Potential benefits of nutritional supplementation in diabetic sarcopenia

in Redox Experimental Medicine
Authors:
Heaji Lee Department of Food and Nutrition, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea

Search for other papers by Heaji Lee in
Current site
Google Scholar
PubMed
Close
,
Soo Jin Yang Department of Food and Nutrition, Seoul Women’s University, Seoul, Korea

Search for other papers by Soo Jin Yang in
Current site
Google Scholar
PubMed
Close
, and
Yunsook Lim Department of Food and Nutrition, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea

Search for other papers by Yunsook Lim in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-3408-8595

Correspondence should be addressed to Y Lim; Email: ylim@khu.ac.kr
Open access
Sign up for journal news

Type 2 diabetes mellitus, one of the metabolic diseases, is a major risk factor for impaired muscle function leading to muscle loss, weakness, and frailty. A lot of studies have suggested that the biological mechanisms which contribute to diabetic sarcopenia, including insulin resistance, altered energy metabolism, oxidative stress, and inflammation. Although different nutritional interventions for diabetic sarcopenia have not been clearly defined, there is no doubt that nutrition plays an essential role in the prevention or delay of muscle loss and maintenance of physical function. In this review, we discuss the recent literature on biological pathways for diabetic sarcopenia and potent nutrients used for attenuating diabetic sarcopenia: dietary proteins, omega-3 fatty acids, vitamin D, vitamin E, and other anti-oxidants for future research.

Abstract

Type 2 diabetes mellitus, one of the metabolic diseases, is a major risk factor for impaired muscle function leading to muscle loss, weakness, and frailty. A lot of studies have suggested that the biological mechanisms which contribute to diabetic sarcopenia, including insulin resistance, altered energy metabolism, oxidative stress, and inflammation. Although different nutritional interventions for diabetic sarcopenia have not been clearly defined, there is no doubt that nutrition plays an essential role in the prevention or delay of muscle loss and maintenance of physical function. In this review, we discuss the recent literature on biological pathways for diabetic sarcopenia and potent nutrients used for attenuating diabetic sarcopenia: dietary proteins, omega-3 fatty acids, vitamin D, vitamin E, and other anti-oxidants for future research.

Introduction

Diabetes mellitus (DM) is a chronic hyperglycemic condition which is caused by low production (type 1 diabetes) or inefficient use (type 2 diabetes) of insulin. Especially type 2 diabetes mellitus (T2DM) has been globally prevalent and has become a major health problem, with around 463 million population affected in 2019 and 700 million people in 2045 (Belma et al. 2019). Chronic hyperglycemic condition in DM often triggers various complications such as neuropathy, retinopathy, and nephropathy. Among various complications related to T2DM, sarcopenia has been recently high-lightened, accompanied by an increased aged population.

Sarcopenia in DM is associated with altered energy metabolism combined with increased proteolysis and impaired protein synthesis, followed by muscle damage under hyperglycemic conditions (Smith et al. 1989, Lecker et al. 1999). Furthermore, changes in energy metabolism are linked to impaired insulin signaling, increased oxidative stress, and inflammation in diabetic pathology.

Loss of muscle mass and strength can be controlled by various lifestyle factors, mainly exercise training and diet. The role of exercise in sarcopenia has been extensively investigated. Moreover, diverse nutritional approaches to improve muscle function and strength have been conducted in various research (Nikoletopoulou et al. 2013, Zhou et al. 2016). However, research focusing on dietary intervention to ameliorate diabetic sarcopenia has not been extensively reviewed (Velázquez-Alva et al. 2020). Particularly, protein supplementation, including branched-chain amino acids (BCCA), has been frequently used to improve muscle health mostly in athletes or healthy subjects (Campbell & Rains 2015). Anti-inflammatory nutrients such as omega-3 fatty acids, vitamin D, and vitamin E have been used to ameliorate sarcopenia and boost muscle functions in DM (Mostad et al. 2006, Amin et al. 2018, Lee & Lim 2018). In addition, some anti-oxidants, including resveratrol, have shown a beneficial effect on muscle damage in DM (Goh et al. 2014, Wang et al. 2018).

In this review, we include major pathological mechanisms associated with hyperglycemia-induced sarcopenia, such as insulin resistance, altered energy metabolism, oxidative stress, and inflammation in T2DM. Furthermore, we focus on the potential benefits of dietary intervention, including proteins, anti-inflammatory, and/or anti-oxidant food components on pathological mechanisms associated with diabetic sarcopenia.

Key pathogenic mechanisms in diabetic sarcopenia

We demonstrated a graphical summary of the biochemical pathways involved in diabetic sarcopenia that is highlighted in Fig. 1.

Figure 1
Figure 1

The plausible mechanisms of potential nutrients in skeletal muscle protein synthesis and degradation. Arrows represent activation and capped lines represent inhibition. Published effects of dietary proteins, omega-3, vitamin D, vitamin E, and other anti-oxidants on signaling pathways associated with diabetic sarcopenia. The binding of insulin to insulin receptor subunit-1 (IRS-1) can activate phosphoinositide 3-kinases (PI3K)/Akt signaling which stimulates mechanistic target of rapamycin (mTOR) pathway. mTOR stimulates protein synthesis by phosphorylation of p70 ribosomal S6 protein kinase (p70S6K). Akt also blocks proteolysis by phosphorylating and inhibiting forkhead transcription factors (FoxOs). The activation of FoxOs and induction of their target atrophic genes activate caspase-dependent proteolysis. 5’adenosine monophosphate-activated protein kinase (AMPK) activation following by energy deficit in skeletal muscle inhibits mTOR activation, thereby reducing protein synthesis. In addition, reactive oxygen species (ROS) and inflammatory cytokines can lead to nuclear factor-kappa B (NF-κB)-dependent upregulation of atrophic genes including Atrogin-1 and MuRF1.

Citation: Redox Experimental Medicine 2022, 1; 10.1530/REM-22-0001

Impaired muscle mass and function in DM (morphology)

A major metabolic defect associated with DM is the failure of appropriate glucose utilization in peripheral tissues such as skeletal muscle (Stanford & Goodyear 2014). Hyperglycemia can act as a powerful risk factor for loss of muscle mass and function called sarcopenia (Bassil & Gougeon 2013). In particular, progressive muscle mass loss, accompanied by declined muscle strength and quality, has been suggested as a potential factor for the link between DM and disability (Bassil & Gougeon 2013).

Human skeletal muscle fibers are classified as slow-twitch oxidative (type 1) and fast-twitch (type 2) fibers (Stuart et al. 2013). Slow-twitch fibers are more sensitive to insulin and have greater glucose uptake ability than fast-twitch fibers (Stuart et al. 2013). In diabetic patients, the fraction of slow fibers was lower than that in the healthy control subjects (Gaster et al. 2000). Furthermore, the expression level of glucose transporter 4 (GLUT4), which is higher in slow-twitch fibers, was reduced in T2DM patients (Hilton et al. 2008). The changes in fiber characteristics can reduce glucose utilization in skeletal muscle of T2DM (Stuart et al. 2013 ).

Furthermore, overweight and obesity, common characteristics in T2DM, are associated with fat infiltration into the muscle (myosteatosis) (Bianchi & Volpato 2016, Hamrick et al. 2016). Fat infiltration in the skeletal muscle can lead to abnormal muscle fiber organization and thus, affects muscle cells proliferation and differentiation (Hamrick et al. 2016). Recent data suggested that skeletal muscles of T2DM patients exhibited an increase in glycolytic fiber and a decrease in capillary density capacity (Hamrick et al. 2016). The increased production of lipid metabolites in skeletal muscle impairs the capacity for normal protein synthesis in T2DM (Bianchi& Volpato 2016). These morphological changes in muscle fiber are also associated with functional impairments in skeletal muscle, as demonstrated by muscle weakness and motor dysfunction (Punkt et al. 1999, Bianchi & Volpato 2016). Thus, the decrease in insulin sensitivity with lipid infiltration is one pathway which can directly affect skeletal muscle health in T2DM.

As a consequence, changes in muscle architecture and fiber type, with gradual loss of muscle strength, have been hypothesized as the primary biological mechanisms responsible for muscle damage in diabetic patients. The pathogenesis of diabetic sarcopenia is multifactorial and attributes to many of these causal pathways that intersect or overlap in relation to hyperglycemia in T2DM.

Altered muscle energy metabolism in DM

There are malfunctions in glucose influx into the skeletal muscle and its utilization of synthesizing ATP in insulin-resistant and diabetic conditions (Boersma et al. 2018, Chadt & Al-Hasani 2020). Subsequent energy deficit due to impaired glucose uptake probably contributes to the increased apoptosis rate in skeletal muscle (Wang et al. 2006). To compensate the energy deficit in muscle cells, protein degradation in skeletal muscle is stimulated (Park et al. 2009), subsequently accelerating the loss of muscle mass in DM (Park et al. 2009). Despite the increased protein catabolism, amino acids are not effectively utilized for ATP synthesis in DM. In normal conditions, BCCAs are catabolized by two main enzymes: BCAA transaminase (BCAT) and branched-chain α-keto acid dehydrogenase (BCKD) (Neinast et al. 2019). However, a diabetic condition often causes BCAA catabolic defects due to limited glucose availability and reduced BCKD activity (Holeček et al. 2018). The diabetes-mediated BCAA catabolic defects aggravate skeletal muscle energy deficit, contributing to muscle loss and dysfunction.

In addition, increased lipid mediates caused by impaired fat utilization in DM lead to activate skeletal muscle proteolysis (Kelley & Simoneau 1994, Pan et al. 1997, Sergi et al. 2019). Reduced β-oxidation triggers diacylglycerol and free fatty acids, which can inhibit insulin receptor subunit-1 (IRS-1) activation in DM (Pan et al. 1997). IRS can activate phosphoinositide 3-kinases (PI3Ks)/Akt signaling, which stimulates myogenesis by mammalian target of rapamycin (mTOR) activation and inhibition of FoxOs activation. Reduced activation of Akt decreases phosphorylation of FoxO, which induces the translocation of target genes and subsequently increases the transcription of muscle RING-finger1 (MuRF1) and atrophy-related ubiquitin ligases Atrogin-1/MAFbx (Meex et al. 2019).

All of the above-mentioned metabolic abnormalities with insulin resistance in skeletal muscle are responsible for the energy deficit related to muscle loss and its dysfunction in DM.

One of the key signaling molecules regulating energy metabolism is 5' adenosine monophosphate-activated protein kinase (AMPK). AMPK, a well-known energy sensor, is activated when ATP is rapidly consumed, generating high amounts of AMP. Phosphorylated AMPK increases sirtuin (SIRT) 1 activity which would result in the deacetylation of peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α) (Cork et al. 2018). PGC1α, as a master regulator of mitochondrial biogenesis, is associated with metabolic regulation such as anti-oxidant defense system and inflammation in skeletal muscle. PGC1α induces glucose uptake in the skeletal muscle by increasing the GLUT 4 translocation (Cork et al. 2018, Kou et al. 2018). In the case of T2DM, the decreased level of PGC1α causes the production of lipid metabolites which can lead to insulin resistance and aberrant energy metabolism (Waldman et al. 2018, Zhou et al. 2019, Lantier et al. 2020, Coppi et al. 2021). Therefore, the regulation of insulin signaling along with AMPK/SIRT1/PGC1α pathway is important for energy homeostasis in various tissues, including skeletal muscle.

Hyperglycemia-induced oxidative stress in skeletal muscle

Hyperglycemia is a major problem in diabetic conditions, working through several mechanisms such as increased production of reactive oxygen species (ROS) and advanced glycation end products (AGEs) (Giacco & Brownlee 2010). AGEs interact with a receptor of AGE (RAGE) present in the cell surface, thus, altering the cell signaling and gene expression. Recent studies showed that an increased level of AGEs in skeletal muscle could cause impaired muscle strength in the elderly (Giacco & Brownlee 2010, Momma et al. 2011). Furthermore, glucose auto-oxidation and protein glycosylation produce ROS, leading to oxidative stress in various tissues, followed by inflammation in DM (Giacco & Brownlee 2010, Luc et al. 2019). In particular, oxidative stress impairs insulin signaling through IRS serine/threonine phosphorylation and decreases GLUT4 translocation and then disturbs glucose uptake in skeletal muscle (Giacco & Brownlee 2010). Patients with T2DM showed excessive oxidative stress in skeletal muscle (Luc et al. 2019). Individuals with poor glycemic control causing oxidative stress also exhibited decreased muscle strength and mass (Sugimoto et al. 2019).

Furthermore, excessive ROS production is indicated as a major factor in muscle protein hyper-catabolism through the activation of the ubiquitin–proteasome pathway (Korovila et al. 2017). Oxidative stress increases the expression of FoxO3a, which activates ubiquitin–proteasome systems such as atrogin-1/MAFbx and MuRF1 in DM (Korovila et al. 2017).

Another T2DM-related factor in the progression of sarcopenia through oxidative stress is impaired mitochondrial dysfunction (Wiegman et al. 2015). As mentioned before, mitochondrial dysfunction due to the accumulation of oxidative damage may trigger apoptosis-induced muscle dysfunction (Korovila et al. 2017). It also has the capacity to dysregulate satellite cell activity that accompanies muscle damage (Xu et al. 2019). Therefore, oxidative stress interfering with muscle growth and development may cause ubiquitin and apoptosis, resulting in diabetic sarcopenia.

Hyperglycemia-induced inflammation in skeletal muscle

Chronic hyperglycemia in T2DM leads to localized inflammation, which can arise through increased immune cell infiltration in inter-myocellular and adipose tissue (AT) around the muscles (Lontchi-Yimagou et al. 2013, Wu & Ballantyne 2017). In addition, increased immune cells in visceral AT can accelerate the release of free fatty acid and migrate to myocytes, causing myocyte inflammation (Hilton et al. 2008). Increased infiltration of immune cells in skeletal muscle activates pro-inflammatory cytokines, which negatively regulate myocyte metabolic functions (Lontchi-Yimagou et al. 2013, Wu & Ballantyne 2017). Skeletal muscle secretes a variety of cytokines such as interleukin (IL)-6, IL-8, and IL-15 and other molecules such as fibroblast growth factor 21 (FGF21), irisin, myonectin, and myostatin (Bonaldo & Sandri 2013). Differentiated cultured myocytes isolated from subjects with T2DM secreted more cytokines such as tumor necrosis factor-alpha (TNF-α) and chemokines such as monocyte chemoattractant protein 1 (MCP-1) compared to those of lean controls (Pedersen et al. 2003, Abbatecola et al. 2004, Muñoz-Cánoves et al. 2013). In T2DM patients, muscle mass and strength were decreased compared to those in non-diabetic controls, and it was in accordance with the increased levels of plasma pro-inflammatory cytokines, including IL-6 and TNF-α (Tsuchiya et al. 2010, Sugimoto et al. 2019, Xu et al. 2019). These inflammatory mediators impair the insulin signaling pathway by inhibiting tyrosine phosphorylation of IRS with modifications of GLUT4 translocation in myocytes (Abbatecola et al. 2004, Tsuchiya et al. 2010, Muñoz-Cánoves et al. 2013, Sugimoto et al. 2019).

On the other hand, increased circulating pro-inflammatory cytokines directly upregulate several protein hydrolysis pathways, resulting in skeletal muscle degradation (Bonaldo & Sandri 2013, Zhou et al. 2016). Increased IL-6 activates janus kinase/signal transducers and activators of transcription (JAK/STAT) catabolic pathway. TNF-α is also the major factor that induces cellular apoptosis in muscle mainly by activating NF-κB and MyoD to cause muscle loss (Mourkioti & Rosenthal 2008, Zhou et al. 2016). NF-κB leads to activate E3 ubiquitin ligase, lysosomal-proteasome, and apoptosis which promote protein degradation in the skeletal muscle. NF-κB also regulates the expression of various pro-inflammatory mediators, which act as positive feedback to activate NF-κB itself, causing continuous muscle damage (Mourkioti & Rosenthal 2008).

These characteristics can interfere with muscle homeostasis and cell death mechanisms promoting losses in skeletal muscle mass, strength, and function, which are considered as the principal components of sarcopenia.

Potential nutritional intervention for diabetic sarcopenia

Previous research has suggested that nutrition plays a major role in diabetic sarcopenia, and nutritional interventions may prevent or reduce muscle loss and maintain physical function. Different nutritional approaches have been studied, focusing on regulating glucose homeostasis through reduced intakes of energy and saturated fat and increased intake of dietary fiber in DM (Nikoletopoulou et al. 2013, Zhou et al. 2016).

Other major pathogenic mechanisms that can be used for the prevention of diabetic sarcopenia are hyperglycemia-induced oxidative stress and inflammation. Sarcopenia is a state of increased oxidative stress and inflammation, causing apoptosis and ubiquitin-proteasome pathway in DM. Hence, we have introduced not only protein sources that help in muscle protein synthesis (MPS) but also dietary anti-inflammatory (vitamin D, omega (w)-3 fatty acids) and anti-oxidant compounds (vitamin E, ultra-trace minerals, and natural compounds) as potential nutritional interventions for diabetic sarcopenia, as summarized in Table 1.

Table 1

Summary of studies depicting cause and effect between nutrient supplementation and diabetic sarcopenia.

Nutrient Model Treatment Key findings Study
Protein Nil whey protein Adults with T2DM performed high-intensity mixed-mode interval training 20 g/day for 10 weeks. ↓ Fasting blood glucose and homeostatic model assessment of insulin resistance

↑ Repetition maximum, peak oxygen consumption, and vastus lateralis muscle thickness.
Gaffney et al. 2018
Leucine Adults with T2DM 2.5 g/day for 6 months No changes in lean tissue mass, fat percentage, muscle strength, and muscle fiber type characteristics Velázquez-Alva et al. 2020
Glutamine Diabetic mice with limb ischemia AIN-93 diet in which a part of the casein was replaced with glutamine ↑ Anti-infammatory monocytes and regulatory T cells in the blood

↓ The percentage of M1 macrophages in muscle tissues

↓ The muscle M1/M2 ratio

↓ Gene expression of IL-6

↑ The levels of PPARγ and myogenic differentiation 1 genes
Pai et al. 2020
Glutamine Diabetic rats 1 g/kg BW for 15 days ↑ Protein synthetic pathway (mTOR/Akt pathway) in skeletal muscle

↓ Protein-degradative signaling pathways (Murf1/Astrogin-1) in Skeletal Muscle
Dollet et al. 2022
Omega-3 fatty acids Fish oil (1.8 g 20:5omega-3, 3.0 g 22:6 omega-3, and 5.9 g total omega-3 fatty acids) Adults with T2DM 17.6 mL/day fish oil for 9 weeks ↓ Insulin sensitivity Alters carbohydrate and fat utilization Mostad et al. 2006
EPA C2C12 myotubes exposed to palmitate (500 μM) 30 μM ↓ Protein kinase C-θ activation

↑ Cellular acylcarnitine profile, insulin-dependent Akt phosphorylation and glucose uptake.
Whitehouse and Tisdale 2001
DHA In vitro

L6 myotubes
0.4 mmol/L ↑ Fat oxidation Aas et al. 2006
Vitamin D Vitamin D (1,25(OH)2D3) Diabetic rats 0.5 μg/kg BW three times weekly for 8 weeks ↑ Insulin sensitivity in skeletal muscles

Sustained muscle atrophy and inflammation
Amin et al. 2018
Vitamin D (1,25(OH)2D3) Obese mice 7 μg/kg, three times/week for 2 months ↑ Muscle insulin signaling Reverted myosteatosis

↓ NF-κB and tumor necrosis factor (TNF-α) activation

↓ lipogenic pathway

↓ RAGE expression in skeletal muscle.
Benetti et al. 2018
Vitamin D (1,25(OH)2D3) Obese mice 1 μg/kg/day, every day for 6 weeks ↓Skeletal muscle loss and insulin resistance in mice.

Regulated glucose homeostasis
Li et al. 2021
Vitamin E α-Tocopherol Diabetic rats 400 mg/kg BW for 21 days ↓ H2O2 levels and oxidative stress

↑ The ratio of glutathione/oxidized glutathione

↑ Protein synthesis and muscle repair
Servais et al. 2007
TRF Diabetic mice 100 mg/kg BW or 300 mg/kg for 12 weeks ↑ IRS-1, Akt, and GLUT4

↑ Mitochondrial biogenesis by activating SIRT1, SIRT3 and AMPK

↓ Oxidative stress (4-hydroxynonenal, protein carbonyls, nuclear factor erythroid 2-related factor 2, and heme oxygenase 1)

↓ Inflammation (NFκB, monocyte chemoattractant protein 1, IL-6, and TNF-α)

↓ Apoptosis (Bax, Bcl2 and caspase-3)
Lee and Lim 2018
TRF Diabetic mice 50 mg/kg BW for 2 weeks Regulates the expression of PPAR target genes

↑ Whole body glucose utilization and insulin sensitivity
Fang et al. 2010
Others RSV Diabetic mice Identical diet containing 0.04% RSV for 8 weeks ↑ Muscle function

↓ Ubiquitin and muscle MuRF-1, and LC3-II and cleaved caspase-3

↑ Mitochondrial biogenesis

↓ Mitophagy

↓ Excess mitochondrial fusion and fission
Goh et al. 2014, Wang et al. 2018
RSV Adults with T2DM 3 g/day for 12 weeks ↑ SIRT1 expression and p-AMPK to AMPK ratio Wang et al. 2018
Oligonol Diabetic mice 20 or 200 mg/kg BW for 10 weeks ↓ NF-κB expression

↑ SIRT1 expression

↓ FoxO3a nuclear localization
Bhakta et al. 2017
LFO KK-Ay mice 0, 1, or 1.5 g/kg BW for 4 weeks ↑ Femoral muscle mass

↓ Expression of MuRF1 and atrogin-1, which

↑ Activation of mTOR and p70 S6K, and phosphorylation of FoxO3a
Yoshioka et al. 2018
Ultra-trace mineral Vanadium Diabetic rats 100 mg/kg BW for 60 days ↑ The loss in body weight

↓ Blood glucose levels

↓ Antioxidant enzyme levels in serum and muscle tissue
Kurt et al. 2011

Dietary proteins

The beneficial effects of dietary proteins have been investigated, but there is insufficient research focusing on the effects of protein supplementation on diabetic sarcopenia (Campbell & Rains 2015, Velázquez-Alva et al. 2020). As protein stimulates MPS by the availability of BCAAs, it can play a central role in skeletal muscle growth. Recent studies have found that protein supplementation attenuated the decline in muscle mass and insulin resistance that potentially prevented the development of T2DM and sarcopenia (Campbell & Rains 2015, Velázquez-Alva et al. 2020). Especially, amino acids are known to induce mTOR pathway. mTOR is conserved serine/threonine kinase and regulates anabolic and catabolic signaling of skeletal muscle. mTOR pathway is activated by its upstream signaling PI3K (Ferretti et al. 2018) and then leads to an increase in S6K1 which stimulates protein synthesis (Bodine et al. 2001).

Researchers have demonstrated that various sources of dietary protein have different effects on muscle health. In a recent study, nil whey protein supplementation improved repetition maximum, peak oxygen consumption, and vastus lateralis muscle thickness in T2DM patients (Gaffney et al. 2018). Furthermore, it ameliorated glucose disposal rate, fasting blood glucose level, and homeostatic model assessment of insulin resistance (Gaffney et al. 2018).

Some previous studies showed that intake of high-quality amino acids effectively reduced muscle mass loss (Campbell & Rains 2015, Pai et al. 2020, Dollet et al. 2022). Among various amino acids, glutamine, the most abundant free amino acid in the body, is known to stimulate protein synthesis and inhibits protein degradation, thereby protecting muscle function in diabetic rats (Dollet et al. 2022, Lambertucci et al. 2022). A recent study demonstrated that glutamine supplementation attenuated insulin resistance in obesity by reducing inflammatory markers and promoting skeletal muscle insulin sensitivity (Dollet et al. 2022). Glutamine also has regulatory roles in enhancing plasma anti-inflammatory monocyte and regulatory T cells in diabetic mice (Pai et al. 2020).

Another study showed that leucine treatment induced protein synthesis by the activation of mTOR signaling (Velázquez-Alva et al. 2020). l-Leucine enhanced muscle glucose uptake with concomitant suppressed proteolytic, glycogenolytic, and gluconeogenetic activities while modulating glucose homeostasis in isolated rat psoas muscle ex vivo (Erukainure et al. 2021). However, leucine treatment did not affect muscle strength and muscle fiber type characteristics in T2DM patients (Leenders et al. 2011).

In summary, supplementation with dietary proteins and amino acids is a promising strategy to increase MPS and attenuates diabetic sarcopenia, although positive evidence was limited. Therefore, further studies are needed to establish the appropriate dosage and timing of supplementation of dietary proteins and amino acids for the management of diabetic sarcopenia.

Omega-3 fatty acids

As an anti-inflammatory agent, omega-3 fatty acids have a beneficial role in skeletal muscle metabolism and function. Omega-3 fatty acids are known to affect muscle health by regulation of the ratio between muscle protein synthesis and breakdown through decreasing inflammation (Huang et al. 2020, Okamura et al. 2020). As natural ligands for PPARγ, omega-3 fatty acids increase the activation of PPARγ, which suppresses NF-κB activation. Omega-3 fatty acids can prevent NF-κB activation and transcription of inflammatory mediators such as COX-2 and TNFα (Vanden Berghe et al. 2003, Liu et al. 2013, Calder et al. 2015). This action prevents the expression of muscle-specific E3 ligase, including MuRF1 and MAFbx/Atrogin-1 causing protein degradation in skeletal muscle (Huang et al. 2011, Ikeno et al. 2022).

In particular, eicosapentaenoic acid (EPA) treatment also enhanced mitochondrial fusion and insulin signaling by the inhibition of inflammation in human primary myotubes (Sergi et al. 2021). Treatment with EPA attenuated muscle loss by the suppression of the ubiquitin–proteasome pathway in rodent models following resistance exercise (Whitehouse & Tisdale 2001, Siriguleng et al. 2021). Another in vivo evidence shown in normal mice demonstrated that docosahexaenoic acid (DHA)-enriched diet supplementation increased skeletal muscle glucose uptake and reduced inflammation (Lam et al. 2011).

Another beneficial effect of omega-3 fatty acids on muscle health is associated with the regulation of lipid metabolism. Abnormal energy metabolism in DM leads to fat accumulation and produces lipotoxic compounds such as ceramide and palmitate, promoting insulin resistance. Recent in vitro studies reported that EPA and DHA improved fatty acid metabolism and then reduced insulin resistance in human skeletal muscle cells, C2C12, and L6 myotubes, notably via their abilities to increase mitochondrial β-oxidation (Aas et al. 2006, Capel et al. 2015, Katsnelson & Ceddia 2020) and Akt phosphorylation (Kim et al. 2016). In T2DM patients, fish oil supplementation increased fat oxidation and glucose utilization which can improve insulin sensitivity (Mostad et al. 2006). Taken together, the results suggest that omega-3 fatty acids may have protective effects against inflammation and abnormal lipid metabolism in skeletal muscle.

Although a more detailed mechanism is required to clarify the effects of omega-3 fatty acids on diabetic patients with sarcopenia, omega-3 fatty acids at physiological doses could participate in the regulation of skeletal muscle metabolism by preventing lipotoxicity and inflammation.

Vitamin D

Recently, the association between vitamin D and skeletal muscle metabolism has been highlighted. Vitamin D may affect the conservation of muscle mass and strength, preventing sarcopenia and frailty (Arik & Ulger 2016).

Administration of vitamin D accelerated the functional restoration of the damaged muscle, improved cell proliferation, and reduced cell death after muscle injury (Stratos et al. 2013). In vitamin D-deficient mice, the levels of atrogin-1 and MuRF1 in skeletal muscle were increased by two-fold compared to those of the controls (Tamura et al. 2017, Dzik & Kaczor 2019). These data demonstrate that vitamin D insufficiency/deficiency is associated with the development of muscle atrophy, but more clear mechanism is needed to be explained.

Vitamin D also regulates the insulin signaling pathway in skeletal muscle (Benetti et al. 2018). Low vitamin D level has been associated with poor glycemic control and physical function (Mirhosseini et al. 2018, Dang et al. 2019). The patients with T2DM were also associated with low level of 25(OH)D (Mendoza-Garcés et al. 2021, Takahashi et al. 2021). In a recent meta-analysis, vitamin D supplementation significantly decreased fasting plasma glucose and HbA1c levels in prediabetic individuals (Poolsup et al. 2016, Pittas et al. 2019). In T2DM rats, oral administration of vitamin D improved insulin sensitivity in skeletal muscle (Amin et al. 2018, Li et al. 2021). In another study, vitamin D restored the impaired muscle insulin signaling and reverted myosteatosis by decreasing the levels of NF-κB and TNF-α (Benetti et al. 2018). However, there are conflicting findings on vitamin D supplementation influencing glucose regulation in DM (Gulseth et al. 2017, Mousa et al. 2017).

On the other hand, vitamin D deficiency in mitochondrial dysfunction can affect the progression of sarcopenia. Mitochondria play a vital role in cellular energy metabolism, but they are also major intracellular sources of ROS (Ott et al. 2007). Ryan et al. demonstrated that 1α,25-dihydroxy vitamin D3 regulated mitochondrial oxygen consumption and dynamics in muscle cells (Ryan et al. 2016). In a recent study, vitamin D supplementation in deficient rodent models improved mitochondria's density and function and protein metabolism (Gogulothu et al. 2020). In another study, vitamin D deficiency increased oxidative stress demonstrated by increasing the expression of SOD1 in skeletal muscle mitochondria and led to lipid and protein peroxidation in patients with chronic low back pain (Dzik et al. 2018). Furthermore, vitamin D treatment protected against skeletal muscle oxidative stress by regulation of SOD and catalase in vitamin D deficiency rats (Bhat & Ismail 2015). These results support that vitamin D can attenuate oxidative stress by regulating mitochondrial ROS generation.

Recently published data indicated that the direct effects of circulating levels of vitamin D on skeletal muscle have to be connected with vitamin D receptor (VDR). VDR located in skeletal muscle plays pivotal roles in both glucose and muscle homeostasis linked to muscle health. Mice with myocyte deletion of VDR have sarcopenia and impaired muscle function (Girgis et al. 2019). Vitamin D treatment activated VDR signaling, thereby inhibiting FoxO1 expression in C2C12 muscle cells. In the previous study, the overexpression of VDR results in skeletal muscle hypertrophy by increasing anabolic signaling, ribosomal biogenesis, and protein synthesis (Bass et al. 2020).

Collectively, the beneficial effects of vitamin D on diabetic sarcopenia can be explained by several mechanisms, including improvement of anabolic/catabolic metabolism and amelioration of mitochondrial dysfunction with reduced oxidative stress as well as increased VDR signaling. Although sarcopenia might be attenuated by vitamin D treatment, a better understanding of the mechanisms is required.

Vitamin E

Vitamin E, with its anti-oxidant and anti-inflammatory properties, has known to have an important role for attenuating the progression of metabolic diseases (Momma et al. 2011, Gonzalez-Calvo et al. 2015). In particular, the potential benefits of vitamin E on muscle damage have been demonstrated in a large number of studies.

Alpha (α)-tocopherol, a predominant vitamin E in the human body, is known to improve protein synthesis and muscle repair by downregulation of oxidative stress in diabetic rats (Servais et al. 2007). Tocopherol supplementation reduced glucocorticoid-induced oxidative stress in rat skeletal muscle (Ohtsuka et al. 1998). Furthermore, α-tocopherol reduced muscle proteolysis by increasing the expression of calpains, caspases-3, -9, and -12, E3 ubiquitin ligases (MAFbx and MuRF1) with the regulation of anti-oxidant enzyme activities (SOD, CAT, GPX) in hindlimb unloading-induced muscle atrophy rodent model (Servais et al. 2007).

In addition, a number of biological properties of tocotrienol rich fraction (TRF) have been identified, such as anti-cancer, anti-diabetes, anti-oxidant, immunomodulatory, and cardio-protective properties (Aragno et al. 2004, Fang et al. 2010, Mahalingam et al. 2011, Nesaretnam et al. 2012, Vasanthi et al. 2012, Lim et al. 2019). Some studies demonstrated that TRF ameliorated oxidative stress in myoblasts (Vasanthi et al. 2012, Lim et al. 2019). In our previous study, TRF attenuated hyperglycemia-induced skeletal muscle oxidative stress demonstrated by reduced Nrf2 related pathway in diabetic mice (Lee & Lim 2018).

On the other hand, previous research has shown that vitamin E dose not only acts as an anti-oxidant but also acts as an anti-inflammatory nutrient (Huey et al. 2008, Fang et al. 2010, Chung et al. 2018). Vitamin E attenuated lipopolysaccharide-induced skeletal muscle damage by regulation of NFκB related inflammation in mice (Servais et al. 2007). TRF also inhibited inflammation by decreasing 20S proteasome activity in myoblast (Huey et al. 2008). Moreover, our group reported that TRF ameliorated NFκB and its associated inflammatory mediators in diabetic mice (Lee & Lim 2018), which attenuates the ubiquitin–proteasome system causing protein degradation (Russell et al. 2007, Qureshi et al. 2010). As inflammation can lead to loss of muscle mass and strength, these findings suggest that muscle dysfunction in diabetic patients can be attenuated by vitamin E supplementation through suppression of NF-κB associated inflammation.

Vitamin E can also ameliorate insulin resistance causing skeletal muscle damage by regulating oxidative stress and inflammation in DM. In a previous study, gamma (γ)-tocopherols ameliorated oxidative stress-induced insulin resistance in L6 myotubes (Cai et al. 2004). Furthermore, TRF improved glucose homeostasis and insulin signaling by regulation of PPAR-related pathways in the skeletal muscle of diabetic mice (Khor et al. 2017). Our group also reported that TRF normalized the insulin signaling pathway by downregulation of hyperglycemia-induced oxidative stress and inflammation in diabetic skeletal muscle (Lee & Lim 2018).

Furthermore, vitamin E supplementation can protect against mitochondrial dysfunction causing muscle proteolysis. Especially, TRF activates PPAR α, γ, and (delta) δ, which regulate mitochondrial biogenesis and energy metabolism (Singh et al. 2008, Fang et al. 2010). Previous studies have reported the ameliorative effects of vitamin E in mitochondria-mediated cell death via the release of apoptotic proteins, causing muscle wasting (Magalhães et al. 2005, 2007, Dillon et al. 2012).

Taken together, the evidence suggests that vitamin E may prevent or/and attenuate diabetic sarcopenia through amelioration of oxidative stress and inflammation accompanied by improving energy metabolism and insulin signaling pathway in skeletal muscle. However, additional investigations are required to confirm the molecular mechanism of vitamin E in protecting diabetic sarcopenia.

Others

Research continues to investigate various natural compounds and ultra-trace minerals targeting skeletal muscle to ameliorate diabetic complications. Some research in the field of diabetic sarcopenia has focused on reducing oxidative stress by anti-oxidant nutrients. In addition to vitamin E, naturally derived compounds such as polyphenols have been reported for their therapeutic potential to alleviate insulin resistance and reduce the risk of metabolic diseases. Among the prominent natural compounds, resveratrol (RSV) is one of the polyphenolic compounds known for its anti-oxidant and anti-inflammatory properties. RSV has shown preventive and therapeutic effects on metabolic diseases, including DM, obesity, and aging-associated disorders. In previous studies, RSV supplementation attenuated sarcopenia by reducing the ubiquitin–proteasome system and the mitochondrial autophagy in diabetic mice (Goh et al. 2014, Wang et al. 2018). In patients with T2DM, RSV supplementation improved energy expenditure by modulating SIRT1 expression and pAMPK/AMPK ratio in skeletal muscle (Wang et al. 2018). These observational studies suggest that RSV may have an attenuative effect on diabetic sarcopenia.

Furthermore, oligonol, mainly found in lychee fruit, consisted of catechins, procyanidins, and other phenolic compounds and is known to have an anti-oxidant effect. Oligonol treatment has been shown to attenuate ROS-related inflammation and prevent oxidative damage in in vitro model of hyperglycemia (Servais et al. 2007). In the T2DM mice model, oligonol supplementation alleviated muscle loss by suppressing atrogin-1 and MuRF1 as well as NFκB-related inflammation (Bhakta et al. 2017).

In addition to RSV and oligonol, licorice flavonoid oil (LFO) has been used for sarcopenia in diabetic mice (Yoshioka et al. 2018). Licorice, a traditional medicine, contains glabridin, which has anti-oxidant and anti-fatigue properties (Liu et al. 2017). In a recent study, LFO supplementation increased skeletal muscle mass by decreasing the expression of MuRF1 and atrogin-1 in the insulin resistance mice model (Yoshioka et al. 2018).

On the other hand, anti-oxidant effects of other ultra-trace minerals such as vanadium have been investigated. Vanadium and vanadium compounds are known to have a modulatory effect on glucose homeostasis by enhancing glucose transport, as well as IR tyrosine kinase activity (Jiang et al. 2016). In a previous study, vanadium administration decreased anti-oxidant enzyme levels, including catalase and SOD, in the skeletal muscle of diabetic mice (Kurt et al. 2011).

Overall, anti-oxidant products provide a promising benefit for inhibiting muscle mass loss since oxidative stress is one of the major risk factors. However, further clinical studies need to be conducted to clarify the beneficial effects of natural anti-oxidant compounds on diabetic sarcopenia.

Conclusions

Recently, numerous epidemiological and clinical research suggested that muscle dysfunction is more common in diabetic patients. Pathological mechanisms such as insulin resistance, abnormal energy metabolism, oxidative stress, and inflammation can all affect various components of muscle dysfunction. Figure 1 illustrates the plausible mechanisms of diabetic sarcopenia and how nutrients mitigate skeletal muscle dysfunction. Evidence suggests that different nutrients treatment can improve and maintain muscle mass and function in diabetic patients. However, more research with extensive investigations is required to establish the effective strategies of nutritional intervention on sarcopenia in DM, which can improve the quality of life of these individuals.

Summary

The prevalence of metabolic and musculoskeletal diseases, which affect individuals to mortality and morbidity, is increasing worldwide. Especially, T2DM characterized by insulin resistance negatively affects muscle health through impairments in protein metabolism, mitochondrial dysfunction, oxidative stress, and inflammation. However, evidence for the effective nutritional intervention for diabetic sarcopenia is lacking. In this review, we demonstrate the impact of various nutrients on attenuating sarcopenia in DM.

Declaration of interest

Yunsook Lim is an Editor of Redox Experimental Medicine. Yunsook Lim was not involved in the review or editorial process for this paper, on which she is listed as an author. The other authors have nothing to disclose.

Funding

This work was supported by a grant (2018R1D1A1B07046778) funded by the Ministry of Education, Science and Technology, Republic of Korea.

Author contribution statement

Y L participated in the conception and design of the work. H L, S J Y, and Y L wrote the manuscript. All authors approved the final manuscript.

References

  • Aas V, Rokling-Andersen MH, Kase ET, Thoresen GH & Rustan AC 2006 Eicosapentaenoic acid (20:5 n-3) increases fatty acid and glucose uptake in cultured human skeletal muscle cells. Journal of Lipid Research 47 366374. (https://doi.org/10.1194/jlr.M500300-JLR200)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Abbatecola AM, Ferrucci L, Grella R, Bandinelli S, Bonafè M, Barbieri M, Corsi AM, Lauretani F, Franceschi C & Paolisso G 2004 Diverse effect of inflammatory markers on insulin resistance and insulin-resistance syndrome in the elderly. Journal of the American Geriatrics Society 52 399404. (https://doi.org/10.1111/j.1532-5415.2004.52112.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Amin SN, Hussein UK, Yassa HD, Hassan SS & Rashed LA 2018 Synergistic actions of vitamin D and metformin on skeletal muscles and insulin resistance of type 2 diabetic rats. Journal of Cellular Physiology 233 57685779. (https://doi.org/10.1002/jcp.26300)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Aragno M, Mastrocola R, Catalano MG, Brignardello E, Danni O & Boccuzzi G 2004 Oxidative stress impairs skeletal muscle repair in diabetic rats. Diabetes 53 10821088. (https://doi.org/10.2337/diabetes.53.4.1082)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Arik G & Ulger Z 2016 Vitamin D in sarcopenia: understanding its role in pathogenesis, prevention and treatment. European Geriatric Medicine 7 207213. (https://doi.org/10.1016/j.eurger.2015.12.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bass JJ, Nakhuda A, Deane CS, Brook MS, Wilkinson DJ, Phillips BE, Philp A, Tarum J, Kadi F & Andersen D et al.2020 Overexpression of the vitamin D receptor (VDR) induces skeletal muscle hypertrophy. Molecular Metabolism 42 101059. (https://doi.org/10.1016/j.molmet.2020.101059)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Belma M, Suvi K, Pouya S & Paraskevi S 2019 IDF Diabetes Atlas, 9 th ed., pp. 1012. International Diabetes Foundation.(https://doi.org/10.1016/j.diabres.2019.107843)

  • Bassil MS & & Gougeon R 2013 Muscle protein anabolism in type 2 diabetes. Current opinion in clinical nutrition and metabolic care 16 8388. (https://doi.org/10.1097/MCO.0b013e32835a88ee)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Benetti E, Mastrocola R, Chiazza F, Nigro D, D'Antona G, Bordano V, Fantozzi R, Aragno M, Collino M & Minetto MA 2018 Effects of vitamin D on insulin resistance and myosteatosis in diet-induced obese mice. PLoS ONE 13 e0189707. (https://doi.org/10.1371/journal.pone.0189707)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bhakta HK, Paudel P, Fujii H, Sato A, Park CH, Yokozawa T, Jung HA & Choi JS 2017 Oligonol promotes glucose uptake by modulating the insulin signaling pathway in insulin-resistant HepG2 cells via inhibiting protein tyrosine phosphatase 1B. Archives of Pharmacal Research 40 13141327. (https://doi.org/10.1007/s12272-017-0970-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bhat M & Ismail A 2015 Vitamin D treatment protects against and reverses oxidative stress induced muscle proteolysis. Journal of Steroid Biochemistry and Molecular Biology 152 171179. (https://doi.org/10.1016/j.jsbmb.2015.05.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bianchi L & & Volpato S 2016 Muscle dysfunction in type 2 diabetes: a major threat to patient's mobility and independence. Acta Diabetol 53 879889. (https://doi.org/10.1007/s00592-016-0880-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R, Zlotchenko E, Scrimgeour A, Lawrence JC & Glass DJ et al.2001 Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nature Cell Biology 3 10141019. (https://doi.org/10.1038/ncb1101-1014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Boersma GJ, Johansson E, Pereira MJ, Heurling K, Skrtic S, Lau J, Katsogiannos P, Panagiotou G, Lubberink M, Kullberg J. 2018 Altered Glucose Uptake in Muscle, Visceral Adipose Tissue, and Brain Predict Whole-Body Insulin Resistance and may Contribute to the Development of Type 2 Diabetes: A Combined PET/MR Study. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme, 50 627639. (https://doi.org/10.1055/a-0643-4739)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bonaldo P & Sandri M 2013 Cellular and molecular mechanisms of muscle atrophy. Disease Models and Mechanisms 6 2539. (https://doi.org/10.1242/dmm.010389)

  • Cai DK, Lee KK, Li M, Tang MK & Chan KM 2004 Ubiquitin expression is up-regulated in human and rat skeletal muscles during aging. Archives of Biochemistry and Biophysics 425 4250. (https://doi.org/10.1016/j.abb.2004.02.027)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Calder PC 2015 Marine omega-3 fatty acids and inflammatory processes: effects, mechanisms and clinical relevance. Biochimica et Biophysica Acta 1851 469484. (https://doi.org/10.1016/j.bbalip.2014.08.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Campbell AP & Rains TM 2015 Dietary protein is important in the practical management of prediabetes and type 2 diabetes. Journal of Nutrition 145 164S169S. (https://doi.org/10.3945/jn.114.194878)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Capel F, Acquaviva C, Pitois E, Laillet B, Rigaudière JP, Jouve C, Pouyet C, Gladine C, Comte B & Vianey Saban C et al.2015 DHA at nutritional doses restores insulin sensitivity in skeletal muscle by preventing lipotoxicity and inflammation. Journal of Nutritional Biochemistry 26 949959. (https://doi.org/10.1016/j.jnutbio.2015.04.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chadt A & Al-Hasani H 2020 Glucose transporters in adipose tissue, liver, and skeletal muscle in metabolic health and disease. Pflugers Archiv 472 12731298 doi:10.1007/s00424-020-02417-x.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chung E, Mo H, Wang S, Zu Y, Elfakhani M, Rios SR, Chyu MC, Yang RS & Shen CL 2018 Potential roles of vitamin E in age-related changes in skeletal muscle health. Nutrition Research 49 2336. (https://doi.org/10.1016/j.nutres.2017.09.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Coppi L, Ligorio S, Mitro N, Caruso D, De Fabiani E & Crestani M 2021 M PGC1s and beyond: disentangling the complex regulation of mitochondrial and cellular metabolism. International Journal of Molecular Sciences 22 6913. (https://doi.org/10.3390/ijms22136913)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cork GK, Thompson J & Slawson C 2018 Real talk: the inter-play between the mTOR, AMPK, and hexosamine biosynthetic pathways in cell signaling. Frontiers in Endocrinology 9 522. (https://doi.org/10.3389/fendo.2018.00522)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dang M, Shore-Lorenti C, McMillan LB, Mesinovic J, Hayes A, Ebeling PR & Scott D 2019 Associations of serum 25-hydroxyvitamin D with physical performance and bone health in overweight and obese older adults. International Journal of Environmental Research and Public Health 16 509. (https://doi.org/10.3390/ijerph16030509)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dillon LM, Rebelo AP & Moraes CT 2012 The role of PGC-1 coactivators in aging skeletal muscle and heart. IUBMB Life 64 231241. (https://doi.org/10.1002/iub.608)

  • Dollet L, Kuefner M, Caria E, Rizo-Roca D, Pendergrast L, Abdelmoez AM, Karlsson HK, Dalbram E, Treebak J & Harada J et al.2022 Glutamine regulates skeletal muscle immunometabolism in type 2 diabetes. Diabetes 14 db200814.(https://doi.org/10.2337/db20-0814)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dzik KP & Kaczor JJ 2019 Mechanisms of vitamin D on skeletal muscle function: oxidative stress, energy metabolism and anabolic state. European Journal of Applied Physiology 119 825839. (https://doi.org/10.1007/s00421-019-04104-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dzik K, Skrobot W, Flis DJ, Karnia M, Libionka W, Kloc W & Kaczor JJ 2018 Vitamin D supplementation attenuates oxidative stress in paraspinal skeletal muscles in patients with low back pain. European Journal of Applied Physiology 118 143151. (https://doi.org/10.1007/s00421-017-3755-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Erukainure OL, Salau VF, Atolani O, Ravichandran R, Banerjee P, Preissner R, Koorbanally NA & Islam MS 2021 L-leucine stimulation of glucose uptake and utilization involves modulation of glucose – lipid metabolic switch and improved bioenergetic homeostasis in isolated rat psoas muscle ex vivo. Amino Acids 53 11351151. (https://doi.org/10.1007/s00726-021-03021-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fang F, Kang Z & Wong C 2010 Vitamin E tocotrienols improve insulin sensitivity through activating peroxisome proliferator-activated receptors. Molecular Nutrition and Food Research 54 345352. (https://doi.org/10.1002/mnfr.200900119)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ferretti R, Moura EG, Dos Santos VC, Caldeira EJ, Conte M, Matsumura CY, Pertille A & Mosqueira M 2018 High-fat diet suppresses the positive effect of creatine supplementation on skeletal muscle function by reducing protein expression of IGF-PI3K-AKT-mTOR pathway. PLoS ONE 13 e0199728. (https://doi.org/10.1371/journal.pone.0199728)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gaffney KA, Lucero A, Stoner L, Faulkner J, Whitfield P, Krebs J & Rowlands DS 2018 Nil whey protein effect on glycemic control after intense mixed-mode training in type 2 diabetes. Medicine and Science in Sports and Exercise 50 1117. (https://doi.org/10.1249/MSS.0000000000001404)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gaster M, Poulsen P, Handberg A, Schroder HD & Beck-Nielsen H 2000 Direct evidence of fiber type-dependent GLUT-4 expression in human skeletal muscle. American Journal of Physiology: Endocrinology and Metabolism 278 E910E916. (https://doi.org/10.1152/ajpendo.2000.278.5.E910)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Giacco F & Brownlee M 2010 Oxidative stress and diabetic complications. Circulation Research 107 10581070. (https://doi.org/10.1161/CIRCRESAHA.110.223545)

  • Girgis CM, Cha KM, So B, Tsang M, Chen J, Houweling PJ, Schindeler A, Stokes R, Swarbrick MM & Evesson FJ et al.2019 Mice with myocyte deletion of vitamin D receptor have sarcopenia and impaired muscle function. Journal of Cachexia, Sarcopenia and Muscle 10 12281240. (https://doi.org/10.1002/jcsm.12460)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gogulothu R, Nagar D, Gopalakrishnan S, Garlapati VR, Kallamadi PR & Ismail A 2020 Disrupted expression of genes essential for skeletal muscle fibre integrity and energy metabolism in vitamin D deficient rats. Journal of Steroid Biochemistry and Molecular Biology 197 105525. (https://doi.org/10.1016/j.jsbmb.2019.105525)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Goh KP, Lee HY, Lau DP, Supaat W, Chan YH & Koh AF 2014 Effects of resveratrol in patients with type 2 diabetes mellitus on skeletal muscle SIRT1 expression and energy expenditure. International Journal of Sport Nutrition and Exercise Metabolism 24 213. (https://doi.org/10.1123/ijsnem.2013-0045)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gonzalez-Calvo L, Joy M, Blanco M, Dervishi E, Molino F, Sarto P, Ripoll G, Serrano M & Calvo JH 2015 Effect of vitamin E supplementation or alfalfa grazing on fatty acid composition and expression of genes related to lipid metabolism in lambs. Journal of Animal Science 93 30443054. (https://doi.org/10.2527/jas.2014-8758)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gulseth HL, Wium C, Angel K, Eriksen EF & Birkeland KI 2017 Effects of vitamin D supplementation on insulin sensitivity and insulin secretion in subjects with type 2 diabetes and vitamin D deficiency: a randomized controlled trial. Diabetes Care 40 872878. (https://doi.org/10.2337/dc16-2302)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hamrick MW, McGee-Lawrence ME & Frechette DM 2016 Fatty infiltration of skeletal muscle: mechanisms and comparisons with bone marrow adiposity. Frontiers in Endocrinology 7 69. (https://doi.org/10.3389/fendo.2016.00069)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hilton TN, Tuttle LJ, Bohnert KL, Mueller MJ & Sinacore DR 2008 Excessive adipose tissue infiltration in skeletal muscle in individuals with obesity, diabetes mellitus, and peripheral neuropathy: association with performance and function. Physical Therapy 88 13361344. (https://doi.org/10.2522/ptj.20080079)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Holeček M 2018 Branched-chain amino acids in health and disease: metabolism, alterations in blood plasma, and as supplements. Nutrition and Metabolism 15 33. (https://doi.org/10.1186/s12986-018-0271-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang F, Wei H, Luo H, Jiang S & Peng J 2011 EPA inhibits the inhibitor of κBα (IκBα)/NF-κB/muscle ring finger 1 pathway in C2C12 myotubes in a PPARγ-dependent manner. British Journal of Nutrition 105 348356. (https://doi.org/10.1017/S0007114510003703)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang YH, Chiu WC, Hsu YP, Lo YL & Wang YH 2020 Effects of omega-3 fatty acids on muscle mass, muscle strength and muscle performance among the elderly: a meta-analysis. Nutrients 12 3739. (https://doi.org/10.3390/nu12123739)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huey KA, Fiscus G, Richwine AF, Johnson RW & Meador BM 2008 In vivo vitamin E administration attenuates interleukin-6 and interleukin-1beta responses to an acute inflammatory insult in mouse skeletal and cardiac muscle. Experimental Physiology 93 12631272. (https://doi.org/10.1113/expphysiol.2008.043190)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ikeno Y, Inomata M, Tsukimura Y, Suzuki Y, Takeuchi H, Harada Y, Kon R, Ikarashi N, Chiba Y & Yamada T et al.2022 Eicosapentaenoic acid suppresses cisplatin-induced muscle atrophy by attenuating the up-regulated gene expression of ubiquitin. Journal of Nutritional Biochemistry 103 108953. (https://doi.org/10.1016/j.jnutbio.2022.108953)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jiang P, Dong Z, Ma B, Ni Z, Duan H, Li X, Wang B, Ma X, Wei Q & Ji X et al.2016 Effect of vanadyl rosiglitazone, a new insulin-mimetic vanadium complexes, on glucose homeostasis of diabetic mice. Applied Biochemistry and Biotechnology 180 841851. (https://doi.org/10.1007/s12010-016-2137-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Katsnelson G & Ceddia RB 2020 Docosahexaenoic and eicosapentaenoic fatty acids differentially regulate glucose and fatty acid metabolism in L6 rat skeletal muscle cells. American Journal of Physiology: Cell Physiology 319 C1120C1129. (https://doi.org/10.1152/ajpcell.00304.2020)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kelley DE & Simoneau JA 1994 Impaired free fatty acid utilization by skeletal muscle in non-insulin-dependent diabetes mellitus. Journal of Clinical Investigation 94 23492356. (https://doi.org/10.1172/JCI117600)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Khor SC, Wan Ngah WZ, Mohd Yusof YA, Abdul Karim N & Makpol S 2017 Tocotrienol-rich fraction ameliorates antioxidant defense mechanisms and improves replicative senescence-associated oxidative stress in human myoblasts. Oxidative Medicine and Cellular Longevity 2017 3868305. (https://doi.org/10.1155/2017/3868305)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim J, Carlson ME, Kuchel GA, Newman JW & Watkins BA 2016 Dietary DHA reduces downstream endocannabinoid and inflammatory gene expression and epididymal fat mass while improving aspects of glucose use in muscle in C57BL/6J mice. International Journal of Obesity 40 129137. (https://doi.org/10.1038/ijo.2015.135)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Korovila I, Hugo M, Castro JP, Weber D, Höhn A, Grune T & Jung T 2017 Proteostasis, oxidative stress and aging. Redox Biology 13 550567. (https://doi.org/10.1016/j.redox.2017.07.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kou G, Li Z, Wu C, Liu Y, Hu Y, Guo L, Xu X & Zhou Z 2018 Citrus tangeretin improves skeletal muscle mitochondrial biogenesis via activating the AMPK-PGC1-α pathway in vitro and in vivo: a possible mechanism for its beneficial effect on physical performance. Journal of Agricultural and Food Chemistry 66 1191711925. (https://doi.org/10.1021/acs.jafc.8b04124)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kurt O, Ozden TY, Ozsoy N, Tunali S, Can A, Akev N & Yanardag R 2011 Influence of vanadium supplementation on oxidative stress factors in the muscle of STZ-diabetic rats. Biometals 24 943949. (https://doi.org/10.1007/s10534-011-9452-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lam YY, Hatzinikolas G, Weir JM, Janovská A, McAinch AJ, Game P, Meikle PJ & Wittert GA 2011 Insulin-stimulated glucose uptake and pathways regulating energy metabolism in skeletal muscle cells: the effects of subcutaneous and visceral fat, and long-chain saturated, n-3 and n-6 polyunsaturated fatty acids. Biochimica et Biophysica Acta 1811 468475. (https://doi.org/10.1016/j.bbalip.2011.04.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lambertucci AC, Lambertucci RH, Hirabara SM, Curi R, Moriscot AS, Alba-Loureiro TC, Guimarães-Ferreira L, Levada-Pires AC, Vasconcelos DA & Sellitti DF et al.2012 Glutamine supplementation stimulates protein-synthetic and inhibits protein-degradative signaling pathways in skeletal muscle of diabetic rats. PLoS ONE 7 e50390. (https://doi.org/10.1371/journal.pone.0050390)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lantier L, Williams AS, Williams IM, Guerin A, Bracy DP, Goelzer M, Foretz M, Viollet B, Hughey CC & Wasserman DH 2020 Reciprocity between skeletal muscle AMPK deletion and insulin action in diet-induced obese mice. Diabetes 69 16361649. (https://doi.org/10.2337/db19-1074)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lecker SH, Salomon V, Price SR, Kwon YT, Mitch WE & Goldberg AL 1999 Ubiquitin conjugation by the N-end rule pathway and mRNAs for its components increase in muscles of diabetic rats. Journal of Clinical Investigation 104 14111420. (https://doi.org/10.1172/JCI7300)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee H & Lim Y 2018 Tocotrienol-rich fraction supplementation reduces hyperglycemia-induced skeletal muscle damage through regulation of insulin signaling and oxidative stress in type 2 diabetic mice. Journal of Nutritional Biochemistry 57 7785. (https://doi.org/10.1016/j.jnutbio.2018.03.016)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leenders M, Verdijk LB, van der Hoeven L, van Kranenburg J, Hartgens F, Wodzig WK, Saris WH & van Loon LJ 2011 Prolonged leucine supplementation does not augment muscle mass or affect glycemic control in elderly type 2 diabetic men. Journal of Nutrition 141 10701076. (https://doi.org/10.3945/jn.111.138495)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li A, Shen P, Liu S, Wang J, Zeng J & Du C 2021 Vitamin D alleviates skeletal muscle loss and insulin resistance by inducing vitamin D receptor expression and regulating the AMPK/SIRT1 signaling pathway in mice. Food Science and Technologyv 42 e47921.(https://doi.org/10.1590/fst.47921)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lim JJ, Wan Zurinah WN, Mouly V & Norwahidah AK 2019 Tocotrienol-rich fraction (TRF) treatment promotes proliferation capacity of stress-induced premature senescence myoblasts and modulates the renewal of satellite cells: microarray analysis. Oxidative Medicine and Cellular Longevity 2019 9141343. (https://doi.org/10.1155/2019/9141343)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu HQ, Qiu Y, Mu Y, Zhang XJ, Liu L, Hou XH, Zhang L, Xu XN, Ji AL & Cao R et al.2013 A high ratio of dietary n-3/n-6 polyunsaturated fatty acids improves obesity-linked inflammation and insulin resistance through suppressing activation of TLR4 in SD rats. Nutrition Research 33 849858. (https://doi.org/10.1016/j.nutres.2013.07.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu HW, Chen YJ, Chang YC & Chang SJ 2017 Oligonol, a low-molecular weight polyphenol derived from lychee, alleviates muscle loss in diabetes by suppressing atrogin-1 and MuRF1. Nutrients 9 1040. (https://doi.org/10.3390/nu9091040)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lontchi-Yimagou E, Sobngwi E, Matsha TE & Kengne AP 2013 Diabetes mellitus and inflammation. Current Diabetes Reports 13 435444. (https://doi.org/10.1007/s11892-013-0375-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Luc K, Schramm-Luc A, Guzik TJ & Mikolajczyk TP 2019 Oxidative stress and inflammatory markers in prediabetes and diabetes. Journal of Physiology and Pharmacology 70 809824. (https://doi.org/10.26402/jpp.2019.6.01)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Magalhães J, Ascensão A, Soares JMC, Ferreira R, Neuparth MJ, Marques F & Duarte JA 2005 Acute and severe hypobaric hypoxia increases oxidative stress and impairs mitochondrial function in mouse skeletal muscle. Journal of Applied Physiology 99 12471253. (https://doi.org/10.1152/japplphysiol.01324.2004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Magalhães J, Ferreira R, Neuparth MJ, Oliveira PJ, Marques F & Ascensão A 2007 Vitamin E prevents hypobaric hypoxiainducedmitochondrial dysfunction in skeletalmuscle. Clinical Science 113 59466. (https://doi.org/10.1042/CS20070075)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mahalingam D, Radhakrishnan AK, Amom Z, Ibrahim N & Nesaretnam K 2011 Effects of supplementation with tocotrienol-rich fraction on immune response to tetanus toxoid immunization in normal healthy volunteers. European Journal of Clinical Nutrition 65 6369. (https://doi.org/10.1038/ejcn.2010.184)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Meex RCR, Blaak EE & van Loon LJC 2019 Lipotoxicity plays a key role in the development of both insulin resistance and muscle atrophy in patients with type 2 diabetes. Obesity Reviews 20 12051217. (https://doi.org/10.1111/obr.12862)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mendoza-Garcés L, Velázquez-Alva MC, Cabrer-Rosales MF, Arrieta-Cruz I, Gutiérrez-Juárez R & Irigoyen-Camacho ME 2021 Vitamin D deficiency is associated with handgrip strength, nutritional status and T2DM in community-dwelling older Mexican women: a cross-sectional study. Nutrients 13 736. (https://doi.org/10.3390/nu13030736)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mirhosseini N, Vatanparast H, Mazidi M & Kimball SM 2018 Vitamin D supplementation, glycemic control, and insulin resistance in prediabetics: a meta-analysis. Journal of the Endocrine Society 2 687709. (https://doi.org/10.1210/js.2017-00472)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Momma H, Niu K, Kobayashi Y, Guan L, Sato M, Guo H, Chujo M, Otomo A, Yufei C & Tadaura H et al.2011 Skin advanced glycation end product accumulation and muscle strength among adult men. European Journal of Applied Physiology 111 15451552. (https://doi.org/10.1007/s00421-010-1779-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mostad IL, Bjerve KS, Bjorgaas MR, Lydersen S & Grill V 2006 Effects of n-3 fatty acids in subjects with type 2 diabetes: reduction of insulin sensitivity and time-dependent alteration from carbohydrate to fat oxidation. American Journal of Clinical Nutrition 84 540550. (https://doi.org/10.1093/ajcn/84.3.540)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mourkioti F & Rosenthal N 2008 NF-kappaB signaling in skeletal muscle: prospects for intervention in muscle diseases. Journal of Molecular Medicine 86 747759. (https://doi.org/10.1007/s00109-008-0308-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mousa A, Naderpoor N, de Courten MP, Teede H, Kellow N, Walker K, Scragg R & de Courten B 2017 Vitamin D supplementation has no effect on insulin sensitivity or secretion in vitamin D-deficient, overweight or obese adults: a randomized placebo-controlled trial. American Journal of Clinical Nutrition 105 13721381. (https://doi.org/10.3945/ajcn.117.152736)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Muñoz-Cánoves P, Scheele C, Pedersen BK & Serrano AL 2013 Interleukin-6 myokine signaling in skeletal muscle: a double-edged sword? FEBS Journal 280 41314148. (https://doi.org/10.1111/febs.12338)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Neinast M, Murashige D & Arany Z 2019 Branched chain amino acids. Annual Review of Physiology 81 139164. (https://doi.org/10.1146/annurev-physiol-020518-114455)

  • Nesaretnam K, Meganathan P, Veerasenan SD & Selvaduray KR 2012 Tocotrienols and breast cancer: the evidence to date. Genes and Nutrition 7 39. (https://doi.org/10.1007/s12263-011-0224-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nikoletopoulou V, Markaki M, Palikaras K & Tavernarakis N 2013 Crosstalk between apoptosis, necrosis and autophagy. Biochimica et biophysica acta 1833 34483459. (https://doi.org/10.1016/j.bbamcr.2013.06.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ohtsuka A, Kojima H, Ohtani T & Hayashi K 1998 Vitamin E reduces glucocorticoid-induced oxidative stress in rat skeletal muscle. Journal of Nutritional Science and Vitaminology 44 779786. (https://doi.org/10.3177/jnsv.44.779)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Okamura T, Hashimoto Y, Miki A, Kaji A, Sakai R, Iwai K, Osaka T, Ushigome E, Hamaguchi M & Yamazaki M et al.2020 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 66 233237. (https://doi.org/10.3164/jcbn.19-85)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ott M, Gogvadze V, Orrenius S & Zhivotovsky B 2007 Mitochondria, oxidative stress and cell death. Apoptosis 12 913922. (https://doi.org/10.1007/s10495-007-0756-2)

  • Pai MH, Lei CS, Su ST, Yeh SL & Hou YC 2020 Effects of dietary glutamine supplementation on immune cell polarization and muscle regeneration in diabetic mice with limb ischemia. European Journal of Nutrition 59 921933. (https://doi.org/10.1007/s00394-019-01951-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pan DA, Lillioja S, Kriketos AD, Milner MR, Baur LA, Bogardus C, Jenkins AB & Storlien LH 1997 Skeletal muscle triglyceride levels are inversely related to insulin action. Diabetes 46 983988. (https://doi.org/10.2337/diab.46.6.983)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Park SW, Goodpaster BH, Lee JS, Kuller LH, Boudreau R, de Rekeneire N, Harris TB, Kritchevsky S, Tylavsky FA & Nevitt M et al.2009 Excessive loss of skeletal muscle mass in older adults with type 2 diabetes. Diabetes Care 32 19931997. (https://doi.org/10.2337/dc09-0264)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pedersen M, Bruunsgaard H, Weis N, Hendel HW, Andreassen BU, Eldrup E, Dela F & Pedersen BK 2003 Circulating levels of TNF-alpha and IL-6-relation to truncal fat mass and muscle mass in healthy elderly individuals and in patients with type-2 diabetes. Mechanisms of Ageing and Development 124 495502. (https://doi.org/10.1016/s0047-6374(0300027-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pittas AG, Dawson-Hughes B, Sheehan P, Ware JH, Knowler WC, Aroda VR, Brodsky I, Ceglia L, Chadha C & Chatterjee R et al.2019 Vitamin D supplementation and prevention of type 2 diabetes. New England Journal of Medicine 381 520530. (https://doi.org/10.1056/NEJMoa1900906)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Poolsup N, Suksomboon N & Plordplong N 2016 Effect of vitamin D supplementation on insulin resistance and glycaemic control in prediabetes: a systematic review and meta-analysis. Diabetic Medicine 33 290299. (https://doi.org/10.1111/dme.12893)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Punkt K, Psinia I, Welt K, Barth W & Asmussen G 1999 Effects on skeletal muscle fibres of diabetes and Ginkgo biloba extract treatment. Acta Histochemica 101 5369. (https://doi.org/10.1016/S0065-1281(9980008-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qureshi AA, Reis JC, Papasian CJ, Morrison DC & Qureshi N 2010 Tocotrienols inhibit lipopolysaccharide-induced pro-inflammatory cytokines in macrophages of female mice. Lipids in Health and Disease 9 143. (https://doi.org/10.1186/1476-511X-9-143)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Russell ST, Eley H & Tisdale MJ 2007 Role of reactive oxygen species in protein degradation in murine myotubes induced by proteolysis-inducing factor and angiotensin II. Cellular Signalling 19 17971806. (https://doi.org/10.1016/j.cellsig.2007.04.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ryan ZC, Craig TA, Folmes CD, Wang X, Lanza IR, Schaible NS, Salisbury JL, Nair KS, Terzic A & Sieck GC et al.2016 1α,25-Dihydroxyvitamin D3 regulates mitochondrial oxygen consumption and dynamics in human skeletal muscle cells. Journal of Biological Chemistry 291 15141528. (https://doi.org/10.1074/jbc.M115.684399)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sergi D, Naumovski N, Heilbronn LK, Abeywardena M, O'Callaghan N, Lionetti L & Luscombe-Marsh N 2019 Mitochondrial (Dys)function and insulin resistance: From pathophysiological molecular mechanisms to the impact of diet. Frontiers in Physiology 10 532. (https://doi.org/10.3389/fphys.2019.00532)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sergi D, Luscombe-Marsh N, Heilbronn LK, Birch-Machin M, Naumovski N, Lionetti L, Proud CG, Abeywardena MY & O'Callaghan N 2021 The inhibition of metabolic inflammation by EPA is associated with enhanced mitochondrial fusion and insulin signaling in human primary myotubes. Journal of Nutrition 151 810819. (https://doi.org/10.1093/jn/nxaa430)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Servais S, Letexier D, Favier R, Duchamp C & Desplanches D 2007 Prevention of unloading-induced atrophy by vitamin E supplementation: links between oxidative stress and soleus muscle proteolysis? Free Radical Biology and Medicine 42 627635. (https://doi.org/10.1016/j.freeradbiomed.2006.12.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Siriguleng S, Koike T, Natsume Y, Jiang H, Mu L & Oshida Y 2021 Eicosapentaenoic acid enhances skeletal muscle hypertrophy without altering the protein anabolic signaling pathway. Physiological Research 70 5565. (https://doi.org/10.33549/physiolres.934534)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Smith OL, Wong CY & Gelfand RA 1989 Skeletal muscle proteolysis in rats with acute streptozotocin-induced diabetes. Diabetes 38 11171122. (https://doi.org/10.2337/diab.38.9.1117)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stanford KI & Goodyear LJ 2014 Exercise and type 2 diabetes: molecular mechanisms regulating glucose uptake in skeletal muscle. Advances in Physiology Education 38 308314. (https://doi.org/10.1152/advan.00080.2014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stratos I, Li Z, Herlyn P, Rotter R, Behrendt AK, Mittlmeier T & Vollmar B 2013 Vitamin D increases cellular turnover and functionally restores the skeletal muscle after crush injury in rats. American Journal of Pathology 182 895904. (https://doi.org/10.1016/j.ajpath.2012.11.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stuart CA, McCurry MP, Marino A, South MA, Howell ME, Layne AS, Ramsey MW & Stone MH 2013 Slow-twitch fiber proportion in skeletal muscle correlates with insulin responsiveness. Journal of Clinical Endocrinology and Metabolism 98 20272036. (https://doi.org/10.1210/jc.2012-3876)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sugimoto K, Tabara Y, Ikegami H, Takata Y, Kamide K, Ikezoe T, Kiyoshige E, Makutani Y, Onuma H & Gondo Y et al.2019 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. Journal of Diabetes Investigation 10 14711479. (https://doi.org/10.1111/jdi.13070)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Takahashi F, Hashimoto Y, Kaji A, Sakai R, Kawate Y, Okamura T, Kondo Y, Fukuda T, Kitagawa N & Okada H et al.2021 Vitamin intake and loss of muscle mass in older people with type 2 diabetes: a prospective study of the KAMOGAWA-DM cohort. Nutrients 13 2335. (https://doi.org/10.3390/nu13072335)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tamura Y, Fujito H, Kawao N & Kaji H 2017 Vitamin D deficiency aggravates diabetes-induced muscle wasting in female mice. Diabetology International 8 5258. (https://doi.org/10.1007/s13340-016-0278-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tsuchiya Y, Hatakeyama H, Emoto N, Wagatsuma F, Matsushita S & Kanzaki M 2010 Palmitate-induced down-regulation of sortilin and impaired GLUT4 trafficking in C2C12 myotubes. Journal of Biological Chemistry 285 3437134381. (https://doi.org/10.1074/jbc.M110.128520)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vanden Berghe W, Vermeulen L, Delerive P, De Bosscher K, Staels B & Haegeman G 2003 A paradigm for gene regulation: inflammation, NF-kappaB and PPAR. Advances in Experimental Medicine and Biology 544 181196. (https://doi.org/10.1007/978-1-4419-9072-3_22)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vasanthi HR, Parameswari RP & Das DK 2012 Multifaceted role of tocotrienols in cardioprotection supports their structure: function relation. Genes and Nutrition 7 1928. (https://doi.org/10.1007/s12263-011-0227-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Velázquez-Alva MC, Irigoyen-Camacho ME, Zepeda-Zepeda MA, Lazarevich I, Arrieta-Cruz I & D’Hyver C 2020 Sarcopenia, nutritional status and type 2 diabetes mellitus: a cross-sectional study in a group of Mexican women residing in a nursing home. Nutrition and Dietetics 77 515522. (https://doi.org/10.1111/1747-0080.12551)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Waldman M, Cohen K, Yadin D, Nudelman V, Gorfil D, Laniado-Schwartzman M, Kornwoski R, Aravot D, Abraham NG & Arad M et al.2018 Regulation of diabetic cardiomyopathy by caloric restriction is mediated by intracellular signaling pathways involving 'SIRT1 and PGC-1α'. Cardiovascular Diabetology 17 111. (https://doi.org/10.1186/s12933-018-0754-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang X, Hu Z, Hu J, Du J & Mitch WE 2006 Insulin resistance accelerates muscle protein degradation: activation of the ubiquitin-proteasome pathway by defects in muscle cell signaling. Endocrinology 147 41604168. (https://doi.org/10.1210/en.2006-0251)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang D, Sun H, Song G, Yang Y, Zou X, Han P & Li S 2018 Resveratrol improves muscle atrophy by modulating mitochondrial quality control in STZ-induced diabetic mice. Molecular Nutrition and Food Research 62 e1700941. (https://doi.org/10.1002/mnfr.201700941)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Whitehouse AS & Tisdale MJ 2001 Downregulation of ubiquitin-dependent proteolysis by eicosapentaenoic acid in acute starvation. Biochemical and Biophysical Research Communications 285 598602. (https://doi.org/10.1006/bbrc.2001.5209)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wiegman CH, Michaeloudes C, Haji G, Narang P, Clarke CJ, Russell KE, Bao W, Pavlidis S, Barnes PJ & Kanerva J et al.2015 Oxidative stress-induced mitochondrial dysfunction drives inflammation and airway smooth muscle remodeling in patients with chronic obstructive pulmonary disease. Journal of Allergy and Clinical Immunology 136 769780. (https://doi.org/10.1016/j.jaci.2015.01.046)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wu H & Ballantyne CM 2017 Skeletal muscle inflammation and insulin resistance in obesity. Journal of Clinical Investigation 127 4354. (https://doi.org/10.1172/JCI88880)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xu D, Jiang Z, Sun Z, Wang L, Zhao G, Hassan HM, Fan S, Zhou W, Han S & Zhang L et al.2019 Mitochondrial dysfunction and inhibition of myoblast differentiation in mice with high-fat-diet-induced pre-diabetes. Journal of Cellular Physiology 234 75107523. (https://doi.org/10.1002/jcp.27512)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yoshioka Y, Yamashita Y, Kishida H, Nakagawa K & Ashida H 2018 Licorice flavonoid oil enhances muscle mass in KK-Ay mice. Life Sciences 205 9196. (https://doi.org/10.1016/j.lfs.2018.05.024)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhou J, Liu B, Liang C, Li Y & Song YH 2016 Cytokine signaling in skeletal muscle wasting. Trends in Endocrinology and Metabolism 27 335347. (https://doi.org/10.1016/j.tem.2016.03.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhou J, Poudel A, Chandramani-Shivalingappa P, Xu B, Welchko R & Li L 2019 Liraglutide induces beige fat development and promotes mitochondrial function in diet induced obesity mice partially through AMPK-SIRT-1-PGC1-α cell signaling pathway. Endocrine 64 271283. (https://doi.org/10.1007/s12020-018-1826-7)

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
Get Permissions
  • Figure 1

    The plausible mechanisms of potential nutrients in skeletal muscle protein synthesis and degradation. Arrows represent activation and capped lines represent inhibition. Published effects of dietary proteins, omega-3, vitamin D, vitamin E, and other anti-oxidants on signaling pathways associated with diabetic sarcopenia. The binding of insulin to insulin receptor subunit-1 (IRS-1) can activate phosphoinositide 3-kinases (PI3K)/Akt signaling which stimulates mechanistic target of rapamycin (mTOR) pathway. mTOR stimulates protein synthesis by phosphorylation of p70 ribosomal S6 protein kinase (p70S6K). Akt also blocks proteolysis by phosphorylating and inhibiting forkhead transcription factors (FoxOs). The activation of FoxOs and induction of their target atrophic genes activate caspase-dependent proteolysis. 5’adenosine monophosphate-activated protein kinase (AMPK) activation following by energy deficit in skeletal muscle inhibits mTOR activation, thereby reducing protein synthesis. In addition, reactive oxygen species (ROS) and inflammatory cytokines can lead to nuclear factor-kappa B (NF-κB)-dependent upregulation of atrophic genes including Atrogin-1 and MuRF1.

  • Aas V, Rokling-Andersen MH, Kase ET, Thoresen GH & Rustan AC 2006 Eicosapentaenoic acid (20:5 n-3) increases fatty acid and glucose uptake in cultured human skeletal muscle cells. Journal of Lipid Research 47 366374. (https://doi.org/10.1194/jlr.M500300-JLR200)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Abbatecola AM, Ferrucci L, Grella R, Bandinelli S, Bonafè M, Barbieri M, Corsi AM, Lauretani F, Franceschi C & Paolisso G 2004 Diverse effect of inflammatory markers on insulin resistance and insulin-resistance syndrome in the elderly. Journal of the American Geriatrics Society 52 399404. (https://doi.org/10.1111/j.1532-5415.2004.52112.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Amin SN, Hussein UK, Yassa HD, Hassan SS & Rashed LA 2018 Synergistic actions of vitamin D and metformin on skeletal muscles and insulin resistance of type 2 diabetic rats. Journal of Cellular Physiology 233 57685779. (https://doi.org/10.1002/jcp.26300)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Aragno M, Mastrocola R, Catalano MG, Brignardello E, Danni O & Boccuzzi G 2004 Oxidative stress impairs skeletal muscle repair in diabetic rats. Diabetes 53 10821088. (https://doi.org/10.2337/diabetes.53.4.1082)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Arik G & Ulger Z 2016 Vitamin D in sarcopenia: understanding its role in pathogenesis, prevention and treatment. European Geriatric Medicine 7 207213. (https://doi.org/10.1016/j.eurger.2015.12.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bass JJ, Nakhuda A, Deane CS, Brook MS, Wilkinson DJ, Phillips BE, Philp A, Tarum J, Kadi F & Andersen D et al.2020 Overexpression of the vitamin D receptor (VDR) induces skeletal muscle hypertrophy. Molecular Metabolism 42 101059. (https://doi.org/10.1016/j.molmet.2020.101059)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Belma M, Suvi K, Pouya S & Paraskevi S 2019 IDF Diabetes Atlas, 9 th ed., pp. 1012. International Diabetes Foundation.(https://doi.org/10.1016/j.diabres.2019.107843)

  • Bassil MS & & Gougeon R 2013 Muscle protein anabolism in type 2 diabetes. Current opinion in clinical nutrition and metabolic care 16 8388. (https://doi.org/10.1097/MCO.0b013e32835a88ee)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Benetti E, Mastrocola R, Chiazza F, Nigro D, D'Antona G, Bordano V, Fantozzi R, Aragno M, Collino M & Minetto MA 2018 Effects of vitamin D on insulin resistance and myosteatosis in diet-induced obese mice. PLoS ONE 13 e0189707. (https://doi.org/10.1371/journal.pone.0189707)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bhakta HK, Paudel P, Fujii H, Sato A, Park CH, Yokozawa T, Jung HA & Choi JS 2017 Oligonol promotes glucose uptake by modulating the insulin signaling pathway in insulin-resistant HepG2 cells via inhibiting protein tyrosine phosphatase 1B. Archives of Pharmacal Research 40 13141327. (https://doi.org/10.1007/s12272-017-0970-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bhat M & Ismail A 2015 Vitamin D treatment protects against and reverses oxidative stress induced muscle proteolysis. Journal of Steroid Biochemistry and Molecular Biology 152 171179. (https://doi.org/10.1016/j.jsbmb.2015.05.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bianchi L & & Volpato S 2016 Muscle dysfunction in type 2 diabetes: a major threat to patient's mobility and independence. Acta Diabetol 53 879889. (https://doi.org/10.1007/s00592-016-0880-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R, Zlotchenko E, Scrimgeour A, Lawrence JC & Glass DJ et al.2001 Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nature Cell Biology 3 10141019. (https://doi.org/10.1038/ncb1101-1014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Boersma GJ, Johansson E, Pereira MJ, Heurling K, Skrtic S, Lau J, Katsogiannos P, Panagiotou G, Lubberink M, Kullberg J. 2018 Altered Glucose Uptake in Muscle, Visceral Adipose Tissue, and Brain Predict Whole-Body Insulin Resistance and may Contribute to the Development of Type 2 Diabetes: A Combined PET/MR Study. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme, 50 627639. (https://doi.org/10.1055/a-0643-4739)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bonaldo P & Sandri M 2013 Cellular and molecular mechanisms of muscle atrophy. Disease Models and Mechanisms 6 2539. (https://doi.org/10.1242/dmm.010389)

  • Cai DK, Lee KK, Li M, Tang MK & Chan KM 2004 Ubiquitin expression is up-regulated in human and rat skeletal muscles during aging. Archives of Biochemistry and Biophysics 425 4250. (https://doi.org/10.1016/j.abb.2004.02.027)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Calder PC 2015 Marine omega-3 fatty acids and inflammatory processes: effects, mechanisms and clinical relevance. Biochimica et Biophysica Acta 1851 469484. (https://doi.org/10.1016/j.bbalip.2014.08.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Campbell AP & Rains TM 2015 Dietary protein is important in the practical management of prediabetes and type 2 diabetes. Journal of Nutrition 145 164S169S. (https://doi.org/10.3945/jn.114.194878)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Capel F, Acquaviva C, Pitois E, Laillet B, Rigaudière JP, Jouve C, Pouyet C, Gladine C, Comte B & Vianey Saban C et al.2015 DHA at nutritional doses restores insulin sensitivity in skeletal muscle by preventing lipotoxicity and inflammation. Journal of Nutritional Biochemistry 26 949959. (https://doi.org/10.1016/j.jnutbio.2015.04.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chadt A & Al-Hasani H 2020 Glucose transporters in adipose tissue, liver, and skeletal muscle in metabolic health and disease. Pflugers Archiv 472 12731298 doi:10.1007/s00424-020-02417-x.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chung E, Mo H, Wang S, Zu Y, Elfakhani M, Rios SR, Chyu MC, Yang RS & Shen CL 2018 Potential roles of vitamin E in age-related changes in skeletal muscle health. Nutrition Research 49 2336. (https://doi.org/10.1016/j.nutres.2017.09.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Coppi L, Ligorio S, Mitro N, Caruso D, De Fabiani E & Crestani M 2021 M PGC1s and beyond: disentangling the complex regulation of mitochondrial and cellular metabolism. International Journal of Molecular Sciences 22 6913. (https://doi.org/10.3390/ijms22136913)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cork GK, Thompson J & Slawson C 2018 Real talk: the inter-play between the mTOR, AMPK, and hexosamine biosynthetic pathways in cell signaling. Frontiers in Endocrinology 9 522. (https://doi.org/10.3389/fendo.2018.00522)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dang M, Shore-Lorenti C, McMillan LB, Mesinovic J, Hayes A, Ebeling PR & Scott D 2019 Associations of serum 25-hydroxyvitamin D with physical performance and bone health in overweight and obese older adults. International Journal of Environmental Research and Public Health 16 509. (https://doi.org/10.3390/ijerph16030509)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dillon LM, Rebelo AP & Moraes CT 2012 The role of PGC-1 coactivators in aging skeletal muscle and heart. IUBMB Life 64 231241. (https://doi.org/10.1002/iub.608)

  • Dollet L, Kuefner M, Caria E, Rizo-Roca D, Pendergrast L, Abdelmoez AM, Karlsson HK, Dalbram E, Treebak J & Harada J et al.2022 Glutamine regulates skeletal muscle immunometabolism in type 2 diabetes. Diabetes 14 db200814.(https://doi.org/10.2337/db20-0814)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dzik KP & Kaczor JJ 2019 Mechanisms of vitamin D on skeletal muscle function: oxidative stress, energy metabolism and anabolic state. European Journal of Applied Physiology 119 825839. (https://doi.org/10.1007/s00421-019-04104-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dzik K, Skrobot W, Flis DJ, Karnia M, Libionka W, Kloc W & Kaczor JJ 2018 Vitamin D supplementation attenuates oxidative stress in paraspinal skeletal muscles in patients with low back pain. European Journal of Applied Physiology 118 143151. (https://doi.org/10.1007/s00421-017-3755-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Erukainure OL, Salau VF, Atolani O, Ravichandran R, Banerjee P, Preissner R, Koorbanally NA & Islam MS 2021 L-leucine stimulation of glucose uptake and utilization involves modulation of glucose – lipid metabolic switch and improved bioenergetic homeostasis in isolated rat psoas muscle ex vivo. Amino Acids 53 11351151. (https://doi.org/10.1007/s00726-021-03021-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fang F, Kang Z & Wong C 2010 Vitamin E tocotrienols improve insulin sensitivity through activating peroxisome proliferator-activated receptors. Molecular Nutrition and Food Research 54 345352. (https://doi.org/10.1002/mnfr.200900119)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ferretti R, Moura EG, Dos Santos VC, Caldeira EJ, Conte M, Matsumura CY, Pertille A & Mosqueira M 2018 High-fat diet suppresses the positive effect of creatine supplementation on skeletal muscle function by reducing protein expression of IGF-PI3K-AKT-mTOR pathway. PLoS ONE 13 e0199728. (https://doi.org/10.1371/journal.pone.0199728)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gaffney KA, Lucero A, Stoner L, Faulkner J, Whitfield P, Krebs J & Rowlands DS 2018 Nil whey protein effect on glycemic control after intense mixed-mode training in type 2 diabetes. Medicine and Science in Sports and Exercise 50 1117. (https://doi.org/10.1249/MSS.0000000000001404)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gaster M, Poulsen P, Handberg A, Schroder HD & Beck-Nielsen H 2000 Direct evidence of fiber type-dependent GLUT-4 expression in human skeletal muscle. American Journal of Physiology: Endocrinology and Metabolism 278 E910E916. (https://doi.org/10.1152/ajpendo.2000.278.5.E910)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Giacco F & Brownlee M 2010 Oxidative stress and diabetic complications. Circulation Research 107 10581070. (https://doi.org/10.1161/CIRCRESAHA.110.223545)

  • Girgis CM, Cha KM, So B, Tsang M, Chen J, Houweling PJ, Schindeler A, Stokes R, Swarbrick MM & Evesson FJ et al.2019 Mice with myocyte deletion of vitamin D receptor have sarcopenia and impaired muscle function. Journal of Cachexia, Sarcopenia and Muscle 10 12281240. (https://doi.org/10.1002/jcsm.12460)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gogulothu R, Nagar D, Gopalakrishnan S, Garlapati VR, Kallamadi PR & Ismail A 2020 Disrupted expression of genes essential for skeletal muscle fibre integrity and energy metabolism in vitamin D deficient rats. Journal of Steroid Biochemistry and Molecular Biology 197 105525. (https://doi.org/10.1016/j.jsbmb.2019.105525)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Goh KP, Lee HY, Lau DP, Supaat W, Chan YH & Koh AF 2014 Effects of resveratrol in patients with type 2 diabetes mellitus on skeletal muscle SIRT1 expression and energy expenditure. International Journal of Sport Nutrition and Exercise Metabolism 24 213. (https://doi.org/10.1123/ijsnem.2013-0045)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gonzalez-Calvo L, Joy M, Blanco M, Dervishi E, Molino F, Sarto P, Ripoll G, Serrano M & Calvo JH 2015 Effect of vitamin E supplementation or alfalfa grazing on fatty acid composition and expression of genes related to lipid metabolism in lambs. Journal of Animal Science 93 30443054. (https://doi.org/10.2527/jas.2014-8758)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gulseth HL, Wium C, Angel K, Eriksen EF & Birkeland KI 2017 Effects of vitamin D supplementation on insulin sensitivity and insulin secretion in subjects with type 2 diabetes and vitamin D deficiency: a randomized controlled trial. Diabetes Care 40 872878. (https://doi.org/10.2337/dc16-2302)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hamrick MW, McGee-Lawrence ME & Frechette DM 2016 Fatty infiltration of skeletal muscle: mechanisms and comparisons with bone marrow adiposity. Frontiers in Endocrinology 7 69. (https://doi.org/10.3389/fendo.2016.00069)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hilton TN, Tuttle LJ, Bohnert KL, Mueller MJ & Sinacore DR 2008 Excessive adipose tissue infiltration in skeletal muscle in individuals with obesity, diabetes mellitus, and peripheral neuropathy: association with performance and function. Physical Therapy 88 13361344. (https://doi.org/10.2522/ptj.20080079)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Holeček M 2018 Branched-chain amino acids in health and disease: metabolism, alterations in blood plasma, and as supplements. Nutrition and Metabolism 15 33. (https://doi.org/10.1186/s12986-018-0271-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang F, Wei H, Luo H, Jiang S & Peng J 2011 EPA inhibits the inhibitor of κBα (IκBα)/NF-κB/muscle ring finger 1 pathway in C2C12 myotubes in a PPARγ-dependent manner. British Journal of Nutrition 105 348356. (https://doi.org/10.1017/S0007114510003703)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang YH, Chiu WC, Hsu YP, Lo YL & Wang YH 2020 Effects of omega-3 fatty acids on muscle mass, muscle strength and muscle performance among the elderly: a meta-analysis. Nutrients 12 3739. (https://doi.org/10.3390/nu12123739)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huey KA, Fiscus G, Richwine AF, Johnson RW & Meador BM 2008 In vivo vitamin E administration attenuates interleukin-6 and interleukin-1beta responses to an acute inflammatory insult in mouse skeletal and cardiac muscle. Experimental Physiology 93 12631272. (https://doi.org/10.1113/expphysiol.2008.043190)