is a notable decrease in glucose uptake in the skeletal muscle of patients with type 2 diabetes mellitus (DM). Endurance exercise can reduce hyperglycemia and improve insulin resistance in patients with type 2 DM. Insulin exerts a variety of effects, many of which are mediated by Akt, including increasing glucose uptake, promoting glycogen synthesis and inhibiting glycogen degradation, increasing free fatty acid uptake, increasing protein synthesis, promoting muscle hypertrophy and inhibiting protein degradation. Skeletal muscle mass progressively declines with aging, resulting in loss of muscle strength and physical function. Sarcopenia is a syndrome characterized by loss of skeletal muscle mass and muscle weakness or loss of physical function, and frailty is another syndrome that has received great interest in recent years. Decreased organ function results in vulnerability to external stress. Frailty is associated with falls, fractures and hospitalization; however, there is the reversibility of returning to a healthy state with appropriate interventions. Frailty is classified into three subgroups: Physical frailty, social frailty and cognitive frailty, whereby sarcopenia is the main component of physical frailty. The present review discusses the associations between sarcopenia, frailty and type 2 DM based on current evidence.

Introduction

Animals, including humans, have been constantly exposed to the danger of starvation during the course of evolution. As a result, animals have acquired the ability to survive by storing excess energy in the form of fat and utilizing it in the starvation status (1,2). Ironically, however, in today's advanced society, where excessive energy intake and lack of exercise have become the norm, this mechanism has become the cause of obesity, muscle weakness and insulin resistance (IR), etc. (1,2).

Skeletal muscle is the largest organ in the human body and plays an important role in energy metabolism and glucose uptake. Skeletal muscle is the most energy-consuming organ in the human body, with a heat expenditure of 30% of the total body in the basal metabolism (3). In patients with type 2 diabetes mellitus (DM), there is a marked decrease in glucose uptake in the skeletal muscle (4,5). It is well known that moderate exercise increases energy expenditure, reduces obesity, and increases glucose metabolism, thereby preventing and improving type 2 DM. Skeletal muscle accounts for about 50% of the body weight, and about 15% of the circulating blood volume is supplied to the skeletal muscle at rest (6). Approximately 20% of the oxygen consumed by the entire body is consumed by the skeletal muscle. Glucose in the blood is transported to the skeletal muscle and metabolized by oxygen. Continuous aerobic exercise increases oxygen uptake and improves glucose metabolism in the skeletal muscle (6). Endurance exercise can improve aerobic metabolism and thus can reduce hyperglycemia in patients with type 2 DM.

Aging-related changes in the body composition are characterized by the followings: (1) a 40% decrease in lean body mass and an increase in fat mass between the ages of 20 and 70 (7). (2) The overall fat mass increases between the ages of 20 and 70, but the peripheral fat tends to decrease compared to the central fat (7). (3) After the age of 70, both lean body mass and fat mass decrease (7). (4) Visceral fat increases with aging (8). (5) Fat is more likely to be deposited in the skeletal muscle and the liver (8). (6) Increased visceral fat is associated with impaired glucose metabolism, and intramuscular and intrahepatic fat deposition is associated with IR through the release of adipokines and free fatty acids (9). (7) Decreased skeletal muscle mass results in a decrease in the basal metabolism of 2–3% per decade after the age of 20, and 4% per decade after the age of 50 (10,11).

The effects of insulin and insulin-like growth factor-1 (IGF-1) can be roughly divided into metabolic and proliferative effects, with insulin mainly exerting the former. Insulin exerts a variety of effects, many of which are mediated by Akt, including increasing glucose uptake, promoting glycogen synthesis and inhibiting glycogen degradation, increasing free fatty acid uptake, increasing protein synthesis, promoting muscle hypertrophy, and inhibiting protein degradation (12,13). In addition, the mitogen-activated protein kinases (MAPKs) pathway promotes cell proliferation. Insulin, together with IGF-1, can increase skeletal muscle mass (12,13). In addition to skeletal muscle, liver and adipose tissue are important insulin target organs, and insulin signaling in these organs mainly promotes protein anabolism. Insulin signaling promotes lipid synthesis and inhibits lipid degradation in the liver and adipose tissue, leading to the storage of excess energy. Inhibition of glycogenesis in the liver is also an important role of insulin (12,13). Fat, on the other hand, contains more energy per equal weight than glucose and other substances (protein: 4.1 kcal/g, fat: 9.3 kcal/g, and glucose: 4.1 kcal/g), making fat the most suitable substance for storing large amounts of energy (14,15). Glycogen is a short-term form of energy storage, whereas fat is a long-term, high-volume form of storage (14,15). Amino acid intake is expected to inhibit muscle atrophy by inhibiting the degradation of skeletal muscle protein as well as promoting the synthesis of skeletal muscle protein (1618).

Skeletal muscle mass progressively declines with aging, leading to a decline in muscle strength and physical function. In 1989, Rosenberg coined the term ‘sarcopenia’ (from the Greek words ‘sarx’ meaning muscle, and ‘penia’ meaning loss) to describe the loss of muscle mass associated with aging. The term ‘sarcopenia’ has been widely accepted, and sarcopenia is now defined as a condition associated with loss of skeletal muscle mass and muscle weakness or loss of physical function (19,20). Guidelines for sarcopenia of the European Working Group on Sarcopenia in Older People (EWGSOP) (21) and the Asian Working Group for Sarcopenia (AWGS) (22) were published in 2010 and 2014, respectively, and in 2016, the Japanese Society of Hepatology issued sarcopenia assessment criteria specific to liver disease (23). In addition, a revised EWGSOP guideline was issued in 2019 (24) and a revised AWGS guideline in 2020 (25). The etiology of sarcopenia is thought to involve several complex factors in addition to aging. These include changes in sex hormones, underlying diseases (i.e., secondary sarcopenia), myokines, IR, poor nutritional intake of proteins and other nutrients, and disuse syndrome due to reduced motor function caused by loss of skeletal muscle mass, etc. (23). Considerable attention has been paid to the relationship between type 2 DM-related IR and skeletal muscle (26). On the other hand, frailty, as well as sarcopenia, is another condition that has received much attention in recent years (20,27,28). Decreased organ function leads to vulnerability to external stress. A condition, in which acute stresses such as infections, surgeries and accidents make it easier for physical functions to decline with decreased organ function, is called frailty (27,28). Frailty is also associated with outcomes such as need for care, death, falls and fractures, and hospitalization, but it encompasses the reversibility of returning to a healthy state with appropriate interventions (27,28). Frailty can be divided into the three main categories: physical frailty, social frailty and cognitive frailty, and sarcopenia is the main component of physical frailty (29). As in the case of sarcopenia, much attention has been paid to the relationship between type 2 DM and frailty. This review outlines the relationship between sarcopenia, frailty and type 2 DM.

Type 2 DM and skeletal muscle

Skeletal muscle takes up more than 80% of the glucose in the peripheral tissues by insulin and uses it for energy or stores the excess energy as glycogen (30). In diabetic patients, the total utilization of insulin in the whole body is about half of that in healthy individuals, which is due to the reduced utilization in the skeletal muscle (30). In other words, skeletal muscle is the main target of insulin and the largest organ that utilizes blood glucose. Therefore, the loss of skeletal muscle mass due to sarcopenia leads to decreased glucose utilization by insulin (i.e., IR). Decreased physical activity due to decreased skeletal muscle mass is also responsible for decreased glucose and lipid metabolism in the skeletal muscle (29). In addition, it is reported that IL-6 secreted by skeletal muscle stimulates the secretion of glucagon like peptide-1 (GLP-1) during exercise, and exercise is also associated with changes in insulin and glucagon secretion (31,32). Liver cirrhosis (LC), which is considered to be a typical form of secondary sarcopenia because of its tendency to cause protein-energy malnutrition, is also associated with a high rate of IR (20,23,3335). LC patients are characterized by postprandial hyperglycemia (33). The organ network between liver and skeletal muscle has recently received much attention (23).

The glucose transporter 4 (GLUT4) is important for the uptake of blood glucose into skeletal muscle cells (36). GLUT4 is normally intracellular, but upon stimulation of glucose uptake, it moves to the cell membrane and acts as a transport pathway for glucose uptake into the cell. When the stimulus subsides, GLUT4 returns to the cell and waits for another opportunity to mobilize. When insulin binds to insulin receptors on the surface of the cell membrane of skeletal muscle, intracellular signaling molecules such as IRS, PI3 kinase, and Akt are activated in turn (36). This information is ultimately transmitted to GLUT4 stores in the cell, resulting in glucose uptake. IR in patients with type 2 DM is thought to be due to abnormalities in this signaling pathway (18,36). In patients with type 2 DM, glucose uptake in peripheral tissues by insulin is reduced. As mentioned earlier, decreased glucose uptake has been shown to originate primarily in the skeletal muscle among the organs of the body, and exercise is a powerful way to increase glucose uptake and improve IR (3). The effects of prolonged exercise, such as marathon running, include the conversion of skeletal muscle to red muscle, an increase in the number of mitochondria, and an increase in the amount of GLUT4 (36,37). Moderate exercise improves mitochondrial function and also increases the ability of blood glucose uptake by increasing GLUT4 levels (36,37). Activation of mitochondrial function increases the beta-oxidation of fatty acids, facilitating the processing of free fatty acids being released from adipose tissue and reducing the accumulation of triglycerides in the liver (38,39). Thus, moderate exercise can efficiently improve dysfunction in the skeletal muscle.

The protein peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) plays an important role in increasing mitochondrial mass and is thought to be the key to improving the condition of lifestyle-related diseases by exercise (40). In other words, increased expression of PGC-1α causes increased mitochondria, change to red muscle, increased energy expenditure, and weight loss as seen in exercise (40). PGC-1α is highly expressed in organs with active metabolism, such as brown adipocytes, skeletal muscle, and liver, and is especially highly expressed in soleus muscle, which has many type I myofibers (described later) among skeletal muscles (41). It is known that PGC-1α expression is increased by sustained exercise (42,43). Mice overexpressing PGC-1α in the skeletal muscle (PGC-1α transgenic mice) showed increased slow-twitch troponin I, myoglobin, and mitochondrial mass (44), characteristics of type I and IIA myofibers (described later), as well as enhanced expression of branched-chain amino acid (BCAA) metabolizing enzymes (45), resulting in increased endurance exercise capacity. It has been reported that expression of PGC-1α improves insulin sensitivity in diabetic mouse models (46).

Type 2 DM and myofiber

Skeletal muscle can be roughly classified into the type I myofibers, which have a slow contraction rate, and the type II myofibers, which have a fast contraction rate and a strong contractile force (47). Type II myofibers can be further divided into the type IIA myofibers with high aerobic glycolytic capacity and the type IIB myofibers with low aerobic glycolytic capacity (47). Type I and IIA myofibers with high aerobic glycolytic capacity have higher mitochondrial density and activity, actively metabolize glucose and lipids, and are less fatigued during endurance exercise (48). Type IIB myofibers have low mitochondrial density and activity, and are mainly involved in anoxic glycolysis in the cytoplasm, making them suitable for exercise that requires instantaneous contraction such as short-distance running (48). The distribution of these myofibers varies from region to region in the body. In the deep layers close to the bone, the proportion of type I and IIA myofibers is higher, and in the superficial layers close to the skin, the proportion of type IIB myofibers is higher (48). The composition of myofiber types changes with various stimuli. For example, the proportion of type I and IIA fibers increases with continuous aerobic exercise (49). On the other hand, it is known that the percentage of type IIB myofibers is higher and the percentage of type I and IIA myofibers is lower in patients with type 2 DM (49). Therefore, increasing the proportion of type I and IIA myofibers may be an effective means of preventing type 2 DM by increasing the number of mitochondria and energy metabolism. The skeletal muscle in patients with type 2 DM has fewer capillaries, regardless of the type of myofiber (50).

Type 2 DM and sarcopenia

According to the data released by the Japan Ministry of Health, Labour and Welfare in 2017, there are 3.289 million people with type 2 DM in Japan, and one in five adults has impaired glucose tolerance. The global diabetic population in 2019 is about 463 million. The International Diabetes Federation predicts that the number of people with type 2 DM will approach 600 million by 2035. Previous reports (Asian subjects) using the AWGS criteria for the diagnosis of sarcopenia reported the frequency of sarcopenia in patients with type 2 DM to be 7–28.8% (5167). Among these reports, the average HbA1c of the target patients was the lowest in the report by Fukuoka et al (average HbA1c=7.0) (66) and was the highest in the report by Murai et al (average HbA1c=9.5) (63).

A 6-year follow-up study of limb skeletal muscle mass using dual-energy X-ray absorption (DXA) in 2,675 elderly people aged 70–79 years reported that the amount of limb muscle mass loss in elderly people with type 2 DM was significantly greater than that in elderly people without type 2 DM (68). In a cross-sectional study in Indians, the prevalence of sarcopenia in patients with type 2 DM was higher than in controls with an odds ratio (OR) of 3.48 (69). A report from Netherlands found significant decrease in skeletal muscle mass and muscle strength in patients with type 2 DM, even after adjusting for age, body mass index (BMI), fasting glucose, HDL cholesterol, BCAAs, and protein intake (70). An analysis of 14,528 individuals in the National Health and Nutrition Examination Surveys III in the United States indicated that sarcopenia is involved in glucose metabolism independently of obesity, that this tendency is stronger in individuals younger than 60 years, and that loss of skeletal muscle mass is a predictor of the type 2 DM incidence (71). When 932 subjects aged 40 years or older were classified into sarcopenia and normal groups using DXA, HbA1c level in the male sarcopenia group were significantly higher than that in the normal group (72). Sugimoto et al (59) examined the relationship between HbA1c levels and the prevalence of sarcopenia in 2,813 subjects aged 40 years and older, and the summary of their results were: i) the prevalence of sarcopenia increased with increasing HbA1c levels, and these linear relationships were particularly marked in non-obese type 2 DM patients with a BMI <22.3 kg/m2 (the prevalence of sarcopenia was: 7.0% in the HbA1c <6.5% group; 18.5% in the HbA1c 6.5–7.0% group; 20.3% in the HbA1c 7.0–8.0% group; and 26.7% in the HbA1c 8.0% or higher group), ii) higher HbA1c levels were more strongly associated with lower skeletal muscle index (OR=5.42) than lower grip strength (OR=1.89) or walking speed (OR=1.13), and iii) no association was found between blood glucose levels and the prevalence of sarcopenia in the elderly people with normal blood glucose levels (59). Lee et al (73) divided 3,132 elderly non-diabetic men aged 65 years or older into quartiles according to homeostasis model assessment of insulin resistance (HOMA-IR) levels, followed them for 5 years using DXA, and they found that elderly men with higher HOMA-IR had greater loss of limb skeletal muscle mass (73). These results suggest that aging-related muscle mass loss may affect HbA1c levels through reduced glucose utilization, or that increased IR may further contribute to sarcopenia by decreasing skeletal muscle protein synthesis and accelerating protein degradation. Hyperglycemia in the elderly is an aggravating factor for sarcopenia, dementia, and depression, and leads to ADL decline (74) (Fig. 1).

Sarcopenia is a risk factor for developing type 2 DM, and type 2 DM is a risk factor for developing sarcopenia (71,75). Regarding the mechanism, Ogawa's research group revealed for the first time that elevated blood glucose levels cause muscle mass loss through the action of two proteins, KLF15 and WWP1 (76). IR prevents the proliferation and growth of skeletal muscle cells, leading to a decrease in the skeletal muscle mass. They reported that in mice with type 2 DM, the amount of a transcription factor called KLF15 protein increases in the skeletal muscle as skeletal muscle mass decreases (76). KLF15 promotes skeletal muscle loss by increasing the expression of genes that cause skeletal muscle degradation and muscle atrophy. Their research confirmed that mice without KLF15 did not lose skeletal muscle mass in diabetes. They also found that (1) the degradation of KLF15 is inhibited by elevated blood glucose levels, (2) it is accumulated in the skeletal muscle, and (3) a protein called WWP1 plays an important role in regulating its degradation. WWP1 is one of the proteins called ‘ubiquitin ligase’, which binds ubiquitin to another protein. Proteins with large amounts of ubiquitin are degraded faster (77). Their study showed that WWP1 specifically binds ubiquitin to KLF15. When blood glucose levels increase, the amount of WWP1 is reduced, resulting in less ubiquitin binding to KLF15, which inhibits KLF15 degradation (76). The characteristics of the skeletal muscle in patients with type 2 DM are listed in Table I.

Table I.

Characteristics of the skeletal muscle in patients with type 2 diabetes mellitus.

Table I.

Characteristics of the skeletal muscle in patients with type 2 diabetes mellitus.

No. Characteristics
1Decreased insulin-induced glucose uptake into the skeletal muscle.
2Higher percentage of type IIB myofibers and lower percentage of type I and IIA myofibers.
3Fewer capillaries regardless of myofiber type.
4Decreased expression of proliferator-activated receptor-γ coactivator-1a and increased expression of Krüppel-like factor 15.

Glucose lowering drugs and sarcopenia

Sodium glucose co-transporter 2 (SGLT2) inhibitors are drugs that tilt the body toward a state of protein catabolism by increasing the amount of urinary glucose, which also reduces body fat, but at the same time reduces muscle mass because of the tilt toward protein catabolism (78,79). Metformin has been shown to inhibit sarcopenia (8082). During acute exercise, enzyme activity increases to burn fat and carbohydrates to compensate for the large amount of energy consumed by the skeletal muscle. In this process, AMP-activated protein kinase (AMPK), which senses the energy status in the skeletal muscle, is activated (83,84). AMPK regulates energy metabolism in cells throughout the body. When the intracellular energy state becomes low energy (i.e., high AMP/ATP ratio) after exercise, AMPK is activated and sends GLUT4 out onto the cell membrane to take up glucose and break down fatty acids to obtain energy (83,84). Metformin activates AMPK (85). In other words, metformin takes glucose into the muscle cells by the same mechanism as the effect of exercise. Bouch et al (53) reported that in a comparison of sarcopenic and non-sarcopenic groups in patients with type 2 DM, the non-sarcopenic group had a significantly higher rate of metformin use (19% vs. 54%) (53). On the other hand, in a cohort study that followed changes in limb skeletal muscle mass in men aged 65 years and older for 3.5 years, limb skeletal muscle mass decreased by 4.4% in the group of diabetic patients who used drugs other than insulin sensitizers, while in the group that used insulin sensitizers, the decrease rate of limb skeletal muscle mass was 1.8% (86). Out of the previous reports using the AWGS criteria for the diagnosis of sarcopenia in Asians (mentioned earlier) (5167), Sugimoto et al (59) reported the lowest frequency of sarcopenia (7%), and insulin preparations were used in about 70% of the non-sarcopenic patients in their study (59). On the other hand, Cui et al (58) reported the highest frequency of sarcopenia (28.8%), and insulin preparations were used in about 70% of sarcopenic patients. Ida et al (62) reported that the use of insulin preparations was significantly higher in the sarcopenia group compared to the non-sarcopenia group (79.5% vs. 62.1%). Thus, further investigations of the effect of insulin preparations on sarcopenia are needed.

Type 2 DM and frailty

The concept of frailty indicates ‘a vulnerable state with reduced recovery to stress’ and ‘a condition that increases the risk of health problems, including falls, disability, and death’. In the elderly people, there is a risk that minor stresses may lead to changes in health status (e.g., from independent to needing care, from mobile to immobile, easily falling, from clear consciousness to delirium, etc.) that are disproportionate to the cause. It is important to take this into consideration during medical interventions (35,87).

Type 2 DM is thought to progress physical frailty and increase the risk of needing care and death through IR and inflammation (88), and clinical data supporting this relationship have been accumulated these days. Several prospective cohort studies and cross-sectional studies have reported the relationship between type 2 DM and frailty. In a prospective cohort study of non-institutionalized individuals aged 60 years or older (observation period, 3.5 years), type 2 DM increased the incidence of new cases of frailty (OR=2.18) (89). In a prospective study of subjects aged 65 years and older, type 2 DM was a significant risk factor for the progression from pre-frail to frailty, especially in patients with macrovascular diseases. Inappropriate lifestyle, abdominal obesity, and poor glycemic and lipid control were associated with the frailty progression, and dietary therapy reduced the risk (90). There are also reports that HOMA-IR is associated with the onset of frailty (91,92), and that frailty is associated with 2-hour blood glucose levels of 75 g oral glucose tolerance test (OGTT) (93,94). On the other hand≠, some longitudinal studies have shown that frailty is a risk factor for the development of type 2 DM, and type 2 DM and frailty can interact and form a vicious circle (95). In patients with type 2 DM, frailty can be a risk factor for pathological fracture (96). The presence of osteoporosis in female patients with type 2 DM predisposes to the development of frailty (97). Not only frailty but also pre-frail can be a poor prognostic factor in patients with type 2 DM (98).

Regarding the relationship between glycemic control and frailty, in a cross-sectional study of the Women's Health and Aging Study, the frequency of frailty was significantly higher in the group with HbA1c 6.5% or higher than in the group with HbA1c less than 6.0% (99), and in a longitudinal study, the frequency of frailty was 3.3 times higher in the group with HbA1c >8% than in the group with HbA1c <5.5% at baseline (100). In a community-based cohort study, higher 5-year mean blood glucose levels were associated with more frailty in non-diabetic patients, whereas a U-shaped association was found in diabetic patients. Baseline fasting blood glucose (FBS) <150 mg/dl (HbA1c=6.9%) had an OR of 1.41, and FBS >190 mg/dl (HbA1c=8.2%) had an OR of 1.30, indicating a higher incidence of frailty, however, the incidence of frailty was the lowest at FBS 170 mg/dl (HbA1c=7.6%) (101). A recent cross-sectional study of elderly diabetic patients in Japan showed that the lower the HbA1c level, the greater the risk of developing frailty (102). Therefore, low HbA1c may be a risk factor for developing frailty in patients with type 2 DM. The reason for this may be that strict glycemic control may lead to frailty via hypoglycemia. In the Japanese guidelines for the treatment of type 2 DM in the elderly, it states that there are no reports that show that good glycemic control inhibits or improves frailty. On the other hand, there are several reports on management goals for type 2 DM patients with frailty. In type 2 DM patients with frailty, there is a report that the group with HbA1c 8–8.9% had preserved ADL and lower risk of death compared to the group with HbA1c 7–7.9%, and caution against over-lowering of blood glucose level is needed (103). As elderly patients may not have hypoglycemic symptoms such as sweating or palpitations despite low blood glucose levels, hypoglycemia may be overlooked in the routine medical care, which is associated with the risk of frailty progression (74). As lifestyle-related diseases such as type 2 DM can be risk factors for frailty, it is important to strictly manage them at least in the middle age. However, it is important to note that in the old age, especially over the age of 75 with reduced physiological reserve, the adverse events associated with strict glycemic control can lead to frailty and even the need for nursing care. In the elderly, hypoglycemia, depression, dementia, and frailty form a vicious cycle through reduced physical activity and reduced dietary intake (74) (Fig. 2)

Conclusion

The relationship between sarcopenia, frailty and type 2 DM was reviewed from the molecular biological and clinical perspectives. The frequency of type 2 DM increases with aging, and in Japan, about 20% of the population over the age of 70 suffer from type 2 DM. In an aging society, the association between sarcopenia or frailty and type 2 DM is an important issue. Skeletal muscles of diabetic patients have a different distribution of myofibers compared to healthy individuals. Hyperglycemia leads to the degradation of muscle proteins. There is a close relationship between the severity of type 2 DM and the frequency of sarcopenia. Hypoglycemia in diabetic patients with frailty should be carefully monitored. It is important to note that appropriate interventions in patients with sarcopenia or frailty can prevent the development of type 2 DM, and appropriate interventions in type 2 DM patients can prevent the development of sarcopenia or frailty, which may lead to improved healthy life expectancy.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

HN drafted the initial manuscript. SF, AA, KY, HO, SN and KH edited and reviewed the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

IR

insulin resistance

DM

diabetes mellitus

IGF-1

insulin-like growth factor-1

EWGSOP

European Working Group on Sarcopenia in Older People

AWGS

Asian Working Group for Sarcopenia

LC

liver cirrhosis

GLUT4

glucose transporter 4

PGC-1α

peroxisome proliferator-activated receptor-γ coactivator-1α

BCAA

branched-chain amino acid

DXA

dual-energy X-ray absorption

OR

odds ratio

BMI

body mass index

HOMA-IR

homeostasis model assessment of insulin resistance

AMPK

AMP-activated protein kinase

FBS

fasting blood glucose

References

1 

Gibbons A: HUMAN EVOLUTION. Why humans are the high-energy apes. Science. 352:6392016. View Article : Google Scholar : PubMed/NCBI

2 

Roth J, Szulc AL and Danoff A: Energy, evolution, and human diseases: An overview. Am J Clin Nutr. 93:875S–883S. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Evans PL, McMillin SL, Weyrauch LA and Witczak CA: Regulation of skeletal muscle glucose transport and glucose metabolism by exercise training. Nutrients. 11:24322019. View Article : Google Scholar : PubMed/NCBI

4 

Seo JA, Kang MC, Yang WM, Hwang WM, Kim SS, Hong SH, Heo JI, Vijyakumar A, Pereira de ML, Uner A, et al: Apolipoprotein J is a hepatokine regulating muscle glucose metabolism and insulin sensitivity. Nat Commun. 11:20242020. View Article : Google Scholar : PubMed/NCBI

5 

Severinsen MCK and Pedersen BK: Muscle-organ crosstalk: The emerging roles of myokines. Endocr Rev. 41:594–609. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Mukund K and Subramaniam S: Skeletal muscle: A review of molecular structure and function, in health and disease. Wiley Interdiscip Rev Syst Biol Med. 12:e14622020. View Article : Google Scholar : PubMed/NCBI

7 

Jackson AS, Janssen I, Sui X, Church TS and Blair SN: Longitudinal changes in body composition associated with healthy ageing: Men, aged 20–96 years. Br J Nutr. 107:1085–1091. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Schosserer M, Grillari J, Wolfrum C and Scheideler M: Age-induced changes in white, brite, and brown adipose depots: A mini-review. Gerontology. 64:229–236. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Wu H and Ballantyne CM: Skeletal muscle inflammation and insulin resistance in obesity. J Clin Invest. 127:43–54. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Kawada T: Basal metabolic rate parameters, sarcopenia, and frailty in older males. J Am Med Dir Assoc. 20:9192019. View Article : Google Scholar : PubMed/NCBI

11 

Zampino M, Semba RD, Adelnia F, Spencer RG, Fishbein KW, Schrack JA, Simonsick EM and Ferrucci L: Greater skeletal muscle oxidative capacity is associated with higher resting metabolic rate: Results from the baltimore longitudinal study of aging. J Gerontol A Biol Sci Med Sci. 75:2262–2268. 2020. View Article : Google Scholar : PubMed/NCBI

12 

Saltiel AR: Insulin signaling in health and disease. J Clin Invest. 131:e1422412021. View Article : Google Scholar : PubMed/NCBI

13 

Kadowaki T, Ueki K, Yamauchi T and Kubota N: SnapShot: Insulin signaling pathways. Cell. 148:624–624.e1. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Burke LM, Kiens B and Ivy JL: Carbohydrates and fat for training and recovery. J Sports Sci. 22:15–30. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Volek JS, Noakes T and Phinney SD: Rethinking fat as a fuel for endurance exercise. Eur J Sport Sci. 15:13–20. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Sato T, Ito Y and Nagasawa T: Regulation of skeletal muscle protein degradation and synthesis by oral administration of lysine in rats. J Nutr Sci Vitaminol (Tokyo). 59:412–419. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Sato T, Ito Y and Nagasawa T: L-Lysine suppresses myofibrillar protein degradation and autophagy in skeletal muscles of senescence-accelerated mouse prone 8. Biogerontology. 18:85–95. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Kamei Y, Hatazawa Y, Uchitomi R, Yoshimura R and Miura S: Regulation of skeletal muscle function by amino acids. Nutrients. 12:2612020. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Nishikawa H, Fukunishi S, Asai A, Nishiguchi S and Higuchi K: Sarcopenia and frailty in liver Cirrhosis. Life (Basel). 11:3992021.PubMed/NCBI

21 

Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, Martin FC, Michel JP, Rolland Y, Schneider SM, et al: Sarcopenia: European consensus on definition and diagnosis: Report of the European working group on sarcopenia in older people. Age Ageing. 39:412–423. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Chen LK, Liu LK, Woo J, Assantachai P, Auyeung TW, Bahyah KS, Chou MY, Chen LY, Hsu PS, Krairit O, et al: Sarcopenia in Asia: Consensus report of the Asian working group for sarcopenia. J Am Med Dir Assoc. 15:95–101. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Nishikawa H, Shiraki M, Hiramatsu A, Moriya K, Hino K and Nishiguchi S: Japan society of hepatology guidelines for sarcopenia in liver disease (1st edition): Recommendation from the working group for creation of sarcopenia assessment criteria. Hepatol Res. 46:951–963. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Cruz-Jentoft AJ, Bahat G, Bauer J, Boirie Y, Bruyère O, Cederholm T, Cooper C, Landi F, Rolland Y, Sayer AA, et al: Sarcopenia: Revised European consensus on definition and diagnosis. Age Ageing. 48:16–31. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Chen LK, Woo J, Assantachai P, Auyeung TW, Chou MY, Iijima K, Jang HC, Kang L, Kim M, Kim S, et al: Asian working group for sarcopenia: 2019 consensus update on sarcopenia diagnosis and treatment. J Am Med Dir Assoc. 21:300–307.e2. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Sampaio RAC, Sewo Sampaio PY, Uchida MC and Arai H: Management of dynapenia, sarcopenia, and frailty: The role of physical exercise. J Aging Res. 2020:81867692020. View Article : Google Scholar : PubMed/NCBI

27 

Satake S and Arai H: Implications of frailty screening in clinical practice. Curr Opin Clin Nutr Metab Care. 20:4–10. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Sugimoto K, Rakugi H, Kojima T, Ishii S, Akishita M, Tamura Y, Araki A, Kozaki K, Senda K, Fukuoka H, et al: Chapter 4 Frailty and specific diseases. Geriatr Gerontol Int. 20 (Suppl 1):S25–S37. 2020. View Article : Google Scholar

29 

Mesinovic J, Zengin A, De Courten B, Ebeling PR and Scott D: Sarcopenia and type 2 diabetes mellitus: A bidirectional relationship. Diabetes Metab Syndr Obes. 12:1057–1072. 2019. View Article : Google Scholar : PubMed/NCBI

30 

DeFronzo RA: Lilly lecture 1987. The triumvirate: Beta-cell, muscle, liver. A collusion responsible for NIDDM. Diabetes. 37:667–687. 1988. View Article : Google Scholar : PubMed/NCBI

31 

Bu L, Cao X, Zhang Z, Wu H, Guo R and Ma M: Decreased secretion of tumor necrosis factor-α attenuates macrophages-induced insulin resistance in skeletal muscle. Life Sci. 244:1173042020. View Article : Google Scholar : PubMed/NCBI

32 

Love KM, Liu J, Regensteiner JG, Reusch JEB and Liu Z: GLP-1 and insulin regulation of skeletal and cardiac muscle microvascular perfusion in type 2 diabetes. J Diabetes. 12:488–498. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Nishikawa H and Osaki Y: Clinical significance of therapy using branched-chain amino acid granules in patients with liver cirrhosis and hepatocellular carcinoma. Hepatol Res. 44:149–158. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Nishikawa H, Enomoto H, Ishii A, Iwata Y, Miyamoto Y, Ishii N, Yuri Y, Takata R, Hasegawa K, Nakano C, et al: Development of a simple predictive model for decreased skeletal muscle mass in patients with compensated chronic liver disease. Hepatol Res. 47:1223–1234. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Nishikawa H, Yoh K, Enomoto H, Iwata Y, Sakai Y, Kishino K, Shimono Y, Ikeda N, Takashima T, Aizawa N, et al: Sarcopenia and frailty in chronic liver damage: Common and different points. In Vivo. 34:2549–2559. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Richter EA and Hargreaves M: Exercise, GLUT4, and skeletal muscle glucose uptake. Physiol Rev. 93:993–1017. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Flores-Opazo M, McGee SL and Hargreaves M: Exercise and GLUT4. Exerc Sport Sci Rev. 48:110–118. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Hood DA, Memme JM, Oliveira AN and Triolo M: Maintenance of skeletal muscle mitochondria in health, exercise, and aging. Annu Rev Physiol. 81:19–41. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Memme JM, Erlich AT, Phukan G and Hood DA: Exercise and mitochondrial health. J Physiol. 599:803–817. 2021. View Article : Google Scholar : PubMed/NCBI

40 

Hatazawa Y, Tadaishi M, Nagaike Y, Morita A, Ogawa Y, Ezaki O, Takai-Igarashi T, Kitaura Y, Shimomura Y, Kamei Y and Miura S: PGC-1α-mediated branched-chain amino acid metabolism in the skeletal muscle. PLoS One. 9:e910062014. View Article : Google Scholar : PubMed/NCBI

41 

Schnyder S and Handschin C: Skeletal muscle as an endocrine organ: PGC-1α, myokines and exercise. Bone. 80:115–125. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Norheim F, Langleite TM, Hjorth M, Holen T, Kielland A, Stadheim HK, Gulseth HL, Birkeland KI, Jensen J and Drevon CA: The effects of acute and chronic exercise on PGC-1α, irisin and browning of subcutaneous adipose tissue in humans. FEBS J. 281:739–749. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Booth FW, Ruegsegger GN, Toedebusch RG and Yan Z: Endurance exercise and the regulation of skeletal muscle metabolism. Prog Mol Biol Transl Sci. 135:129–151. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Islam H, Hood DA and Gurd BJ: Looking beyond PGC-1α: Emerging regulators of exercise-induced skeletal muscle mitochondrial biogenesis and their activation by dietary compounds. Appl Physiol Nutr Metab. 45:11–23. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Jang C, Oh SF, Wada S, Rowe GC, Liu L, Chan MC, Rhee J, Hoshino A, Kim B, Ibrahim A, et al: A branched-chain amino acid metabolite drives vascular fatty acid transport and causes insulin resistance. Nat Med. 22:421–426. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Arany Z, He H, Lin J, Hoyer K, Handschin C, Toka O, Ahmad F, Matsui T, Chin S, Wu PH, et al: Transcriptional coactivator PGC-1 alpha controls the energy state and contractile function of cardiac muscle. Cell Metab. 1:259–271. 2005. View Article : Google Scholar : PubMed/NCBI

47 

Cretoiu D, Pavelescu L, Duica F, Radu M, Suciu N and Cretoiu SM: Myofibers. Adv Exp Med Biol. 1088:23–46. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Drake JC and Yan Z: Precision remodeling: How exercise improves mitochondrial quality in myofibers. Curr Opin Physiol. 10:96–101. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Hickey MS, Carey JO, Azevedo JL, Houmard JA, Pories WJ, Israel RG and Dohm GL: Skeletal muscle fiber composition is related to adiposity and in vitro glucose transport rate in humans. Am J Physiol. 268((3 Pt 1)): E453–E457. 1995.PubMed/NCBI

50 

Märin P, Krotkiewski M, Andersson B and Björntorp P: Muscle fiber composition and capillary density in women and men with NIDDM. Diabetes Care. 17:382–386. 1994. View Article : Google Scholar : PubMed/NCBI

51 

Kaji A, Hashimoto Y, Kobayashi Y, Sakai R, Okamura T, Miki A, Hamaguchi M, Kuwahata M, Yamazaki M and Fukui M: Sarcopenia is associated with tongue pressure in older patients with type 2 diabetes: A cross-sectional study of the KAMOGAWA-DM cohort study. Geriatr Gerontol Int. 19:153–158. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Sazlina SG, Lee PY, Chan YM, A Hamid MS and Tan NC: The prevalence and factors associated with sarcopenia among community living elderly with type 2 diabetes mellitus in primary care clinics in Malaysia. PLoS One. 15:e02332992020. View Article : Google Scholar : PubMed/NCBI

53 

Bouchi R, Fukuda T, Takeuchi T, Minami I, Yoshimoto T and Ogawa Y: Sarcopenia is associated with incident albuminuria in patients with type 2 diabetes: A retrospective observational study. J Diabetes Investig. 8:783–787. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Hashimoto Y, Kaji A, Sakai R, Hamaguchi M, Okada H, Ushigome E, Asano M, Yamazaki M and Fukui M: Sarcopenia is associated with blood pressure variability in older patients with type 2 diabetes: A cross-sectional study of the KAMOGAWA-DM cohort study. Geriatr Gerontol Int. 18:1345–1349. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Okamura T, Mik A, Hashimoto Y, Kaji A, Sakai R, Osaka T, Hamaguchi M, Yamazaki M and Fukui M: Deficiency of energy intake rather than protein is associated with sarcopenia in early patients with type 2 diabetes: A cross-sectional study of the KAMOGAWA-DM cohort. J Diabetes. 11:477–483. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Okamura T, Hashimoto Y, Miki A, Kaji A, Sakai R, Iwai K, Osaka T, Kitagawa N, Ushigome E, Hamaguchi M, et al: High brain natriuretic peptide is associated with sarcopenia in patients with type 2 diabetes: A cross-sectional study of KAMOGAWA-DM cohort study. Endocr J. 66:369–377. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Bouchi R, Fukuda T, Takeuchi T, Nakano Y, Murakami M, Minami I, Izumiyama H, Hashimoto K, Yoshimoto T and Ogawa Y: Insulin treatment attenuates decline of muscle mass in Japanese patients with type 2 diabetes. Calcif Tissue Int. 101:1–8. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Cui M, Gang X and Wang G, Xiao X, Li Z, Jiang Z and Wang G: A cross-sectional study: Associations between sarcopenia and clinical characteristics of patients with type 2 diabetes. Medicine (Baltimore). 99:e187082020. View Article : Google Scholar : PubMed/NCBI

59 

Sugimoto K, Tabara Y, Ikegami H, Takata Y, Kamide K, Ikezoe T, Kiyoshige E, Makutani Y, Onuma H, Gondo Y, et al: Hyperglycemia in non-obese patients with type 2 diabetes is associated with low muscle mass: The multicenter study for clarifying evidence for sarcopenia in patients with diabetes mellitus. J Diabetes Investig. 10:1471–1479. 2019. View Article : Google Scholar : PubMed/NCBI

60 

Fung FY, Koh YLE, Malhotra R, Ostbye T, Lee PY, Shariff Ghazali S and Tan NC: Prevalence of and factors associated with sarcopenia among multi-ethnic ambulatory older Asians with type 2 diabetes mellitus in a primary care setting. BMC Geriatr. 19:1222019. View Article : Google Scholar : PubMed/NCBI

61 

Mori H, Kuroda A, Ishizu M, Ohishi M, Takashi Y, Otsuka Y, Taniguchi S, Tamaki M, Kurahashi K, Yoshida S, et al: Association of accumulated advanced glycation end-products with a high prevalence of sarcopenia and dynapenia in patients with type 2 diabetes. J Diabetes Investig. 10:1332–1340. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Ida S, Murata K, Nakadachi D, Ishihara Y, Imataka K, Uchida A, Monguchi K, Kaneko R, Fujiwara R and Takahashi H: Association between dynapenia and decline in higher-level functional capacity in older men with diabetes. Geriatr Gerontol Int. 18:1393–1397. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Murai J, Nishizawa H, Otsuka A, Fukuda S, Tanaka Y, Nagao H, Sakai Y, Suzuki M, Yokota S, Tada H, et al: Low muscle quality in Japanese type 2 diabetic patients with visceral fat accumulation. Cardiovasc Diabetol. 17:1122018. View Article : Google Scholar : PubMed/NCBI

64 

Osaka T, Hamaguchi M, Hashimoto Y, Ushigome E, Tanaka M, Yamazaki M and Fukui M: Decreased the creatinine to cystatin C ratio is a surrogate marker of sarcopenia in patients with type 2 diabetes. Diabetes Res Clin Pract. 139:52–58. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Fukuda T, Bouchi R, Takeuchi T, Tsujimoto K, Minami I, Yoshimoto T and Ogawa Y: Sarcopenic obesity assessed using dual energy X-ray absorptiometry (DXA) can predict cardiovascular disease in patients with type 2 diabetes: A retrospective observational study. Cardiovasc Diabetol. 17:552018. View Article : Google Scholar : PubMed/NCBI

66 

Fukuoka Y, Narita T, Fujita H, Morii T, Sato T, Sassa MH and Yamada Y: Importance of physical evaluation using skeletal muscle mass index and body fat percentage to prevent sarcopenia in elderly Japanese diabetes patients. J Diabetes Investig. 10:322–330. 2019. View Article : Google Scholar : PubMed/NCBI

67 

Murata Y, Kadoya Y, Yamada S and Sank T: Sarcopenia in elderly patients with type 2 diabetes mellitus: Prevalence and related clinical factors. Diabetol Int. 9:136–142. 2017. View Article : Google Scholar : PubMed/NCBI

68 

Park SW, Goodpaster BH, Lee JS, Kuller LH, Boudreau R, de Rekeneire N, Harris TB, Kritchevsky S, Tylavsky FA, Nevitt M, et al: Excessive loss of skeletal muscle mass in older adults with type 2 diabetes. Diabetes Care. 32:1993–1997. 2009. View Article : Google Scholar : PubMed/NCBI

69 

Anbalagan VP, Venkataraman V, Pradeepa R, Deepa M, Anjana RM and Mohan V: The prevalence of presarcopenia in Asian Indian individuals with and without type 2 diabetes. Diabetes Technol Ther. 15:768–775. 2013. View Article : Google Scholar : PubMed/NCBI

70 

Leenders M, Verdijk LB, van der Hoeven L, Adam JJ, van Kranenbur J, Nilwik R and van Loon LJ: Patients with type 2 diabetes show a greater decline in muscle mass, muscle strength, and functional capacity with aging. J Am Med Dir Assoc. 14:585–592. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Srikanthan P, Hevener AL and Karlamangla AS: Sarcopenia exacerbates obesity-associated insulin resistance and dysglycemia: Findings from the National health and nutrition examination survey III. PLoS One. 5:e108052010. View Article : Google Scholar : PubMed/NCBI

72 

Sanada K, Miyachi M, Tanimoto M, Yamamoto K, Murakami H, Okumura S, Gando Y, Suzuki K, Tabata I and Higuchi M: A cross-sectional study of sarcopenia in Japanese men and women: Reference values and association with cardiovascular risk factors. Eur J Appl Physiol. 110:57–65. 2010. View Article : Google Scholar : PubMed/NCBI

73 

Lee CG, Boyko EJ, Strotmeyer ES, Lewis CE, Cawthon PM, Hoffman AR, Everson-Rose SA, Barrett-Connor E and Orwoll ES; Osteoporotic Fractures in Men Study Research Group, : Association between insulin resistance and lean mass loss and fat mass gain in older men without diabetes mellitus. J Am Geriatr Soc. 59:1217–1224. 2011. View Article : Google Scholar : PubMed/NCBI

74 

Araki A and Ito H: Diabetes mellitus and geriatric syndromes. Geriatr Gerontol Int. 9:105–114. 2009. View Article : Google Scholar : PubMed/NCBI

75 

Kim TN, Park MS, Yang SJ, Yoo HJ, Kang HJ, Song W, Seo JA, Kim SG, Kim NH, Baik SH, et al: Prevalence and determinant factors of sarcopenia in patients with type 2 diabetes: The Korean sarcopenic obesity study (KSOS). Diabetes Care. 33:1497–1499. 2010. View Article : Google Scholar : PubMed/NCBI

76 

Hirata Y, Nomura K, Senga Y, Okada Y, Kobayashi K, Okamoto S, Minokoshi Y, Imamura M, Takeda S, Hosooka T and Ogawa W: Hyperglycemia induces skeletal muscle atrophy via a WWP1/KLF15 axis. JCI Insight. 4:e1249522019. View Article : Google Scholar : PubMed/NCBI

77 

Varshavsky A: The ubiquitin system, autophagy, and regulated protein degradation. Annu Rev Biochem. 86:123–128. 2017. View Article : Google Scholar : PubMed/NCBI

78 

Sasaki T: Sarcopenia, frailty circle and treatment with sodium-glucose cotransporter 2 inhibitors. J Diabetes Investig. 10:193–195. 2019. View Article : Google Scholar : PubMed/NCBI

79 

Sasaki T, Sugawara M and Fukuda M: Sodium-glucose cotransporter 2 inhibitor-induced changes in body composition and simultaneous changes in metabolic profile: 52-week prospective LIGHT (Luseogliflozin: The components of weight loss in Japanese patients with type 2 diabetes mellitus) study. J Diabetes Investig. 10:108–117. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Chen F, Xu S, Wang Y, Chen F, Cao L, Liu T, Huang T, Wei Q, Ma G, Zhao Y and Wang D: Risk factors for sarcopenia in the elderly with type 2 diabetes mellitus and the effect of metformin. J Diabetes Res. 2020:39504042020. View Article : Google Scholar : PubMed/NCBI

81 

Laksmi PW, Setiati S, Tamin TZ, Soewondo P, Rochmah W, Nafrialdi N and Prihartono J: Effect of metformin on handgrip strength, gait speed, myostatin serum level, and health-related quality of life: A double blind randomized controlled trial among non-diabetic pre-frail elderly patients. Acta Med Indones. 49:118–127. 2017.PubMed/NCBI

82 

Yang Y, Liao Z and Xiao Q: Metformin ameliorates skeletal muscle atrophy in Grx1 KO mice by regulating intramuscular lipid accumulation and glucose utilization. Biochem Biophys Res Commun. 533:1226–1232. 2020. View Article : Google Scholar : PubMed/NCBI

83 

Pirkmajer S, Petrič M and Chibalin AV: The role of AMPK in regulation of Na+, K+-ATPase in skeletal muscle: Does the gauge always plug the sink? J Muscle Res Cell Motil. 42:77–97. 2021. View Article : Google Scholar : PubMed/NCBI

84 

McGee SL and Hargreaves M: Exercise adaptations: Molecular mechanisms and potential targets for therapeutic benefit. Nat Rev Endocrinol. 16:495–505. 2020. View Article : Google Scholar : PubMed/NCBI

85 

LaMoia TE and Shulman GI: Cellular and molecular mechanisms of metformin action. Endocr Rev. 42:77–96. 2021. View Article : Google Scholar : PubMed/NCBI

86 

Lee CG, Boyko EJ, Barrett-Connor E, Miljkovic I, Hoffman AR, Everson-Rose SA, Lewis CE, Cawthon PM, Strotmeyer ES and Orwoll ES; Osteoporotic Fractures in Men (MrOS) Study Research Group, : Insulin sensitizers may attenuate lean mass loss in older men with diabetes. Diabetes Care. 34:2381–2386. 2011. View Article : Google Scholar : PubMed/NCBI

87 

Clegg A, Young J, Iliffe S, Rikkert MO and Rockwood K: Frailty in elderly people. Lancet. 381:752–762. 2013. View Article : Google Scholar : PubMed/NCBI

88 

Morley JE, Malmstrom TK, Rodriguez-Mañas L and Sinclair AJ: Frailty, Sarcopenia and diabetes. J Am Med Dir Assoc. 15:853–859. 2014. View Article : Google Scholar : PubMed/NCBI

89 

García-Esquinas E, Graciani A, Guallar-Castillón P, López-García E, Rodríguez-Mañas L and Rodríguez-Artalejo F: Diabetes and risk of frailty and its potential mechanisms: A prospective cohort study of older adults. J Am Med Dir Assoc. 16:748–754. 2015. View Article : Google Scholar : PubMed/NCBI

90 

Espinoza SE, Jung I and Hazuda H: Frailty transitions in the San Antonio longitudinal study of aging. J Am Geriatr Soc. 60:652–660. 2012. View Article : Google Scholar : PubMed/NCBI

91 

Barzilay JI, Blaum C, Moore T, Xue QL, Hirsch CH, Walston JD and Fried LP: Insulin resistance and inflammation as precursors of frailty: The cardiovascular health study. Arch Intern Med. 167:635–641. 2007. View Article : Google Scholar : PubMed/NCBI

92 

Pérez-Tasigchana RF, León-Muñoz LM, Lopez-Garcia E, Gutierrez-Fisac JL, Laclaustra M, Rodríguez-Artalejo F and Guallar-Castillón P: Metabolic syndrome and insulin resistance are associated with frailty in older adults: A prospective cohort study. Age Ageing. 46:807–812. 2017. View Article : Google Scholar : PubMed/NCBI

93 

Walston J, McBurnie MA, Newman A, Tracy RP, Kop WJ, Hirsch CH, Gottdiener J and Fried LP; Cardiovascular Health Study, : Frailty and activation of the inflammation and coagulation systems with and without clinical comorbidities: Results from the cardiovascular health study. Arch Intern Med. 162:2333–2341. 2002. View Article : Google Scholar : PubMed/NCBI

94 

Kalyani RR, Varadhan R, Weiss CO, Fried LP and Cappola AR: Frailty status and altered glucose-insulin dynamics. J Gerontol A Biol Sci Med Sci. 67:1300–1306. 2012. View Article : Google Scholar : PubMed/NCBI

95 

Veronese N, Stubbs B, Fontana L, Trevisan C, Bolzetta F, De Rui M, Sartori L, Musacchio E, Zambon S, Maggi S, et al: Frailty is associated with an increased risk of incident type 2 diabetes in the elderly. J Am Med Dir Assoc. 17:902–907. 2016. View Article : Google Scholar : PubMed/NCBI

96 

Li G, Prior JC, Leslie WD, Thabane L, Papaioannou A, Josse RG, Kaiser SM, Kovacs CS, Anastassiades T, Towheed T, et al: Frailty and risk of fractures in patients with Type 2 Diabetes. Diabetes Care. 42:507–513. 2019. View Article : Google Scholar : PubMed/NCBI

97 

Tamura H, Miyamoto T, Tamaki A, Nawa G and Konya H: Osteoporosis complication is a risk factor for frailty in females with type 2 diabetes mellitus. J Phys Ther Sci. 31:621–624. 2019. View Article : Google Scholar : PubMed/NCBI

98 

Chao CT, Wang J and Chien KL; COhort of GEriatric Nephrology in NTUH (COGENT) study group, : Both pre-frailty and frailty increase healthcare utilization and adverse health outcomes in patients with type 2 diabetes mellitus. Cardiovasc Diabetol. 17:1302018. View Article : Google Scholar : PubMed/NCBI

99 

Blaum CS, Xue QL, Tian J, Semba RD, Fried LP and Walston J: Is hyperglycemia associated with frailty status in older women? J Am Geriatr Soc. 57:840–847. 2009. View Article : Google Scholar : PubMed/NCBI

100 

Kalyani RR, Tian J, Xue QL, Walston J, Cappola AR, Fried LP, Brancati FL and Blaum CS: Hyperglycemia and incidence of frailty and lower extremity mobility limitations in older women. J Am Geriatr Soc. 60:1701–1707. 2012. View Article : Google Scholar : PubMed/NCBI

101 

Zaslavsky O, Walker RL, Crane PK, Gray SL and Larson EB: glucose levels and risk of frailty. J Gerontol A Biol Sci Med Sci. 71:1223–1229. 2016. View Article : Google Scholar : PubMed/NCBI

102 

Yanagita I, Fujihara Y, Eda T, Tajima M, Yonemura K, Kawajiri T, Yamaguchi N, Asakawa H, Nei Y, Kayashima Y, et al: Low glycated hemoglobin level is associated with severity of frailty in Japanese elderly diabetes patients. J Diabetes Investig. 9:419–425. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Yau CK, Eng C, Cenzer IS, Boscardin WJ, Rice-Trumble K and Lee SJ: Glycosylated hemoglobin and functional decline in community-dwelling nursing home-eligible elderly adults with diabetes mellitus. J Am Geriatr Soc. 60:1215–1221. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2021
Volume 24 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Nishikawa H, Fukunishi S, Asai A, Yokohama K, Ohama H, Nishiguchi S and Higuchi K: Sarcopenia, frailty and type 2 diabetes mellitus (Review). Mol Med Rep 24: 854, 2021
APA
Nishikawa, H., Fukunishi, S., Asai, A., Yokohama, K., Ohama, H., Nishiguchi, S., & Higuchi, K. (2021). Sarcopenia, frailty and type 2 diabetes mellitus (Review). Molecular Medicine Reports, 24, 854. https://doi.org/10.3892/mmr.2021.12494
MLA
Nishikawa, H., Fukunishi, S., Asai, A., Yokohama, K., Ohama, H., Nishiguchi, S., Higuchi, K."Sarcopenia, frailty and type 2 diabetes mellitus (Review)". Molecular Medicine Reports 24.6 (2021): 854.
Chicago
Nishikawa, H., Fukunishi, S., Asai, A., Yokohama, K., Ohama, H., Nishiguchi, S., Higuchi, K."Sarcopenia, frailty and type 2 diabetes mellitus (Review)". Molecular Medicine Reports 24, no. 6 (2021): 854. https://doi.org/10.3892/mmr.2021.12494