Next Article in Journal
Characterization of Fruit Sorbet Matrices with Added Value from Zizyphus jujuba and Stevia rebaudiana
Next Article in Special Issue
Health Effects of Whole Grains: A Bibliometric Analysis
Previous Article in Journal
Effect of Cephalosporin Antibiotics on the Activity of Yoghurt Cultures
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bioactive Components in Whole Grains for the Regulation of Skeletal Muscle Function

China-Canada Joint Lab of Food Nutrition and Health, Beijing Technology & Business University, Beijing 100048, China
*
Author to whom correspondence should be addressed.
Foods 2022, 11(18), 2752; https://doi.org/10.3390/foods11182752
Submission received: 25 July 2022 / Revised: 30 August 2022 / Accepted: 2 September 2022 / Published: 7 September 2022

Abstract

:
Skeletal muscle plays a primary role in metabolic health and physical performance. Conversely, skeletal muscle dysfunctions such as muscular dystrophy, atrophy and aging-related sarcopenia could lead to frailty, decreased independence and increased risk of hospitalization. Dietary intervention has become an effective approach to improving muscle health and function. Evidence shows that whole grains possess multiple health benefits compared with refined grains. Importantly, there is growing evidence demonstrating that bioactive substances derived from whole grains such as polyphenols, γ-oryzanol, β-sitosterol, betaine, octacosanol, alkylresorcinols and β-glucan could contribute to enhancing myogenesis, muscle mass and metabolic function. In this review, we discuss the potential role of whole-grain-derived bioactive components in the regulation of muscle function, emphasizing the underlying mechanisms by which these compounds regulate muscle biology. This work will contribute toward increasing awareness of nutraceutical supplementation of whole grain functional ingredients for the prevention and treatment of muscle dysfunctions.

1. Introduction

Whole grains refer to the whole, milled into powdered, fragmented or pressed pieces of cereal glume, whose proportion of endosperm, germ and bran is essentially the same as that in intact glume [1]. It has become evident that whole grains and whole-grain-based products possess enormous nutritional and health-promoting properties due to the germ and bran fractions, which contains unique bioactive components (dietary fiber, β-glucan, phenolics, carotenoids and tocotrienol) compared to refined grains [2,3]. Scientific studies showed that the components derived from whole grains exhibit positive modulation on skeletal muscle function. Based on established research, polyphenols extracted from wheat bran, such as alkylresorcinol [4] and ferulic acid [5], could alleviate muscle atrophy caused by obesity. The intake of more dietary fiber is beneficial for promoting muscle mass by improving the composition and metabolic capacity of the gut microbiota [6]. This evidence indicates the potential benefit of bioactive components in whole grains on skeletal muscle function. In addition, the emergence of sports nutrition foods demonstrates the beneficial effect of bioactive components in whole grains. Sports nutrition supplements with bioactive ingredients such as γ-oryzanol and octacosanol have gained increased attention in the functional food market and attracted growing interest especially in the elderly population and athletes [7]. For instance, γ-oryzanol has been commercially approved for enhancing the physical fitness of athletes, used to improve capacity to resist muscle fatigue and enhance muscle strength [8]. Functional drinks and capsules supplemented with octacosanol (0.07 mg/mL and 51 mg/kg, respectively) are shown to exert anti-fatigue, reaction sensitivity improvement and physical recovery effects on athletes [9].
Skeletal muscle dysregulation occurs in chronic syndrome caused by obesity and aging, which is characterized by inflammatory reaction, poor mitochondrial function and muscle protein loss [10]. Muscle tissue function depends on the growth and differentiation of myotubes, the transformation of muscle fiber types and mitochondrial function [11,12,13]. The reduction in myotube size will lead to skeletal muscle atrophy, while the improvement in muscle fiber cross-sectional area (CSA) is positively associated with muscle strength and mass [14]. Generally, skeletal muscle is composed of slow-twitch myofibers (types I and IIa) and fast-twitch myofibers (types IIb and IIx). These two muscle types are the basis of muscle plasticity in response to various functional demands [15]. The change in isoforms of myosin heavy chain (MyHC) serves as an important sign for muscle-fiber-type transformation [16]. Exercise and disuse of muscle could stimulate muscle-fiber-type transformation [17]. In addition, mitochondria are involved in the regulation of muscle metabolic function through adaptive changes in mitochondrial quantity, structure and quality [18]. The occurrence of muscle dysfunction is found to be directly related to the impairment of the mitochondrial network [19]. Taken together, these factors mentioned above have been implicated in the pathogenesis of muscle-related diseases, including muscular dystrophy, atrophy and aging-related sarcopenia (Figure 1).
Muscular dystrophy (MD) is characterized by atrophy and weakness of muscle tissue, resulting in degeneration and gradual aggravation of muscles controlling movement. Mitochondrial biogenesis dysfunction and decreased ATP production are important inducements of muscular dystrophy [20]. AMPK has emerged as a key regulator of muscular dystrophy by regulating mitochondrial autophagy and biogenesis-related proteins including PGC-1a, NRF1, NRF-2/GABP and SIRT1 [21]. Multiple regulators can modulate skeletal muscle metabolism through activation of AMPK among which FNIP1 is a key regulatory protein that regulates mitochondrial function, muscle-fiber-type transformation and skeletal muscle health [22]. FNIP1 knockdown could lead to the activation of AMPK and enhancement of mitochondria number, alleviating the symptom of muscular dystrophy [23]. In addition, the decrease in autophagy flux could also cause muscular dystrophy, while AMPK-activated autophagy is reported to be beneficial for improving myofiber structure and function in mice with muscular dystrophy [24]. Moreover, AMPK could also be modulated by mTOR signaling via phosphorylation, which inhibits autophagosome-wrapping-damaged organelles and the process of lysosome-caused degradation [25].
Skeletal muscle atrophy is the consequence of muscle protein degradation exceeding protein synthesis [26]. The destruction of myogenesis and decrease in CSA caused by excessive autophagy are two main reasons for muscle atrophy. SIRT2 is reported to modulate skeletal muscle atrophy by inhibiting excessive autophagy [27] and improve myotube differentiation and hypertrophy [28]. Enhanced mitochondrial function is conducive to improving muscle atrophy [29], while the reduction of mitochondrial biogenesis and oxidative phosphorylation leads to the production of ROS, which causes excessive muscle protein degradation [30]. Mitochondrial transcription factor A (TFAM) binds and coats mtDNA, protecting it from ROS-induced degradation and maintaining mitochondrial function [26]. On the other hand, muscle atrophy is accompanied by slow-to-fast shift in muscle fiber type [31]. The overexpression of FOXO3a could stimulate this process, while PGC-1α could alleviate muscle atrophy through inhibiting the expression of FOXO3a [31]. In addition, TGF-β is mediated by Smad3/ERK-dependent signaling to regulate myotube proliferation and differentiation. It has been reported that TGF-β could inhibit the level of myosin heavy chain (MyHC) and decrease muscle CSA, resulting in skeletal muscle atrophy [32].
Sarcopenia is characterized by a loss of muscle mass and function that reduces mobility. As reported, aging-induced sarcopenia is characterized by the preferential loss of fast-twitch muscle fibers and transformation from type II (fast-twitch muscle fibers) to type I muscle fibers (slow-twitch muscle fibers) [33]. The dysfunction of myotube regeneration and lysosomal degradation are also involved in sarcopenia [34]. It has been proven that activation and overexpression of SIRT1 reduces skeletal muscle aging. SIRT1 directly phosphorylates and enhances the activity of PGC-1α, which targets the hypoxia-inducible factor 2 α (HIF2α) to control muscle fiber transformation [35]. In addition, early growth response 1 (EGR1) has been reported to bind with SIRT1 to improve oxidative-stress-impaired myotube differentiation [36]. Systemic inflammation is one of the main causes of skeletal muscle loss and sarcopenia. For instance, NF-κB signaling is activated in aging people which induces an inflammatory effect and the production of ROS in mitochondria. Moreover, excessive autophagy is aggravated by NF-κB signaling, which causes the degradation of muscle protein [37].
Emerging evidence suggests that nutritional intervention is helpful to improve myotube proliferation, regulate myofiber-type transformation and prevent or delay muscle protein degradation, benefiting muscle mass and metabolic function. In this review, we summarize the bioactive components derived from whole grains (Table 1) and explore the potential role of whole grain bioactive components in improving skeletal muscle function, and further discuss the possible regulatory mechanisms.

2. Whole Grain Bioactive Compounds in the Regulation of Skeletal Muscle Function

Whole grain consumption has been demonstrated to have an impressive effect on chronic disease prevention, such as type 2 diabetes, cardiovascular disease, cancer and muscle disease associated with obesity and aging [1,38]. Herein, we discuss the evidence which supports the role of whole grain functional ingredients in regulating muscle health through their ability to promote myogenesis, affect muscle-fiber-type conversion, reduce muscle protein loss and enhance muscle metabolic function (Table 2 and Figure 2).

2.1. Phenolic Compounds

2.1.1. Phenolic Acids

Ferulic acid is a phenolic acid widespread in whole grains such as oat [39], wheat [40], rice [41], quinoa [42] and corn [43], which is evidenced to possess an impressive effect on modulating muscle function. It has been reported to prevent insulin resistance in skeletal muscle [44]. It can also promote glucose uptake and regulate fatty acid oxidative decomposition in a dose-dependent manner, promoting redox balance in rat muscle [5]. By triggering the PI3K/CpkC pathway, ferulic acid is proved to promote glucose uptake and glycogen synthesis, protecting against insulin resistance in L6 myotubes [45]. After 30 days of dietary ferulic acid supplementation, the number and CSA of muscle fiber are enhanced in a zebrafish model. Ferulic acid also accelerated the muscle tissue growth of zebrafish by upregulating the expression of MyoD and myogenin [46]. In addition, ferulic acid is proven to modulate PGC-1α-activated mitogenesis and energy metabolism via upregulating the expression of NRF1, TFAM and TFB2M [47]. In C2C12 cells, ferulic acid exerts an anti-fatigue function via increasing succinate dehydrogenase (SDH) activity and decreasing lactate dehydrogenase (LDH) activity. Moreover, ferulic acid could stimulate the transformation from fast-twitch muscle fibers to slow-twitch muscle fibers via the SIRT1/AMPK signaling pathway [48]. This effect is further verified in a weaned piglets model, demonstrating that SIRT1/AMPK/PGC-1α signaling was involved in ferulic-acid-mediated muscle transformation and anti-fatigue function [49]. This evidence demonstrates the positive effect of ferulic acid on muscle metabolism and function.
p-coumaric acid is widely distributed in whole grains including rice [50], quinoa [42], barley [51] and wheat [40]. It has been found that p-coumaric acids possess the ability to regulate myogenic differentiation and lipid metabolism of skeletal muscle. For instance, p-coumaric acid could increase the expression of myogenic regulatory factors myogenin and MyoD via activating AMPK signaling, improving myogenic differentiation of C2C12 cells [52]. In the L6 cell model, p-coumaric acid could increase fatty acid β-oxidation through stimulating ACC phosphorylation and the expression of PPARγ, preventing skeletal muscle against oxidative stress and inflammation caused by fatty acid accumulation [53].

2.1.2. Resveratrol

Resveratrol is a natural source of antioxidant and mitochondrial nutrients which can be found in whole grains, such as buckwheat [54]. Resveratrol shows a positive effect on mitochondrial-dysfunction-induced muscle atrophy. For instance, in a dexamethasone-induced mouse model, resveratrol alleviates dexamethasone-induced mitochondrial dysfunction and muscle atrophy by blocking AMPK/FOXO3 signaling [55]. In high-fat-diet-induced aged mice, muscle loss and myofiber size decrease are reversed by resveratrol supplementation. Resveratrol could improve the mitochondrial dysfunction and oxidative-stress-induced muscle atrophy via activation of the PKA/LKB1/AMPK pathway [56]. Moreover, resveratrol has been reported to enhance muscle proliferation and differentiation in obese mouse muscle. The level of muscle regeneration proteins including MyoG, Myf5 and Pax7 is upregulated by resveratrol administration [57]. In response to skeletal-muscle glucose metabolism disorders caused by obesity, resveratrol can reduce insulin resistance by increasing muscle glycogen synthesis and reducing ROS levels in high-fat-diet-fed mice [58]. In the C2C12 model, resveratrol promotes AKT activation and glucose absorption, as well as decreased glutathione (GSH) level. These results suggest that resveratrol could alleviate muscle insulin resistance by modulating redox levels [59]. Resveratrol is evidenced to alleviate TNF-α-induced muscle hypertrophy and muscle atrophy in C2C12 cells via inhibiting the atrophy-related ubiquitin ligase through upregulating AKT/mTOR/FOXO1 signaling [60]. Furthermore, resveratrol displays impressive activity in promoting the transformation of muscle fiber types. The expression of myosin heavy chain (MyHC) 1, MyHC2a and MyHC2x in mouse muscle is increased with resveratrol supplementation, indicating the transformation from fast- to slow-twitch muscle fibers. This conversion is achieved by activating the AdiopR1-AMPK-PGC-1α signaling pathway [61]. In C2C12 myotubes, resveratrol supplementation increased the activities of lactate dehydrogenase (LDH) and malate dehydrogenase (MDH) while it reversed the elevated level of miR-22-3p, suggesting its anti-fatigue effect via stimulating the transformation of muscle fiber from fast-twitch to slow-twitch type [62]. In addition, resveratrol exerts an anti-fatigue effect in contusion-induced-injury mice, shown as the increased activity of lactate dehydrogenase (LDH) and creatine kinase (CK), displaying an important role in muscle metabolic regulation [63].

2.1.3. Flavonoids

As a special subclass of flavonoids, quercetin is a natural bioactive compound abundantly present in common cereal grains, especially in buckwheat [64], followed by rice [50], quinoa [42], corn [43] and oat [39]. Quercetin has displayed notable antioxidative and anti-inflammatory effects and shows potential benefits for muscle function [65]. By increasing the expression of adiponectin, quercetin stimulates muscle-fiber-type transformation from fast-twitch to slow-twitch myofibers [66]. It has been also demonstrated that quercetin increases slow-twitch myofibers by regulating AMPK/SIRT1 signaling [67]. Heme oxygenase-1 (HO-1) serves as an essential factor to decrease the inflammatory response and oxidative stress. Mediated by an HO-1/NRF2-dependent mechanism, quercetin is found to alleviate obesity-induced muscle atrophy [68]. In relation to the mitochondrial network, quercetin has been shown to significantly improve mitogenesis in denervated mice, through which muscle atrophy is improved [69]. Moreover, it is shown that quercetin ameliorates TNF-α-induced skeletal muscle insulin resistance in C2C12 cells [70]. The anti-fatigue function of quercetin might be related to its ability to scavenge free radicals [71]. Overloaded exercise causes excessive ROS generation, which further leads to mitochondrial dysfunction and muscle protein loss [72]. In a BALB/c mouse model, quercetin displayed obvious fatigue resistance activity, shown as enhanced muscle glycogen content, elevated mitochondrial fatty acid β-oxidation and decreased oxidative stress [73]. These findings demonstrate the impressive effect of quercetin on improving muscle mitochondrial function and alleviating the inflammatory reaction in the muscle.
Oligomeric procyanidins (OPCs) are mainly found in dark-colored grains, such as black rice and red rice [74]. As reported, OPCs could effectively inhibit glucose metabolism disorder in muscle tissue caused by diabetes. OPCs are shown to improve lipid metabolism by inhibiting mTOR signaling and enhancing glucose homeostasis and insulin sensitivity in the skeletal muscle of diabetic mice [75]. In obese mice, OPC supplementation increases glycogen synthesis and glucose uptake, exerting an antidiabetic effect in an insulin-independent manner [76]. Moreover, oral OPC administration alleviates acute hyperglycemia and improves insulin sensitivity via promoting AMPK-signaling-mediated GLUT4 translocation in ICR mice [77].
Cyanidin-3-glucoside (Cy3G) widely exists in pigmented whole grains, such as black rice [50], purple corn [43], oat, wheat and rye [78]. In vivo and in vitro studies have shown that Cy3G possesses anti-obesity and antidiabetic effects [79,80]. It has been demonstrated that as effective radical scavengers, Cy3G exerts an anti-diabetes effect via reducing oxidative stress in human skeletal muscle cells [81]. In an ICR mouse model, oral supplementation with Cy3G shows improved exercise endurance, performed as weight-loaded swimming time. Moreover, the increased level of lactic acid accumulated after excessive exercise is reversed by Cy3G via activating PGC-1α signaling, indicating that Cy3G exerts its anti-fatigue effect through modulating the PGC-1α-mediated lactic acid metabolism [82]. Consistent with the in vivo data, Cy3G could also upregulate PGC-1α expression in C2C12 myotubes, improving muscle metabolic function [82].
Catechins are flavanols widely found in whole grains such as wheat [83], barley and buckwheat [84]. The isomers of catechins including catechin, epicatechin (EC) and epigallocatechin gallate (EGCG) are proved to increase muscle mass and muscle strength, enhancing the exercise endurance of rats [85]. Catechins could upregulate the expression of Myf5 in muscle stem cells via activating Akt phosphorylation and thus improve muscle regeneration [86]. In the C2C12 cell model, catechin supplementation was found to promote myotube differentiation, shown as an increased level of MyoD, MyoG and MyHC, suggesting its improvement effect on skeletal muscle regeneration and repair [87]. Moreover, catechins have shown impressive modulation of the promotion of mitochondrial function in skeletal muscle. In an LCR (low running capacity) rat model, EC treatment improved mitochondrial biogenesis and respiratory capacity via activating p38 MAPK signaling in the skeletal muscle, enhancing muscle resistance to fatigue [88]. Using a diabetic rat model, EGCG supplementation could alleviate diabetes-induced mitochondrial dysfunction in skeletal muscle, characterized by increased expression of PGC-1α, MFN2 and COXⅠ. The improvement effect of catechins is possibly achieved by promoting mitochondrial autophagy via activating the ROS-ERK/JNK-p53 pathway [89]. Moreover, EGCG could target Wnt signaling and increase the level of downstream MyoD, promoting muscle regeneration [90]. In a rat model of sarcopenia, EGCG administration could increase skeletal muscle mass which is presumably attributed to the downregulated muscle protein degradation mediated by the ubiquitin–proteasome system (UPS) and the upregulated muscle protein synthesis mediated by IGF-1 [91]. Moreover, EGCG inhibits the expression of oxidatively modified protein induced by electrical stimulation in rat skeletal muscle, preventing the muscle from oxidative damage [92]. These data suggest that catechin could facilitate skeletal muscle function through promoting myotube differentiation, enhancing mitochondrial function and defending oxidative stress.
Rutin is a flavonol glycoside and is widely distributed in whole grains such as buckwheat [84], wheat [93] and quinoa [94]. The antioxidant and anti-inflammatory properties of rutin are demonstrated from both in vivo and in vitro studies [95]. The decreased mitochondrial number and mitochondrial biogenesis in skeletal muscle are associated with obesity [96]. In an obese rat model, rutin supplementation significantly enhances mitochondrial DNA (mtDNA) content and mitochondrial biogenesis in skeletal muscle by activating AMPK signaling. The expression levels of PGC-1α, NRF1, TFAM and SIRT1 are also increased, indicating that rutin could restore obesity-induced skeletal muscle mitochondrial dysfunction [97]. Using a forced swimming mouse model, rutin administration was proved to upregulate PGC-1α-mediated mitochondrial biogenesis and decrease the level of lactic acid in skeletal muscle, improving the fatigue-resistance capacity of mouse muscle [98]. In addition, rutin was found to inhibit oxidative stress and inflammation-induced muscle injury. Specifically, rutin intervention improves the inflammatory state by inhibiting NF-κB signaling, manifested in the decreased expression of IL-6 and iNOS, preventing muscle injury of C2C12 cells [99].

2.2. Carotenoids

Carotenoids are lipid-soluble phytochemicals and the inactive precursor of vitamin A [100], which are abundantly distributed in corn, wheat and barley [101]. As the main component of carotenoids, lutein is reported to improve muscle function via alleviating oxidative stress [102]. In a rat model of ischemia-reperfusion (IR) muscle injury, lutein supplementation decreases the production of ROS and the expression of COX-2 by inhibiting NF-κB signaling, protecting against skeletal muscle IR injury [103]. These observations imply the muscle improvement benefit of lutein presumably via its antioxidant and anti-inflammatory effect.
β-carotenes are the most ubiquitous and stable natural pigments in nature [104]. β-carotene supplementation could enhance muscle mass by promoting IGF-1-mediated muscle protein synthesis and reducing ubiquitin–protease-mediated muscle protein degradation in an atrophy mouse model [105]. Atrogin-1 and MuRF1 are identified as muscle-specific ubiquitin ligases that are upregulated in skeletal muscle under atrophy-inducing conditions [106]. β-carotene treatment is reported to increase muscle mass and decrease the level of Atrogin-1 and MuRF1 in denervated mice, exhibiting a promotion effect on oxidative-stress-induced muscle atrophy [107].

2.3. Tocotrienol

Tocotrienol, the unsaturated form of vitamin E, mainly exists in buckwheat [108], rice, rye and oat [39]. Tocotrienol receives attention due to its unique biological properties such as antioxidant and anti-inflammatory activity [109]. Increasing evidence has shown that tocopherol exhibits a positive effect on maintaining muscle function. It has been proven that dietary supplementation of tocotrienol improves muscle atrophy and insulin resistance in type 2 diabetic mice. This effect is achieved by activating AMPK/SIRT1/PGC-1α signaling and increasing mitochondrial biogenesis in the skeletal muscle, indicating that tocotrienol improves muscle disorder via upregulating mitochondrial function [110]. In addition, muscle differentiation and regeneration are essential for protecting against aging-related loss of muscle mass and strength. Tocotrienol treatment is found to increase the expression of myoblast-differentiation-related proteins such as MyoD, MyoG and MRF4. Moreover, tocotrienol increased the miR-206 level in human skeletal muscle myoblasts (HSMMs), which resulted in an increased expression of IGF1R and decreased expression of Pax7, promoting muscle cell differentiation [111]. Through transcriptomic analysis, tocotrienol was found to promote muscle cell regeneration via Wnt signaling and alleviate aging-related muscle loss through the FOXO pathway [112]. Additionally, Chung et al. reported that tocotrienol administration improved insulin intolerance and increased soleus muscle weight of obese mice, indicating its potential to ameliorate muscle dysfunction [113].

2.4. β-Glucan

β-glucan is a soluble dietary fiber which exists abundantly in oat [39], rice [114], wheat, barley and rye [108]. β-glucan has been evidenced to have a positive effect on muscle health [115]. For example, dietary supplementation with oat β-glucan is found to decrease the activity of lactate dehydrogenase and creatine kinase in serum, while it increases the glycogen content in the gastrocnemius muscle of rats. These results indicate that oat β-glucan facilitates the recovery of trained rats from fatigue [116]. In addition, β-glucan is found to promote the transformation of fast-twitch muscle fibers to slow-twitch type in the C2C12 model. Moreover, the levels of myoblast proliferation markers such as Myf5 and Mox2 are increased by β-glucan treatment, suggesting the anti-fatigue effect via improving muscle fiber proliferation and transformation [117]. Mitochondria are responsible for providing energy during immune action and the enhancement of mitochondrial function is conducive to preventing oxidative stress. In the Duchenne muscular dystrophy (DMD) zebrafish model, β-glucan intervention improves mitochondrial respiration and zebrafish exercise capacity, suggesting that β-glucan could alleviate the symptoms of DMD through regulating mitochondria function [118].

2.5. γ-Oryzanol

γ-oryzanol is a mixture of feruloyl esters with triterpenol as the main component and is mainly found in rice, corn and wheat [119,120]. γ-oryzanol supplementation has been evidenced to improve exercise performance such as grip strength and running time in aged mice. Additionally, γ-oryzanol has been found to promote the formation of slow-twitch muscle fiber, which is a fatigue-resistant myofiber type and conductive to the prevention of muscle aging. By binding to PPARδ and ERRγ directly, γ-oryzanol could upregulate mitochondrial biogenesis and alleviate inflammation, reducing the occurrence of muscle weakness of the aged mice [121]. Moreover, γ-oryzanol supplementation is demonstrated to promote GLUT4 translocation, alleviating insulin resistance in obese mice [122]. In addition, the antioxidant function of γ-oryzanol in muscle could be enhanced under an exercise condition [123], suggesting the physical exercise combined with γ-oryzanol supplementation might be a beneficial strategy to prevent muscle dysregulation.

2.6. β-Sitosterol

As the most abundant phytosterol, β-sitosterol exists widely in plant products such as barley [51], wheat, oat and quinoa [39,124]. It is considered to be a mild free-radical scavenger and has positive effects on maintaining cell membrane stability, as well as inhibiting inflammation and Alzheimer’s disease [125]. In ICR mice, β-sitosterol promotes mitochondrial electron transport and mitochondrial oxidative phosphorylation, enhancing muscle strength. In addition, β-sitosterol promotes the energy metabolism of C2C12 cells by activating UCP3 and inducing mitochondrial uncoupling [126]. Meanwhile, β-sitosterol dramatically enhanced mitochondrial biogenesis in chicken skeletal muscle via activating the PGC-1α/TFAM pathway [127]. In an L6 cell model, β-sitosterol could promote glucose uptake and lipid metabolism by activating ACC phosphorylation. A decreased level of triglycerides and cholesterol is achieved with LKB1-mediated AMPK activation [128]. Likewise, β-sitosterol supplementation is proved to facilitate GLUT4 translocation and upregulate insulin sensitivity in HFD-induced diabetic rats, manifested in the increased activity of glycolytic and gluconeogenesis enzymes [129]. Taken together, these data suggest the promoting effect of β-sitosterol on mitochondrial function and glucose metabolism in the skeletal muscle.

2.7. Alkylresorcinols

Alkylresorcinols (ARs) are phenolic lipids and exist abundantly in the bran part of cereal grains such as wheat, barley and rye [108]. They consists of various homologues distinguished by the length of the alkyl chain (C17:0−C25:0) attached to the phenyl ring [3]. ARs derived from wheat bran extracts have been indicated to prevent muscle atrophy in denervated mice via regulating the disruption of fatty acid metabolism induced by lipid autophagy [4]. In addition, dietary supplementation containing ARs was found to protect against isoproterenol-induced myofibrillar degeneration in rats via inhibiting oxidative damage caused by lipid accumulation [130]. 5-heptadecylresorcinol (AR-C17) as the main homologue of ARs has been proven to have the best anti-inflammatory effect among the identified ARs homologues in our previous study [3]. We found that AR-C17 alleviated muscle dysfunction via upregulating SIRT3/PGC-1α-mediated mitochondrial function. The mitochondria content and mitochondrial-biogenesis-related proteins such TFAM and NRF1 were significantly increased, contributing to enhanced exercise performance [131].

2.8. Betaine

Betaine is a natural antioxidant widely present in common cereals including whole grain wheat, barley, rye and oat [132]. Betaine supplementation is reported to improve muscle strength and power [133]. IGF-1 stimulates myotube growth and differentiation by activating the expression of downstream MyoD and myogenin. Betaine is reported to increase the expression of IGF-1 in murine myoblasts [134]. AKT is considered as a key regulator of muscle signaling and protein synthesis. Additionally, betaine supplementation could increase anabolism and weaken catabolism in muscle tissue by stimulating IGF-1/AKT signaling [135]. In an in vitro C2C12 model, betaine supplementation promoted myotube differentiation by upregulating the secretion of miR-29b-3p. In addition, through the activation of NFATc1/MyoD signaling, betaine promotes the transformation from fast-twitch to slow-twitch myofibers [136]. Moreover, betaine could promote muscle energy metabolism by upregulating mitochondrial biogenesis, accompanied by increased glucose consumption and ATP production in C2C12 cells [137].

2.9. Octacosanol

Octacosanol is a primary aliphatic alcohol that is abundant in rice [138] and wheat [139]. According to literature data, octacosanol has been identified as an anti-fatigue agent which could be stored in skeletal muscle after serial doses administration [140]. In a trained rats model, dietary supplementation with octacosanol also showed a better physical recovery after exhaustion and there was a shift from fast-twitch to slow-twitch myofibers in skeletal muscle [141]. In addition, orally administrated octacosanol might be converted into fatty acids and related to energy utilization, indicating its potential to improve exercise capacity through upregulating the energy supply [142]. The underlying mechanism of the anti-fatigue effect by octacosanol might be through increasing the expression of Prx and decreasing the expression of Trim63 in trained mice. Based on the gene ontology analysis, this improvement might be mediated by the Bcl3/TLRs/MAPK signaling pathway [143]. Overall, octacosanol resists fatigue possibly via modulating muscle-fiber-type transition and muscular energy metabolism.
Table 1. The structure and content of bioactive components in whole grains.
Table 1. The structure and content of bioactive components in whole grains.
ComponentsStructureDerived GrainsContent (mg/kg)References
γ-oryzanolFoods 11 02752 i001Rice1550.0–8400.0 [119]
Wheat 297.0–584.0[120]
Corn200.0–250.0[120]
β-sitosterolFoods 11 02752 i002Wheat 365.6–673.1[124]
Barley510.0–676.0[51]
Quinoa76.0–556.0[39]
Oat210.0–409.2[39]
AlkylresorcinolsFoods 11 02752 i003
Rye 360.0–3200.0[108]
Wheat 317.0–1732.0[108]
Ferulic acidFoods 11 02752 i004Barley 41.0–210.0 [108]
Corn 55.2 [43]
Wheat 472.0–813.8[40]
Barley 41.0–210.0 [108]
Quinoa126.8–281.7 [42]
Brown rice7.1–52.7[50]
Rice155.6–271.1[41]
Oat1493.6[39]
p-coumaric acidFoods 11 02752 i005Black rice84.8 ± 1.4[50]
Red rice176.9 ± 2.5[50]
Brown rice76.1–152.0[50]
Wheat 23.8–35.6 [40]
Barley0.8–58.4[51]
Quinoa31.3–42.0 [42]
ResveratrolFoods 11 02752 i006Buckwheat 5.7–7.9[54]
QuercetinFoods 11 02752 i007Buckwheat 3700.0[64]
Rice22.0–28.0[50]
Barley1.4–8.7[51]
Quinoa 11.3–42.8 [42]
Corn15.8[43]
Oat89.0[39]
Oligomeric procyanidinsFoods 11 02752 i008Black rice3500.0[74]
Red rice200.0[74]
Cyanidin-3-glucosideFoods 11 02752 i009Wheat1590.0[78]
Rice2682.0–4700.0[50]
Purple corn 1430.0[43]
Rye2270.0[78]
Barley1020.0[78]
Oat430.0[78]
CatechinsFoods 11 02752 i010Barley1312.47 ± 7.11[84]
Wheat355.87[83]
Buckwheat46.47 ± 0.17[84]
RutinFoods 11 02752 i011Wheat236.2[93]
Quinoa609.1[94]
Buckwheat69.95 ± 2.25[84]
CarotenoidsFoods 11 02752 i012Corn 9.69–13.0[101]
Wheat32.1–39.7[101]
Barley0.15–10.5[101]
BetaineFoods 11 02752 i013Oat200.0–1000.0[132]
Wheat270.0–1110.0[132]
Rye444.0–2213.0[132]
Barley460.0–980.0[132]
OctacosanolFoods 11 02752 i014Rice 95.7 [138]
Wheat0.4–8.9 [139]
β-glucanFoods 11 02752 i015Rice 140.0–570.0 [114]
Barley 4000.0–7000.0[108]
Oat 51,800.0–282,000.0[39]
Rye 1200.0–2900.0[108]
Wheat 400.0–1400.0[108]
TocotrienolFoods 11 02752 i016Black rice31.9 ± 0.4[50]
Red rice36.9 ± 1.6[50]
Brown rice19.6 ± 0.4[50]
Wheat 27.81 [108]
Rye 27.78[108]
Barley 18.73 [108]
Oat 11.59[108]
Buckwheat54.6–552.2[39]

3. Clinical Trials

Whole grains have been shown to possess a range of human health benefits due to their unique health-promoting nutrients in the bran, endosperm and germ tissues [38]. For instance, dietary fiber which is better preserved in whole grains has been reported to prevent age-associated declines in skeletal muscle mass. According to the analysis aimed at adults aged 40 and older in America during 2011–2018, higher dietary fiber intakes are associated with increased muscle strength, as well as improvements in muscle glucose homeostasis [144]. After supplementation with whole grain oats rich in dietary fiber, female individuals are found to have improved exercise capacity, along with increased utilization rate of glycogen and respiratory exchange ratio while decreased free fatty acid content in muscle tissue [145]. In a random trial, dietary intervention with high cereal fiber (whole grain wheat/rye: 30–50 g/day) effectively decreased muscle fat accumulation and improved the situation of insulin resistance and inflammatory reaction in the muscle of T2D patients [146]. Consistently, adopting a healthy diet pattern rich in whole grains is conducive to the improvement in muscle function in adults aged 60 or older, characterized by a higher muscle mass and lower rate of sarcopenia [147]. Interestingly, people with a diet rich in catechins are found to have improved skeletal muscle strength. For instance, 62 males (aged 70) who received an 8-week catechin supplementation (1 mg/kg/d) showed improved muscle strength and decreased expression of MSTN, suggesting the beneficial effect of catechin on aging-related sarcopenia [148]. Moreover, patients with type 2 diabetes who received catechin supplementation (100 mg/d) for 3 months were found to have enhanced mitochondrial biogenesis and reduced mitochondrial damage in the skeletal muscle, alleviating diabetes-induced muscle dysfunction [149]. As a whole grain bioactive component, carotenoids exhibit notable antioxidant and senescence retarding effects and are inversely associated with sarcopenic symptomology [150]. For overweight and elderly people, a diet rich in carotene is conducive to maintaining skeletal muscle function and reducing the development of sarcopenia [151]. In addition, carotene supplementation could decrease the inflammation level and oxidative damage in the muscle of adults aged 65 [152]. Moreover, according to the survey that investigated the effect of carotene on muscle health of 80 adults aged 50 years old, carotene intervention significantly increased muscle insulin sensitivity, suggesting its positive effect on muscle glucose metabolism [153]. In a recent survey, betaine administration also showed an impressive effect on the regulation of skeletal muscle mass. After receiving betaine supplement for 3 years, a significant reduction in muscle loss was observed in the 1242 male participants aged 41–60 [154]. Additionally, adolescent handball players were found with higher exercise endurance and stronger anti-fatigue performance after two weeks of betaine supplementation (2.5 g/day) [155]. Among 23 male subjects, 11 of whom accepted betaine supplementation (2.5 g/day) along with 6-week training showed improved muscle mass and decreased muscle fat accumulation [156]. In summary, these clinical data demonstrate the physiological activity of whole grain components and their regulation effect on skeletal muscle dysfunction.
Table 2. Bioactive compounds derived from whole grains for the regulation of muscle function.
Table 2. Bioactive compounds derived from whole grains for the regulation of muscle function.
CompoundsExperimental ModelEffective DoseTargetsTarget ProcessEffectReferences
γ-oryzanolMale C57BL/6 mice14.78 mg/kg/dPPARδ
ERRγ
Upregulate mitochondrial biogenesis and promote fatty acid β-oxidation Reduce muscle weakness, alleviate inflammation and enhance muscle strength[121]
Male Wistar rats0.5% (w/w)GLUT4Promote antioxidant and anti-inflammatory effectsAlleviate insulin resistance in muscle[122]
β-sitosterolICR mice
C2C12 cells
35 or 350 μg/kg/d 15 μMUCP3Upregulate the activity of complex proteins in the mitochondrial electron transport chain and induce mitochondrial uncouplingMeet energy demand and promote muscle strength[126]
Broilers 100 mg/kgPGC-1α/TFAM signalingUpregulate oxidative status and mitochondrial biogenesisPromote muscle performance[127]
L6 cells20 μMLKB1/AMPK signalingIncrease ACC phosphorylation and triglyceride metabolismPromote glucose homeostasis and lipid metabolism[128]
Male diabetic rats20 mg/kg/dRab/IRAP/Munc 18 pathwayUpregulate GLUT4 translocation and glycolytic and gluconeogenesis enzymesAlleviate insulin resistance in muscle[129]
Male C57BL/6 mice200 mg/kg/dFoxO1 signalingAlleviate muscle protein loss and inhibit protein degradationAlleviate muscle atrophy[157]
AlkylresorcinolsMale C57BL/6 mice0.4% (w/w)Pyruvate dehydrogenase kinase 4 (PDK4)Promote lipid-autophagy-induced lipid metabolism disorderInhibit the reduction in muscle size and prevent muscle atrophy[4]
Male Wistar rats400 mg/kg/dGlutathione peroxidase (GPx)Inhibit oxidative damage caused by lipid accumulation and increase the expression of lactate dehydrogenase (LDH), alanine transaminase (ALT)Alleviate myocardial infarction and exert an anti-fatigue effect[130]
Male C57BL/6J mice30 or 150 mg/kg/dSIRT3/PGC-1α signalingIncrease mitochondrial content and mitochondrial biogenesisImprove exercise capacity[131]
Ferulic acidMale SD rats0.6 mg/kg/dPKCεInhibit lipid-accumulation-induced inflammationAlleviate insulin resistance[44]
Isolated rat psoas muscle cells from Sprague Dawley male albino rats15–240 µg/mLα-glucosidase and α-amylasePromote fatty acid oxidative decomposition and inhibit carbohydrate and lipid hydrolyzing enzymesAlleviate oxidative stress and mitigate redox imbalance [5]
C2C12 cells25 μMPI3K/Cpkc signalingPromote glucose uptake and glycogen synthesisImprove insulin resistance[45]
Zebrafish0.06 mg/dMyoD
myogenin
Promote muscle growth Increase the CSA of muscle fiber and muscle mass[46]
Duroc × Landrace × Yorkshire (DLY) weaned piglets0.05% or 0.45% (w/w) SIRT1/AMPK/PGC-1α signalingImprove the activity of SDH and MDH,
increase the expression of slow MyHC protein
Increase the proportion of slow-twitch fiber and promote mitochondria function[47]
C2C12 cells0.5 or 1 μMSIRT1/AMPK signalingIncrease the protein level of slow MyHC and decrease the protein level of fast MyHCPromote slow oxidative muscle fiber formation and inhibit fast muscle fiber formation and exert an anti-fatigue function[48]
p-coumaric acidL6 cells100 μMAMPK signalingPromote the fatty acid β-oxidationInhibit lipid-accumulation-induced inflammation in muscle[53]
C2C12 cells0.1 mMAMPK signalingIncrease expression of myogenin and myoDImprove myogenic differentiation [52]
ResveratrolMale C57 BL/6J mice50 mg/kg/dAMPK/
FOXO3 signaling
Improve mitochondrial functionImprove muscle atrophy[55]
Male C57BL/6J mice0.4% (w/w)AMPK/PGC-1α signalingIncrease the level of muscle regeneration proteins including MyoG, Myf5 and Pax7 and mitochondrial biogenesisEnhance muscle proliferation, differentiation and regeneration of impaired muscle [57]
L6 cells25 μMPKA/LKB1/
AMPK pathway
Improve mitochondrial dysfunction and oxidative stressIncrease muscle mass and myofiber size and improve induced muscle atrophy[56]
Male C57BL/6J mice15 mg/kg/d Increase muscle glycogen synthesis and reduce ROS levelsReduce insulin resistance and promote lipid metabolism[58]
C2C12 cells50 μMAKT signalingModulate redox levels and glucose absorptionReduce insulin resistance[59]
C2C12 cells100 μMAKT/mTOR/FOXO1 signalingInhibit the atrophy-related ubiquitin ligaseImprove muscle hypertrophy and muscle atrophy[60]
Male Kunming mice400 mg/kg/dAdiopR1–AMPK–PGC-1α signalingIncrease the expression of myosin heavy chain (MyHC) 1, MyHC2a and MyHC2xImprove the transformation from fast- to slow-twitch muscle fibers and exercise performance[61]
C2C12 cells50 μMmiR-22-3pIncrease the activities of lactate dehydrogenase (SDH) and malate dehydrogenase (MDH)Promote muscle-fiber-type conversion from fast-twitch to slow-twitch muscle fibers and exert an anti-fatigue effect[62]
Male ICR mice25 mg/kg/d Increase the activities of LDH (lactic dehydrogenase) and creatine kinase (CK)Improve muscle recovery and inflammation[63]
QuercetinMale Balb/c mice0.5% (w/w)Adiponectin signalingDecrease the fast MyHC
and MyHC IIb protein expression
Promote muscle-fiber-type transformation from fast-twitch to slow-twitch muscle fibers[66]
Male C57BL/6 mice0.05% (w/w)HO-1/NRF2 signalingDecrease inflammatory response and oxidative stressReduce obesity-induced muscle atrophy[68]
Male C57BL/6 mice0.2% (w/w)PGC-1α signalingImprove mitochondrial biogenesis and oxidative phosphorylationAlleviate disuse-induced muscle atrophy[69]
C2C12 cells20 µMAMPK signaling Enhance insulin-stimulated glucose uptake and decrease inflammatory responseAmeliorate inflammation-induced insulin resistance[70]
Oligomeric procyanidins (OPCs)Male ICR mice15 mg/kg/dmTOR signalingIncrease glucose uptake and glycolysis, improve heat generation and inhibit gluconeogenesis and lipogenesisImprove glucose homeostasis, lipid metabolism and insulin sensitivity[75]
Human primary skeletal muscle cells 10 or 25 μMAKT signalingIncrease glycogen synthesis and glucose uptakeImprove glucose utilization and alleviate insulin resistance[76]
Male ICR mice10 μg/kg/dAMPK signalingPromote GLUT4 translocationIncrease insulin sensitivity[77]
Cyanidin-3-glucoside (Cy3G)Male ICR mice1 mg/kg/dPGC-1α signalingImprove mitochondrial content and mitochondrial biogenesisImprove exercise capacity[82]
Human skeletal muscle cells100 µM Inhibit the activity of α-amylase and α-glucosidase Alleviate diabetes[81]
Catechin C2C12 cells20 µMMyoD, MyoG, and MyHCPromote myotube differentiationImprove skeletal muscle regeneration and repair[87]
Male C57BL/6 mice25 mg/kg/d Wnt signalingPromote myotube differentiationPromote muscle regeneration [90]
C2C12 cells10 μMAkt Promote myotube differentiationImprove muscle regeneration[86]
Male rats1.0 mg/kg/dp38 MAPK signalingPromote mitochondrial respiratory capacity and mitochondrial biogenesisEnhance the ability of resisting fatigue[88]
Male Goto–Kakizaki (GK) rats100 mg/kg/dROS-ERK/JNK-p53 pathwayPromote mitochondrial autophagy Alleviate diabetic-induced sarcopenia[89]
Male Sprague Dawley rats200 mg/kg/dIGF-1Downregulate UPS-mediated muscle protein degradation and upregulate IGF-1-mediated muscle protein synthesisIncrease muscle mass[91]
Male SD rats0.1% (w/w) Inhibit the expression of oxidative-modified proteinsPrevent muscle from oxidative stress induced by free radicals[92]
RutinMale Sprague Dawley rats0.1% (w/w)AMPK signalingEnhance mitochondrial DNA (mtDNA) content and mitochondrial biogenesisImprove obesity-induced muscle mitochondrial dysfunction[97]
Male C57BL/6 mice60 mg/kg/dPGC-1α signalingUpregulate mediated mitochondrial biogenesis and decrease the level of lactic acidImprove fatigue-resistance capacity[98]
C2C12 cells100 μMNF-κB signalingDecrease the expression of IL-6 and iNOS and the production of ROSInhibit oxidative-stress-induced skeletal muscle injury[99]
Lutein Male Wistar rats0.5 mg/kg/dNF-κB signaling Reduce oxidative stress and inflammation and decrease the production of ROS Improve skeletal muscle IR injury [103]
β-caroteneMale Kwl: ddY mice0.5 mg/kg/dIGF-1Promote protein synthesis and reduce ubiquitin-mediated muscle protein degradationIncrease muscle mass and prevent muscle hypertrophy[105]
C2C12 cells10 μMFOXO3ADecrease the level of Atrogin-1 and MuRF1Increase muscle mass and exhibit an improvement effect on oxidative-stress-induced muscle atrophy[107]
BetaineC2C12 cells10 mMNFATc1/
MyoD signaling
Upregulate the expression of miR-29b-3p and promote myotube differentiation and the expression of slow MyHC proteins Promote muscle cell differentiation and the transformation from fast muscle to slow muscle fiber[136]
C2C12 cells10 mMIGF-1 signalingIncrease the expression of MyoD and myogeninPromote muscle fiber differentiation and growth[134]
C2C12 cells2 or 5 mMPGC-1α signalingIncrease mitochondrial biogenesis and ATP productionPromote muscle differentiation and the transformation from fast muscle to slow muscle fiber[137]
OctacosanolMale Wistar rats2.0 μCi/dose Promote energy mobilization and energy supplyEnhance physical performance[140]
Male C57BL/6 mice200 mg/kg/dBcl3/TLRs/MAPK signalingIncrease the expression of Prx, Trim63 and ATPase activityExert an anti-fatigue effect[143]
Male SD rats0.75% (w/w)Creatine phosphorylation Promote the shift from fast-twitch to slow-twitch myofibersExert an anti-fatigue effect[141]
Male Wistar rats2.0 μCi/doseACC phosphorylationMeet energy demandImprove exercise capacity[142]
β-glucanMale SD rats312.5 mg/kg/d Decrease the activity of lactate dehydrogenase and the creatine kinase Increase exercise capacity and facilitate the recovery from fatigue[116]
C2C12 cells20 mg/mLMyf5 and Mox2Increase muscle cell proliferation and differentiationPromote the transformation from fast muscle fibers to slow muscle fibers[117]
Duchenne muscular dystrophy (DMD) zebrafish model8 mg/LMitochondrial respiration enzymeImprove mitochondrial respiration and prevent oxidative stressImprove exercise capacity[118]
TocotrienolMale C57BL/6J mice100 or 300 mg/kg/d AMPK/SIRT1/PGC-1α signaling Upregulate the expression of proliferation and differentiation related proteins
Increase mitochondrial biogenesis
Prevent diabetes-related skeletal muscle atrophy[110]
Human skeletal muscle myoblasts50 μg/mL miR-206Increase the expression of IGF1R and decrease the expression of Pax7Promote muscle cell proliferation and differentiation[111]
Stress-induced premature senescence (SIPS) Human skeletal muscle myoblasts (CHQ5B)50 μg/mLWnt signaling FOXO pathwayDownregulate the expression of MSTN and increase the expression of muscle cells regeneration related proteins such as EREG, SHC1 and SHC3Promote muscle cell regeneration and alleviate muscle loss[112]
Male C57BL/6J mice400 mg/kg/dCOXⅠ-ⅤPromote mitochondrial respiration and reduce lipid peroxidationIncrease muscle mass and improve glucose homeostasis[113]

4. Conclusions and Perspectives

Based on the included studies, bioactive components in whole grains exhibited important roles in regulating muscle dysfunction that can benefit individuals with related muscle diseases. Significantly, this evidence suggests that nutritional strategies are a promising way to prevent and modulate metabolic-related disorders in muscles. However, further clinical research studies are needed as there is a limited number of trials in which participants with skeletal muscle diseases are specifically targeted. Additionally, evaluation of multimodal intervention strategies such as exercise training combined with dietary supplementation, as well as the appropriate dosage and duration for the intervention are also needed which might provide novel therapeutic strategies to counteract skeletal-muscle-related diseases.

Author Contributions

Conceptualization, Q.L. and Z.W.; investigation, J.L. and J.W.; writing—original draft preparation, Q.L.; writing—review and editing, Z.W.; visualization, H.Y. and S.S.; funding acquisition, Z.W. and J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National Natural Science Foundation of China (32172207 and 32001704) and the General Project of Beijing Municipal Education Commission (KM202210011011).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CSA: muscle fiber cross-sectional area; MyHC: myosin heavy chain; MD: muscular dystrophy; ATP: adenosine-triphosphate; AMPK: the adenosine monophosphate (AMP)-activated protein kinase; PGC-1a: peroxisome proliferators-activated receptor-alpha; NRF1: nuclear respiratory factor 1; TFAM: mitochondrial transcription factor A; GABP: GA binding protein transcription factor; FNIP1: folliculin interacting protein 1; mTOR: molecular target of rapamycin; mtDNA: mitochondrial DNA; ROS: reactive oxygen species; SIRT2: recombinant sirtuin 2; TGF-β: transforming growth factor-β; Smad3: mothers against DPP homolog 3; ERK: extra cellular signal-regulated kinase; FOXO3a: fork head box O3a; ERRγ: estrogen-related receptor γ; PPARδ: peroxisome proliferator-activated receptor δ; UCP3: uncoupling protein 3; MyoD: myogenic differentiation antigen; TFB2M: transcription factor B2; TNF-α: tumor necrosis factor-α; GSK-3β: glycogen synthase kinase-3β; OPCs: oligomeric procyanidins; ACC: acetyl CoA carboxylase; IGF-1: insulin-like growth factor 1; NFATc1: nuclear factor of activated T-cells; Cy3G: cyanidin-3-glucoside; Pax7: paired box gene 7; HSMM: human skeletal muscle myoblasts; SDH: succinate dehydrogenase; LDH: lactate dehydrogenase; Myf5: Myogenic determining factor 5; CK-MB: Creatine Kinase MB Isoenzyme; Mox2: Mesenchyme Homeobox 2; Prx: periaxin; Trim63: tripartite motif-containing 63; TLRs: Toll-like receptors; MAPK: mitogen-activated protein kinase; GO: Gene ontology; PKA: protein kinase A; LKB1: Liver kinase B1; NFATc1: Homo sapiens nuclear factor of activated T-cells.

References

  1. Liu, J.; Yu, L.L.; Wu, Y. Bioactive Components and Health Beneficial Properties of Whole Wheat Foods. J. Agric. Food Chem. 2020, 68, 12904–12915. [Google Scholar] [CrossRef] [PubMed]
  2. Nogala-Katucka, M.; Kawka, A.; Dwiecki, K.; Siger, A. Evaluation of bioactive compounds in cereals study of wheat, barley, oat and selected grain products. Acta Sci. Pol.-Technol. Aliment. 2020, 19, 405–423. [Google Scholar] [CrossRef]
  3. Liu, J.; Hao, Y.; Wang, Z.; Ni, F.; Wang, Y.; Gong, L.; Sun, B.; Wang, J. Identification, Quantification, and Anti-inflammatory Activity of 5-n-Alkylresorcinols from 21 Different Wheat Varieties. J. Agric. Food Chem. 2018, 66, 9241–9247. [Google Scholar] [CrossRef] [PubMed]
  4. Hiramoto, S.; Yahata, N.; Saitoh, K.; Yoshimura, T.; Wang, Y.; Taniyama, S.; Nikawa, T.; Tachibana, K.; Hirasaka, K. Dietary supplementation with alkylresorcinols prevents muscle atrophy through a shift of energy supply. J. Nutr. Biochem. 2018, 61, 147–154. [Google Scholar] [CrossRef]
  5. Salau, V.F.; Erukainure, O.L.; Koorbanally, N.A.; Islam, M.S. Ferulic acid promotes muscle glucose uptake and modulate dysregulated redox balance and metabolic pathways in ferric-induced pancreatic oxidative injury. J. Food Biochem. 2022, 46, e13641. [Google Scholar] [CrossRef]
  6. Strasser, B.; Wolters, M.; Weyh, C.; Kruger, K.; Ticinesi, A. The Effects of Lifestyle and Diet on Gut Microbiota Composition, Inflammation and Muscle Performance in Our Aging Society. Nutrients 2021, 13, 2045. [Google Scholar] [CrossRef]
  7. Arenas-Jal, M.; Sune-Negre, J.M.; Perez-Lozano, P.; Garcia-Montoya, E. Trends in the food and sports nutrition industry: A review. Crit. Rev. Food Sci. Nutr. 2020, 60, 2405–2421. [Google Scholar] [CrossRef]
  8. Manore, M.; Meeusen, R.; Roelands, B.; Moran, S.; Popple, A.D.; Naylor, M.J.; Burke, L.M.; Stear, S.J.; Castell, L.M. BJSM reviews: A-Z of nutritional supplements: Dietary supplements, sports nutrition foods and ergogenic aids for health and performance-Part 16. Br. J. Sports Med. 2011, 45, 73–74. [Google Scholar] [CrossRef] [PubMed]
  9. Peeling, P.; Castell, L.M.; Derave, W.; de Hon, O.; Burke, L.M. Sports Foods and Dietary Supplements for Optimal Function and Performance Enhancement in Track-and-Field Athletes. Int. J. Sport Nutr. Exerc. Metab. 2019, 29, 198–209. [Google Scholar] [CrossRef]
  10. Tomlinson, D.J.; Erskine, R.M.; Morse, C.I.; Winwood, K.; Onambele-Pearson, G. The impact of obesity on skeletal muscle strength and structure through adolescence to old age. Biogerontology 2016, 17, 467–483. [Google Scholar] [CrossRef] [Green Version]
  11. Liu, X.; Du, Y.; Trakooljul, N.; Brand, B.; Muryni, E.; Krischek, C.; Wicke, M.; Schwerin, M.; Wimmers, K.; Ponsuksili, S. Muscle Transcriptional Profile Based on Muscle Fiber, Mitochondrial Respiratory Activity, and Metabolic Enzymes. Int. J. Biol. Sci. 2015, 11, 1348–1362. [Google Scholar] [CrossRef] [PubMed]
  12. Blaauw, B.; Schiaffino, S.; Reggiani, C. Mechanisms Modulating Skeletal Muscle Phenotype. Compr. Physiol. 2013, 3, 1645–1687. [Google Scholar] [CrossRef]
  13. Khor, S.C.; Razak, A.M.; Ngah, W.Z.W.; Yusof, Y.A.M.; Karim, N.A.; Makpol, S. The Tocotrienol-Rich Fraction Is Superior to Tocopherol in Promoting Myogenic Differentiation in the Prevention of Replicative Senescence of Myoblasts. PLoS ONE 2016, 11, e0149265. [Google Scholar] [CrossRef]
  14. Lecker, S.H.; Goldberg, A.L.; Mitch, W.E. Protein degradation by the ubiquitin-proteasome pathway in normal and disease states. J. Am. Soc. Nephrol. 2006, 17, 1807–1819. [Google Scholar] [CrossRef] [PubMed]
  15. Zhu, L.-N.; Ren, Y.; Chen, J.-Q.; Wang, Y.-Z. Effects of myogenin on muscle fiber types and key metabolic enzymes in gene transfer mice and C2C12 myoblasts. Gene 2013, 532, 246–252. [Google Scholar] [CrossRef]
  16. Qaisar, R.; Renaud, G.; Hedstrom, Y.; Pollanen, E.; Ronkainen, P.; Kaprio, J.; Alen, M.; Sipila, S.; Artemenko, K.; Bergquist, J.; et al. Hormone replacement therapy improves contractile function and myonuclear organization of single muscle fibres from postmenopausal monozygotic female twin pairs. J. Physiol.-Lond. 2013, 591, 2333–2344. [Google Scholar] [CrossRef] [PubMed]
  17. Qaisar, R.; Bhaskaran, S.; Van Remmenn, H. Muscle fiber type diversification during exercise and regeneration. Free. Radic. Biol. Med. 2016, 98, 56–67. [Google Scholar] [CrossRef] [PubMed]
  18. Hoppeler, H.; Fluck, M. Plasticity of skeletal muscle mitochondria: Structure and function. Med. Sci. Sports Exerc. 2003, 35, 95–104. [Google Scholar] [CrossRef]
  19. Gan, Z.; Fu, T.; Kelly, D.P.; Vega, R.B. Skeletal muscle mitochondrial remodeling in exercise and diseases. Cell Res. 2018, 28, 969–980. [Google Scholar] [CrossRef]
  20. Heydemann, A. Skeletal Muscle Metabolism in Duchenne and Becker Muscular Dystrophy-Implications for Therapies. Nutrients 2018, 10, 796. [Google Scholar] [CrossRef] [Green Version]
  21. Narkar, V.A.; Downes, M.; Yu, R.T.; Embler, E.; Wang, Y.-X.; Banayo, E.; Mihaylova, M.M.; Nelson, M.C.; Zou, Y.; Juguilon, H.; et al. AMPK and PPAR delta Agonists are exercise mimetics. Cell 2008, 134, 405–415. [Google Scholar] [CrossRef]
  22. Liu, J.; Liang, X.; Zhou, D.; Lai, L.; Xiao, L.; Liu, L.; Fu, T.; Kong, Y.; Zhou, Q.; Vega, R.B.; et al. Coupling of mitochondrial function and skeletal muscle fiber type by a miR-499/Fnip1/AMPK circuit. Embo Mol. Med. 2016, 8, 1212–1228. [Google Scholar] [CrossRef]
  23. Reyes, N.L.; Banks, G.B.; Tsang, M.; Margineantu, D.; Gu, H.; Djukovic, D.; Chan, J.; Torres, M.; Liggitt, H.D.; Hirenallur-S, D.K.; et al. Fnip1 regulates skeletal muscle fiber type specification, fatigue resistance, and susceptibility to muscular dystrophy. Proc. Natl. Acad. Sci. USA 2015, 112, 424–429. [Google Scholar] [CrossRef] [PubMed]
  24. Ljubicic, V.; Jasmin, B.J. AMP-activated protein kinase at the nexus of therapeutic skeletal muscle plasticity in Duchenne muscular dystrophy. Trends Mol. Med. 2013, 19, 614–624. [Google Scholar] [CrossRef]
  25. Spaulding, H.R.; Kelly, E.M.; Quindry, J.C.; Sheffield, J.B.; Hudson, M.B.; Selsby, J.T. Autophagic dysfunction and autophagosome escape in the mdx mus musculus model of Duchenne muscular dystrophy. Acta Physiol. 2018, 222, e12944. [Google Scholar] [CrossRef]
  26. Theilen, N.T.; Kunkel, G.H.; Tyagi, S.C. The Role of Exercise and TFAM in Preventing Skeletal Muscle Atrophy. J. Cell. Physiol. 2017, 232, 2348–2358. [Google Scholar] [CrossRef]
  27. Komatsu, R.; Okazaki, T.; Ebihara, S.; Kobayashi, M.; Tsukita, Y.; Nihei, M.; Sugiura, H.; Niu, K.; Ebihara, T.; Ichinose, M. Aspiration pneumonia induces muscle atrophy in the respiratory, skeletal, and swallowing systems. J. Cachexia Sarcopenia Muscle 2018, 9, 643–653. [Google Scholar] [CrossRef]
  28. Han, Z.; Chang, C.; Zhu, W.; Zhang, Y.; Zheng, J.; Kang, X.; Jin, G.; Gong, Z. Role of SIRT2 in regulating the dexamethasone-activated autophagy pathway in skeletal muscle atrophy. Biochem. Cell Biol. 2021, 99, 562–569. [Google Scholar] [CrossRef] [PubMed]
  29. Theilen, N.T.; Jeremic, N.; Weber, G.J.; Tyagi, S.C. TFAM overexpression diminishes skeletal muscle atrophy after hindlimb suspension in mice. Arch. Biochem. Biophys. 2019, 666, 138–147. [Google Scholar] [CrossRef] [PubMed]
  30. Siu, P.M. Muscle Apoptotic Response to Denervation, Disuse, and Aging. Med. Sci. Sports Exerc. 2009, 41, 1876–1886. [Google Scholar] [CrossRef]
  31. Falduto, M.T.; Czerwinski, S.M.; Hickson, R.C. Glucocorticoid-induced muscle atrophy prevention by exercise in fast-twitch fibers. J. Appl. Physiol. 1990, 69, 1058–1062. [Google Scholar] [CrossRef] [PubMed]
  32. Abrigo, J.; Campos, F.; Simon, F.; Riedel, C.; Cabrera, D.; Vilos, C.; Cabello-Verrugio, C. TGF-beta requires the activation of canonical and non-canonical signalling pathways to induce skeletal muscle atrophy. Biol. Chem. 2018, 399, 253–264. [Google Scholar] [CrossRef] [PubMed]
  33. Lin, I.H.; Chang, J.-L.; Hua, K.; Huang, W.-C.; Hsu, M.-T.; Chen, Y.-F. Skeletal muscle in aged mice reveals extensive transformation of muscle gene expression. BMC Genet. 2018, 19, 55. [Google Scholar] [CrossRef]
  34. de Sire, R.; Rizzatti, G.; Ingravalle, F.; Pizzoferrato, M.; Petito, V.; Lopetuso, L.; Graziani, C.; de Sire, A.; Mentella, M.C.; Mele, M.C.; et al. Skeletal muscle-gut axis: Emerging mechanisms of sarcopenia for intestinal and extra intestinal diseases. Minerva Gastroenterol. Dietol. 2018, 64, 351–362. [Google Scholar] [CrossRef]
  35. Rasbach, K.A.; Gupta, R.K.; Ruas, J.L.; Wu, J.; Naseri, E.; Estall, J.L.; Spiegelman, B.M. PGC-1 alpha regulates a HIF2 alpha-dependent switch in skeletal muscle fiber types. Proc. Natl. Acad. Sci. USA 2010, 107, 21866–21871. [Google Scholar] [CrossRef] [PubMed]
  36. Pardo, P.S.; Boriek, A.M. The physiological roles of Sirt1 in skeletal muscle. Aging-Us 2011, 3, 430–437. [Google Scholar] [CrossRef]
  37. Sakuma, K.; Aoi, W.; Yamaguchi, A. Molecular mechanism of sarcopenia and cachexia: Recent research advances. Pflug. Arch.-Eur. J. Physiol. 2017, 469, 573–591. [Google Scholar] [CrossRef] [PubMed]
  38. Wang, Z.; Yang, Z.; Liu, J.; Hao, Y.; Sun, B.; Wang, J. Potential Health Benefits of Whole Grains: Modulation of Mitochondrial Biogenesis and Energy Metabolism. J. Agric. Food Chem. 2021, 69, 14065–14074. [Google Scholar] [CrossRef]
  39. Raguindin, P.F.; Itodo, O.A.; Stoyanov, J.; Dejanovic, G.M.; Gamba, M.; Asllanaj, E.; Minder, B.; Bussler, W.; Metzger, B.; Muka, T.; et al. A systematic review of phytochemicals in oat and buckwheat. Food Chemistry 2021, 338, 127982. [Google Scholar] [CrossRef] [PubMed]
  40. Saini, P.; Kumar, N.; Kumar, S.; Mwaurah, P.W.; Panghal, A.; Attkan, A.K.; Singh, V.K.; Garg, M.K.; Singh, V. Bioactive compounds, nutritional benefits and food applications of colored wheat: A comprehensive review. Crit. Rev. Food Sci. Nutr. 2021, 61, 3197–3210. [Google Scholar] [CrossRef] [PubMed]
  41. Yu, L.; Li, G.; Li, M.; Xu, F.; Beta, T.; Bao, J. Genotypic variation in phenolic acids, vitamin E and fatty acids in whole grain rice. Food Chemistry 2016, 197, 776–782. [Google Scholar] [CrossRef]
  42. Han, Y.; Chi, J.; Zhang, M.; Zhang, R.; Fan, S.; Huang, F.; Xue, K.; Liu, L. Characterization of saponins and phenolic compounds: Antioxidant activity and inhibitory effects on alpha-glucosidase in different varieties of colored quinoa (Chenopodium quinoa Willd). Biosci. Biotechnol. Biochem. 2019, 83, 2128–2139. [Google Scholar] [CrossRef]
  43. Cristianini, M.; Sanchez, J.S. Extraction of bioactive compounds from purple corn using emerging technologies: A review. J. Food Sci. 2020, 85, 862–869. [Google Scholar] [CrossRef]
  44. Gogoi, B.; Chatterjee, P.; Mukherjee, S.; Buragohain, A.K.; Bhattacharya, S.; Dasgupta, S. A polyphenol rescues lipid induced insulin resistance in skeletal muscle cells and adipocytes. Biochem. Biophys. Res. Commun. 2014, 452, 382–388. [Google Scholar] [CrossRef] [PubMed]
  45. Kang, B.-B.; Chiang, B.-H. Amelioration of insulin resistance using the additive effect of ferulic acid and resveratrol on vesicle trafficking for skeletal muscle glucose metabolism. Phytother. Res. 2020, 34, 808–816. [Google Scholar] [CrossRef]
  46. Wen, Y.; Ushio, H. Ferulic Acid Promotes Hypertrophic Growth of Fast Skeletal Muscle in Zebrafish Model. Nutrients 2017, 9, 66. [Google Scholar] [CrossRef] [PubMed]
  47. Anis, E.; Zafeer, M.F.; Firdaus, F.; Islam, S.N.; Khan, A.A.; Ali, A.; Hossain, M.M. Ferulic acid reinstates mitochondrial dynamics through PGC1 alpha expression modulation in 6-hydroxydopamine lesioned rats. Phytother. Res. 2020, 34, 214–226. [Google Scholar] [CrossRef]
  48. Chen, X.; Guo, Y.; Jia, G.; Zhao, H.; Liu, G.; Huang, Z. Ferulic acid regulates muscle fiber type formation through the Sirt1/AMPK signaling pathway. Food Funct. 2019, 10, 259–265. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, Y.; Chen, X.; Huang, Z.; Chen, D.; Yu, B.; Chen, H.; Yu, J.; Luo, Y.; Zheng, P.; He, J. Effects of dietary ferulic acid supplementation on growth performance and skeletal muscle fiber type conversion in weaned piglets. J. Sci. Food Agric. 2021, 101, 5116–5123. [Google Scholar] [CrossRef] [PubMed]
  50. Gong, E.S.; Liu, C.; Li, B.; Zhou, W.; Chen, H.; Li, T.; Wu, J.; Zeng, Z.; Wang, Y.; Si, X.; et al. Phytochemical profiles of rice and their cellular antioxidant activity against ABAP induced oxidative stress in human hepatocellular carcinoma HepG2 cells. Food Chem. 2020, 318, 126484. [Google Scholar] [CrossRef]
  51. Li, Y.; Li, T.; Liu, R.H. Bioactive compounds of highland barley and their health benefits. J. Cereal Sci. 2022, 103, 103366. [Google Scholar] [CrossRef]
  52. Ilavenil, S.; Kim, D.H.; Srigopalram, S.; Arasu, M.V.; Lee, K.D.; Lee, J.C.; Lee, J.S.; Renganathan, S.; Choi, K.C. Potential Application of p-Coumaric Acid on Differentiation of C2C12 Skeletal Muscle and 3T3-L1 Preadipocytes-An In Vitro and In Silico Approach. Molecules 2016, 21, 997. [Google Scholar] [CrossRef]
  53. Yoon, S.-A.; Kang, S.-I.; Shin, H.-S.; Kang, S.-W.; Kim, J.-H.; Ko, H.-C.; Kim, S.-J. p-Coumaric acid modulates glucose and lipid metabolism via AMP-activated protein kinase in L6 skeletal muscle cells. Biochem. Biophys. Res. Commun. 2013, 432, 553–557. [Google Scholar] [CrossRef] [PubMed]
  54. Nemcova, L.; Zima, J.; Barek, J.; Janovska, D. Determination of resveratrol in grains, hulls and leaves of common and tartary buckwheat by HPLC with electrochemical detection at carbon paste electrode. Food Chem. 2011, 126, 374–378. [Google Scholar] [CrossRef]
  55. Liu, J.; Peng, Y.; Wang, X.; Fan, Y.; Qin, C.; Shi, L.; Tang, Y.; Cao, K.; Li, H.; Long, J.; et al. Mitochondrial Dysfunction Launches Dexamethasone-Induced Skeletal Muscle Atrophy via AMPK/FOXO3 Signaling. Mol. Pharm. 2016, 13, 73–84. [Google Scholar] [CrossRef] [PubMed]
  56. Huang, Y.; Zhu, X.; Chen, K.; Lang, H.; Zhang, Y.; Hou, P.; Ran, L.; Zhou, M.; Zheng, J.; Yi, L.; et al. Resveratrol prevents sarcopenic obesity by reversing mitochondrial dysfunction and oxidative stress via the PKA/LKB1/AMPK pathway. Aging-Us 2019, 11, 2217–2240. [Google Scholar] [CrossRef]
  57. Niu, W.; Wang, H.; Wang, B.; Mao, X.; Du, M. Resveratrol improves muscle regeneration in obese mice through enhancing mitochondrial biogenesis. J. Nutr. Biochem. 2021, 98, 108804. [Google Scholar] [CrossRef]
  58. Gong, L.; Guo, S.; Zou, Z. Resveratrol ameliorates metabolic disorders and insulin resistance in high-fat diet-fed mice. Life Sci. 2020, 242, 117212. [Google Scholar] [CrossRef]
  59. Quan, Y.; Hua, S.; Li, W.; Zhan, M.; Li, Y.; Lu, L. Resveratrol bidirectionally regulates insulin effects in skeletal muscle through alternation of intracellular redox homeostasis. Life Sci. 2020, 242, 117188. [Google Scholar] [CrossRef]
  60. Wang, D.-T.; Yin, Y.; Yang, Y.-J.; Lv, P.-J.; Shi, Y.; Lu, L.; Wei, L.-B. Resveratrol prevents TNF-alpha-induced muscle atrophy via regulation of Akt/mTOR/FoxO1 signaling in C2C12 myotubes. Int. Immunopharmacol. 2014, 19, 206–213. [Google Scholar] [CrossRef]
  61. Jiang, Q.; Cheng, X.; Cui, Y.; Xia, Q.; Yan, X.; Zhang, M.; Lan, G.; Liu, J.; Shan, T.; Huang, Y. Resveratrol regulates skeletal muscle fibers switching through the AdipoR1-AMPK-PGC-1 alpha pathway. Food Funct. 2019, 10, 3334–3343. [Google Scholar] [CrossRef]
  62. Wen, W.; Chen, X.; Huang, Z.; Chen, D.; Chen, H.; Luo, Y.; He, J.; Zheng, P.; Yu, J.; Yu, B. Resveratrol regulates muscle fiber type conversion via miR-22-3p and AMPK/SIRT1/PGC-1 alpha pathway. J. Nutr. Biochem. 2020, 77, 108297. [Google Scholar] [CrossRef] [PubMed]
  63. Hsu, Y.-J.; Ho, C.-S.; Lee, M.-C.; Ho, C.-S.; Huang, C.-C.; Kan, N.-W. Protective Effects of Resveratrol Supplementation on Contusion Induced Muscle Injury. Int. J. Med. Sci. 2020, 17, 53–62. [Google Scholar] [CrossRef]
  64. Gabr, A.M.M.; Fayek, N.M.; Mahmoud, H.M.; El-Bahr, M.K.; Ebrahim, H.S.; Sytar, O.; El-Halawany, A.M. Effect of Light Quality and Media Components on Shoot Growth, Rutin, and Quercetin Production from Common Buckwheat. ACS Omega 2022, 7, 26566–26572. [Google Scholar] [CrossRef]
  65. Paulina Luna-Castillo, K.; Lin, S.; Francisco Munoz-Valle, J.; Vizmanos, B.; Lopez-Quintero, A.; Marquez-Sandoval, F. Functional Food and Bioactive Compounds on the Modulation of the Functionality of HDL-C: A Narrative Review. Nutrients 2021, 13, 1165. [Google Scholar] [CrossRef]
  66. Chen, X.; Liang, D.; Huang, Z.; Jia, G.; Zhao, H.; Liu, G. Quercetin regulates skeletal muscle fiber type switching via adiponectin signaling. Food Funct. 2021, 12, 2693–2702. [Google Scholar] [CrossRef] [PubMed]
  67. Iwabu, M.; Yamauchi, T.; Okada-Iwabu, M.; Sato, K.; Nakagawa, T.; Funata, M.; Yamaguchi, M.; Namiki, S.; Nakayama, R.; Tabata, M.; et al. Adiponectin and AdipoR1 regulate PGC-1 alpha and mitochondria by Ca2+ and AMPK/SIRT1. Nature 2010, 464, 1313–1319. [Google Scholar] [CrossRef] [PubMed]
  68. Kim, Y.; Kim, C.-S.; Joe, Y.; Chung, H.T.; Ha, T.Y.; Yu, R. Quercetin Reduces Tumor Necrosis Factor Alpha-Induced Muscle Atrophy by Upregulation of Heme Oxygenase-1. J. Med. Food 2018, 21, 551–559. [Google Scholar] [CrossRef]
  69. Mukai, R.; Matsui, N.; Fujikura, Y.; Matsumoto, N.; Hou, D.-X.; Kanzaki, N.; Shibata, H.; Horikawa, M.; Iwasa, K.; Hirasaka, K.; et al. Preventive effect of dietary quercetin on disuse muscle atrophy by targeting mitochondria in denervated mice. J. Nutr. Biochem. 2016, 31, 67–76. [Google Scholar] [CrossRef]
  70. Dai, X.; Ding, Y.; Zhang, Z.; Cai, X.; Bao, L.; Li, Y. Quercetin But Not Quercitrin Ameliorates Tumor Necrosis Factor-Alpha-Induced Insulin Resistance in C2C12 Skeletal Muscle Cells. Biol. Pharm. Bull. 2013, 36, 788–795. [Google Scholar] [CrossRef] [Green Version]
  71. Chen, C.; Yang, J.-S.; Lu, C.-C.; Chiu, Y.-J.; Chen, H.-C.; Chung, M.-I.; Wu, Y.-T.; Chen, F.-A. Effect of Quercetin on Dexamethasone-Induced C2C12 Skeletal Muscle Cell Injury. Molecules 2020, 25, 3267. [Google Scholar] [CrossRef]
  72. Liang, Q.; Chen, Y.; Li, C.; Lu, L. Quercetin attenuates Ox-LDL-induced calcification in vascular smooth muscle cells by regulating ROS-TLR4 signaling pathway. Nan Fang Yi Ke Da Xue Xue Bao J. South. Med. Univ. 2018, 38, 980–985. [Google Scholar] [CrossRef]
  73. Chen, X.; Liang, D.; Huang, Z.; Jia, G.; Zhao, H.; Liu, G. Anti-fatigue effect of quercetin on enhancing muscle function and antioxidant capacity. J. Food Biochem. 2021, 45, e13968. [Google Scholar] [CrossRef] [PubMed]
  74. Finocchiaro, F.; Ferrari, B.; Gianinetti, A.; Dall’Asta, C.; Galaverna, G.; Scazzina, F.; Pellegrini, N. Characterization of antioxidant compounds of red and white rice and changes in total antioxidant capacity during processing. Mol. Nutr. Food Res. 2007, 51, 1006–1019. [Google Scholar] [CrossRef]
  75. Li, X.; Sui, Y.; Wu, Q.; Xie, B.; Sun, Z. Attenuated mTOR Signaling and Enhanced Glucose Homeostasis by Dietary Supplementation with Lotus Seedpod Oligomeric Procyanidins in Streptozotocin (STZ)-Induced Diabetic Mice. J. Agric. Food Chem. 2017, 65, 3801–3810. [Google Scholar] [CrossRef]
  76. Bowser, S.M.; Moore, W.T.; McMillan, R.P.; Dorenkott, M.R.; Goodrich, K.M.; Ye, L.; O’Keefe, S.F.; Hulver, M.W.; Neilson, A.P. High-molecular-weight cocoa procyanidins possess enhanced insulin-enhancing and insulin mimetic activities in human primary skeletal muscle cells compared to smaller procyanidins. J. Nutr. Biochem. 2017, 39, 48–58. [Google Scholar] [CrossRef]
  77. Yamashita, Y.; Wang, L.; Nanba, F.; Ito, C.; Toda, T.; Ashida, H. Procyanidin Promotes Translocation of Glucose Transporter 4 in Muscle of Mice through Activation of Insulin and AMPK Signaling Pathways. PLoS ONE 2016, 11, e0161704. [Google Scholar] [CrossRef] [PubMed]
  78. Corol, D.-I.; Ravel, C.; Raksegi, M.; Bedo, Z.; Charmet, G.; Beale, M.H.; Shewry, P.R.; Ward, J.L. Effects of Genotype and Environment on the Contents of Betaine, Choline, and Trigonelline in Cereal Grains. J. Agric. Food Chem. 2012, 60, 5471–5481. [Google Scholar] [CrossRef]
  79. Guo, H.; Guo, J.; Jiang, X.; Li, Z.; Ling, W. Cyanidin-3-O-beta-glucoside, a typical anthocyanin, exhibits antilipolytic effects in 3T3-L1 adipocytes during hyperglycemia: Involvement of FoxO1-mediated transcription of adipose triglyceride lipase. Food Chem. Toxicol. 2012, 50, 3040–3047. [Google Scholar] [CrossRef]
  80. Matsukawa, T.; Inaguma, T.; Han, J.; Villareal, M.O.; Isoda, H. Cyanidin-3-glucoside derived from black soybeans ameliorate type 2 diabetes through the induction of differentiation of preadipocytes into smaller and insulin-sensitive adipocytes. J. Nutr. Biochem. 2015, 26, 860–867. [Google Scholar] [CrossRef]
  81. Giang Thanh Thi, H.; Kase, E.T.; Wangensteen, H.; Barsett, H. Phenolic Elderberry Extracts, Anthocyanins, Procyanidins, and Metabolites Influence Glucose and Fatty Acid Uptake in Human Skeletal Muscle Cells. J. Agric. Food Chem. 2017, 65, 2677–2685. [Google Scholar] [CrossRef]
  82. Matsukawa, T.; Motojima, H.; Sato, Y.; Takahashi, S.; Villareal, M.O.; Isoda, H. Upregulation of skeletal muscle PGC-1 alpha through the elevation of cyclic AMP levels by Cyanidin-3-glucoside enhances exercise performance. Sci. Rep. 2017, 7, 44799. [Google Scholar] [CrossRef] [PubMed]
  83. Dinelli, G.; Segura Carretero, A.; Di Silvestro, R.; Marotti, I.; Fu, S.; Benedettelli, S.; Ghiselli, L.; Fernandez Gutierrez, A. Determination of phenolic compounds in modern and old varieties of durum wheat using liquid chromatography coupled with time-of-flight mass spectrometry. J. Chromatogr. A 2009, 1216, 7229–7240. [Google Scholar] [CrossRef]
  84. Hes, M.; Dziedzic, K.; Gorecka, D.; Drozdzynska, A.; Gujska, E. Effect of Boiling in Water of Barley and Buckwheat Groats on the Antioxidant Properties and Dietary Fiber Composition. Plant Foods Hum. Nutr. 2014, 69, 276–282. [Google Scholar] [CrossRef]
  85. Li, P.; Liu, A.; Xiong, W.; Lin, H.; Xiao, W.; Huang, J.; Zhang, S.; Liu, Z. Catechins enhance skeletal muscle performance. Crit. Rev. Food Sci. Nutr. 2020, 60, 515–528. [Google Scholar] [CrossRef] [PubMed]
  86. Kim, A.R.; Kim, K.M.; Byun, M.R.; Hwang, J.-H.; Park, J.I.; Oh, H.T.; Kim, H.K.; Jeong, M.G.; Hwang, E.S.; Hong, J.-H. Catechins activate muscle stem cells by Myf5 induction and stimulate muscle regeneration. Biochem. Biophys. Res. Commun. 2017, 489, 142–148. [Google Scholar] [CrossRef] [PubMed]
  87. Li, P.; Liu, A.; Liu, C.; Qu, Z.; Xiao, W.; Huang, J.; Liu, Z.; Zhang, S. Role and mechanism of catechin in skeletal muscle cell differentiation. J. Nutr. Biochem. 2019, 74, 108225–108260. [Google Scholar] [CrossRef]
  88. Huettemann, M.; Lee, I.; Perkins, G.A.; Britton, S.L.; Koch, L.G.; Malek, M.H. (-)-Epicatechin is associated with increased angiogenic and mitochondrial signalling in the hindlimb of rats selectively bred for innate low running capacity. Clin. Sci. 2013, 124, 663–674. [Google Scholar] [CrossRef]
  89. Yan, J.; Feng, Z.; Liu, J.; Shen, W.; Wang, Y.; Wertz, K.; Weber, P.; Long, J.; Liu, J. Enhanced autophagy plays a cardinal role in mitochondrial dysfunction in type 2 diabetic Goto-Kakizaki (GK) rats: Ameliorating effects of (-)-epigallocatechin-3-gallate. J. Nutr. Biochem. 2012, 23, 716–724. [Google Scholar] [CrossRef] [PubMed]
  90. Kim, A.R.; Kim, K.M.; Byun, M.R.; Hwang, J.-H.; Park, J.I.; Oh, H.T.; Jeong, M.G.; Hwang, E.S.; Hong, J.-H. (-)-Epigallocatechin-3-gallate stimulates myogenic differentiation through TAZ activation. Biochem. Biophys. Res. Commun. 2017, 486, 378–384. [Google Scholar] [CrossRef]
  91. Meador, B.M.; Mirza, K.A.; Tian, M.; Skelding, M.B.; Reaves, L.A.; Edens, N.K.; Tisdale, M.J.; Pereira, S.L. The Green Tea Polyphenol Epigallocatechin-3-Gallate (EGCg) Attenuates Skeletal Muscle Atrophy in a Rat Model of Sarcopenia. J. Frailty Aging 2015, 4, 209–215. [Google Scholar] [CrossRef] [PubMed]
  92. Nagasawa, T.; Hayashi, H.; Fujimaki, N.; Nishizawa, N.; Kitts, D.D. Induction of oxidatively modified proteins in skeletal muscle by electrical stimulation and its suppression by dietary supplementation of (-)-epigallocatechin gallate. Biosci. Biotechnol. Biochem. 2000, 64, 1004–1010. [Google Scholar] [CrossRef]
  93. Li, Y.; Ma, D.; Sun, D.; Wang, C.; Zhang, J.; Xie, Y.; Guo, T. Total phenolic, flavonoid content, and antioxidant activity of flour, noodles, and steamed bread made from different colored wheat grains by three milling methods. Crop J. 2015, 3, 328–334. [Google Scholar] [CrossRef]
  94. Maria Gomez-Caravaca, A.; Segura-Carretero, A.; Fernandez-Gutierrez, A.; Caboni, M.F. Simultaneous Determination of Phenolic Compounds and Saponins in Quinoa (Chenopodium quinoa Willd) by a Liquid Chromatography-Diode Array Detection-Electrospray Ionization-Time-of-Flight Mass Spectrometry Methodology. J. Agric. Food Chem. 2011, 59, 10815–10825. [Google Scholar] [CrossRef] [PubMed]
  95. Sheu, J.R.; Hsiao, G.; Chou, P.H.; Shen, M.Y.; Chou, D.S. Mechanisms involved in the antiplatelet activity of rutin, a glycoside of the flavonol quercetin, in human platelets. J. Agric. Food Chem. 2004, 52, 4414–4418. [Google Scholar] [CrossRef] [PubMed]
  96. Kim, J.Y.; Hickner, R.C.; Cortright, R.L.; Dohm, G.L.; Houmard, J.A. Lipid oxidation is reduced in obese human skeletal muscle. Am. J. Physiol.-Endocrinol. Metab. 2000, 279, E1039–E1044. [Google Scholar] [CrossRef] [PubMed]
  97. Seo, S.; Lee, M.-S.; Chang, E.; Shin, Y.; Oh, S.; Kim, I.-H.; Kim, Y. Rutin Increases Muscle Mitochondrial Biogenesis with AMPK Activation in High-Fat Diet-Induced Obese Rats. Nutrients 2015, 7, 8152–8169. [Google Scholar] [CrossRef]
  98. Su, K.-Y.; Yu, C.Y.; Chen, Y.-W.; Huang, Y.-T.; Chen, C.-T.; Wu, H.-F.; Chen, Y.-L.S. Rutin, a Flavonoid and Principal Component of Saussurea Involucrata, Attenuates Physical Fatigue in a Forced Swimming Mouse Model. Int. J. Med. Sci. 2014, 11, 528–537. [Google Scholar] [CrossRef] [PubMed]
  99. Liu, S.; Adewole, D.; Yu, L.; Sid, V.; Wang, B.; Karmin, O.; Yang, C. Rutin attenuates inflammatory responses induced by lipopolysaccharide in an in vitro mouse muscle cell (C2C12) model. Poult. Sci. 2019, 98, 2756–2764. [Google Scholar] [CrossRef]
  100. Weber, D.; Grune, T. The contribution of beta-carotene to vitamin A supply of humans. Mol. Nutr. Food Res. 2012, 56, 251–258. [Google Scholar] [CrossRef]
  101. Banwo, K.; Olojede, A.O.; Adesulu-Dahunsi, A.T.; Verma, D.K.; Thakur, M.; Tripathy, S.; Singh, S.; Patel, A.R.; Gupta, A.K.; Aguilar, C.N.; et al. Functional importance of bioactive compounds of foods with Potential Health Benefits: A review on recent trends. Food Biosci. 2021, 43, 101320. [Google Scholar] [CrossRef]
  102. Loskutov, I.G.; Khlestkina, E.K. Wheat, Barley, and Oat Breeding for Health Benefit Components in Grain. Plants 2021, 10, 86. [Google Scholar] [CrossRef] [PubMed]
  103. Cheng, F.; Zhang, Q.; Yan, F.-F.; Wan, J.-F.; Lin, C.-S. Lutein protects against ischemia/reperfusion injury in rat skeletal muscle by modulating oxidative stress and inflammation. Immunopharmacol. Immunotoxicol. 2015, 37, 329–334. [Google Scholar] [CrossRef]
  104. Nagao, A. Metabolism of Carotenoids in Mammals. In Carotenoids: Biosynthetic and Biofunctional Approaches; Misawa, N., Ed.; Advances in Experimental Medicine and Biology; Springer: Singapore, 2021; Volume 1261, pp. 67–78. [Google Scholar]
  105. Kitakaze, T.; Harada, N.; Imagita, H.; Yamaji, R. beta-Carotene Increases Muscle Mass and Hypertrophy in the Soleus Muscle in Mice. J. Nutr. Sci. Vitaminol. 2015, 61, 481–487. [Google Scholar] [CrossRef] [PubMed]
  106. Foletta, V.C.; White, L.J.; Larsen, A.E.; Leger, B.; Russell, A.P. The role and regulation of MAFbx/atrogin-1 and MuRF1 in skeletal muscle atrophy. Pflug. Arch. -Eur. J. Physiol. 2011, 461, 325–335. [Google Scholar] [CrossRef] [PubMed]
  107. Ogawa, M.; Kariya, Y.; Kitakaze, T.; Yamaji, R.; Harada, N.; Sakamoto, T.; Hosotani, K.; Nakano, Y.; Inui, H. The preventive effect of beta-carotene on denervation-induced soleus muscle atrophy in mice. Br. J. Nutr. 2013, 109, 1349–1358. [Google Scholar] [CrossRef]
  108. Siurek, B.; Rosicka-Kaczmarek, J.; Nebesny, E. Bioactive compounds in cereal grains—Occurrence, structure, technological significance and nutritional benefits-a review. Food Sci. Technol. Int. 2012, 18, 559–568. [Google Scholar] [CrossRef]
  109. Borneo, R.; Leon, A.E. Whole grain cereals: Functional components and health benefits. Food Funct. 2012, 3, 110–119. [Google Scholar] [CrossRef]
  110. Lee, H.; Lim, Y. Tocotrienol-rich fraction supplementation reduces hyperglycemia-induced skeletal muscle damage through regulation of insulin signaling and oxidative stress in type 2 diabetic mice. J. Nutr. Biochem. 2018, 57, 77–85. [Google Scholar] [CrossRef]
  111. Razak, A.M.; Khor, S.C.; Jaafar, F.; Karim, N.A.; Makpol, S. Targeting myomiRs by tocotrienol-rich fraction to promote myoblast differentiation. Genes Nutr. 2018, 13, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Lim, J.J.; Zurinah, W.N.W.; Moul, V.; Norwahidah, A.K. Tocotrienol-Rich Fraction (TRF) Treatment Promotes Proliferation Capacity of Stress-Induced Premature Senescence Myoblasts and Modulates the Renewal of Satellite Cells: Microarray Analysis. Oxidative Med. Cell. Longev. 2019, 2019, 9141343. [Google Scholar] [CrossRef]
  113. Chung, E.; Campise, S.N.; Joiner, H.E.; Tomison, M.D.; Kaur, G.; Dufour, J.M.; Cole, L.; Ramalingam, L.; Moustaid-Moussa, N.; Shen, C.-L. Effect of annatto-extracted tocotrienols and green tea polyphenols on glucose homeostasis and skeletal muscle metabolism in obese male mice. J. Nutr. Biochem. 2019, 67, 36–43. [Google Scholar] [CrossRef] [PubMed]
  114. Jung, T.-D.; Shin, G.-H.; Kim, J.-M.; Choi, S.-I.; Lee, J.-H.; Lee, S.J.; Park, S.J.; Woo, K.S.; Oh, S.K.; Lee, O.-H. Comparative Analysis of gamma-Oryzanol, beta-Glucan, Total Phenolic Content and Antioxidant Activity in Fermented Rice Bran of Different Varieties. Nutrients 2017, 9, 571. [Google Scholar] [CrossRef]
  115. Prasadi, V.P.N.; Joye, I.J. Dietary Fibre from Whole Grains and Their Benefits on Metabolic Health. Nutrients 2020, 12, 3045. [Google Scholar] [CrossRef]
  116. Xu, C.; Lv, J.; Lo, Y.M.; Cui, S.W.; Hu, X.; Fan, M. Effects of oat beta-glucan on endurance exercise and its anti-fatigue properties in trained rats. Carbohydr. Polym. 2013, 92, 1159–1165. [Google Scholar] [CrossRef]
  117. Li, Y.; Fan, Y.; Pan, H.; Qian, H.; Qi, X.; Wu, G.; Zhang, H.; Xu, M.; Rao, Z.; Wang, L.; et al. Effects of functional beta-glucan on proliferation, differentiation, metabolism and its anti-fibrosis properties in muscle cells. Int. J. Biol. Macromol. 2018, 117, 287–293. [Google Scholar] [CrossRef]
  118. Brogi, L.; Marchese, M.; Cellerino, A.; Licitra, R.; Naef, V.; Mero, S.; Bibbiani, C.; Fronte, B. beta-Glucans as Dietary Supplement to Improve Locomotion and Mitochondrial Respiration in a Model of Duchenne Muscular Dystrophy. Nutrients 2021, 13, 1619. [Google Scholar] [CrossRef] [PubMed]
  119. Ratseewo, J.; Meeso, N.; Siriamornpun, S. Changes in amino acids and bioactive compounds of pigmented rice as affected by far-infrared radiation and hot air drying. Food Chem. 2020, 306, 125644. [Google Scholar] [CrossRef]
  120. Zhu, D.; Sanchez-Ferrer, A.; Nystroem, L. Antioxidant Activity of Individual Steryl Ferulates from Various Cereal Grain Sources. J. Nat. Prod. 2016, 79, 308–316. [Google Scholar] [CrossRef] [PubMed]
  121. Ahn, J.; Son, H.J.; Seo, H.D.; Ha, T.Y.; Ahn, J.; Lee, H.; Shin, S.H.; Jung, C.H.; Jang, Y.J. gamma-Oryzanol Improves Exercise Endurance and Muscle Strength by Upregulating PPAR delta and ERR gamma Activity in Aged Mice. Mol. Nutr. Food Res. 2021, 65, 2000652. [Google Scholar] [CrossRef] [PubMed]
  122. Mattei, L.; Francisqueti-Ferron, F.V.; Garcia, J.L.; Ferron, A.J.T.; Silva, C.C.V.d.A.; Gregolin, C.S.; Nakandakare-Maia, E.T.; Silva, J.d.C.P.; Moreto, F.; Minatel, I.O.; et al. Antioxidant and anti-inflammatory properties of gamma- oryzanol attenuates insulin resistance by increasing GLUT-4 expression in skeletal muscle of obese animals. Mol. Cell. Endocrinol. 2021, 537, 111423. [Google Scholar] [CrossRef]
  123. Dahleh, M.M.M.; Araujo, S.M.; Bortolotto, V.C.; Pinheiro, F.C.; Poetini, M.R.; Musachio, E.A.S.; Meichtry, L.B.; Couto, S.d.F.; Prigol, M. Exercise associated with gamma-oryzanol supplementation suppresses oxidative stress and prevents changes in locomotion in Drosophila melanogaster. Free. Radic. Res. 2021, 55, 198–209. [Google Scholar] [CrossRef]
  124. Chen, C.Y.O.; Kamil, A.; Blumberg, J.B. Phytochemical composition and antioxidant capacity of whole wheat products. Int. J. Food Sci. Nutr. 2015, 66, 63–70. [Google Scholar] [CrossRef] [PubMed]
  125. Ayaz, M.; Junaid, M.; Ullah, F.; Subhan, F.; Sadiq, A.; Ali, G.; Ovais, M.; Shahid, M.; Ahmad, A.; Wadood, A.; et al. Anti-Alzheimer’s Studies on beta-Sitosterol Isolated from Polygonum hydropiper L. Front. Pharmacol. 2017, 8, 697. [Google Scholar] [CrossRef] [PubMed]
  126. Wong, H.S.; Leong, P.K.; Chen, J.; Leung, H.Y.; Chan, W.M.; Ko, K.M. beta-Sitosterol increases mitochondrial electron transport by fluidizing mitochondrial membranes and enhances mitochondrial responsiveness to increasing energy demand by the induction of uncoupling in C2C12 myotubes. J. Funct. Foods 2016, 23, 253–260. [Google Scholar] [CrossRef]
  127. Cheng, Y.; Chen, Y.; Li, J.; Qu, H.; Zhao, Y.; Wen, C.; Zhou, Y. Dietary beta-Sitosterol Improves Growth Performance, Meat Quality, Antioxidant Status, and Mitochondrial Biogenesis of Breast Muscle in Broilers. Animals 2019, 9, 71. [Google Scholar] [CrossRef]
  128. Hwang, S.-L.; Kim, H.-N.; Jung, H.-H.; Kim, J.-E.; Choi, D.-K.; Hur, J.-M.; Lee, J.-Y.; Song, H.; Song, K.-S.; Huh, T.-L. Beneficial effects of beta-sitosterol on glucose and lipid metabolism in L6 myotube cells are mediated by AMP-activated protein kinase. Biochem. Biophys. Res. Commun. 2008, 377, 1253–1258. [Google Scholar] [CrossRef] [PubMed]
  129. Pei, J.; Prasad, M.; Mohamed Helal, G.; El-Sherbiny, M.; Abdelmonem Elsherbini, D.M.; Rajagopal, P.; Palanisamy, C.P.; Veeraraghavan, V.P.; Jayaraman, S.; Surapaneni, K.M. Beta-Sitosterol Facilitates GLUT4 Vesicle Fusion on the Plasma Membrane via the Activation of Rab/IRAP/Munc 18 Signaling Pathways in Diabetic Gastrocnemius Muscle of Adult Male Rats. Bioinorg. Chem. Appl. 2022, 2022, 7772305. [Google Scholar] [CrossRef] [PubMed]
  130. Dianita, R.; Jantan, I.; Amran, A.Z.; Jalil, J. Protective Effects of Labisia pumila var. alata on Biochemical and Histopathological Alterations of Cardiac Muscle Cells in Isoproterenol-Induced Myocardial Infarction Rats. Molecules 2015, 20, 4746–4763. [Google Scholar] [CrossRef]
  131. Wang, Z.; Li, Q.; Hao, Y.; Wang, Z.; Yang, H.; Liu, J.; Wang, J. Protective effect of 5-heptadecylresorcinol against obesity-associated skeletal muscle dysfunction by modulating mitochondrial biogenesis via the activation of SIRT3/PGC-1α signaling pathway. J. Funct. Foods 2022, 95, 105178. [Google Scholar] [CrossRef]
  132. Filipcev, B.; Kojic, J.; Krulj, J.; Bodroza-Solarov, M.; Ilic, N. Betaine in Cereal Grains and Grain-Based Products. Foods 2018, 7, 49. [Google Scholar] [CrossRef]
  133. Ismaeel, A. Effects of betaine supplementation on muscle strength and power: A systematic review. J. Strength Cond. Res. 2017, 31, 2338–2346. [Google Scholar] [CrossRef] [PubMed]
  134. Senesi, P.; Luzi, L.; Montesano, A.; Mazzocchi, N.; Terruzzi, I. Betaine supplement enhances skeletal muscle differentiation in murine myoblasts via IGF-1 signaling activation. J. Transl. Med. 2013, 11, 174. [Google Scholar] [CrossRef]
  135. Apicella, J.M.; Lee, E.C.; Bailey, B.L.; Saenz, C.; Anderson, J.M.; Craig, S.A.S.; Kraemer, W.J.; Volek, J.S.; Maresh, C.M. Betaine supplementation enhances anabolic endocrine and Akt signaling in response to acute bouts of exercise. Eur. J. Appl. Physiol. 2013, 113, 793–802. [Google Scholar] [CrossRef]
  136. Du, J.; Shen, L.; Zhang, P.; Tan, Z.; Cheng, X.; Luo, J.; Zhao, X.; Yang, Q.; Gu, H.; Jiang, A.a.; et al. The regulation of skeletal muscle fiber-type composition by betaine is associated with NFATc1/MyoD. J. Mol. Med. 2018, 96, 685–700. [Google Scholar] [CrossRef]
  137. Ma, J.; Meng, X.; Kang, S.Y.; Zhang, J.; Jung, H.W.; Park, Y.-K. Regulatory effects of the fruit extract of Lycium chinense and its active compound, betaine, on muscle differentiation and mitochondrial biogenesis in C2C12 cells. Biomed. Pharmacother. 2019, 118, 109297. [Google Scholar] [CrossRef] [PubMed]
  138. Jung, D.M.; Lee, M.J.; Yoon, S.H.; Jung, M.Y. A Gas Chromatography-Tandem Quadrupole Mass Spectrometric Analysis of Policosanols in Commercial Vegetable Oils. J. Food Sci. 2011, 76, C891–C899. [Google Scholar] [CrossRef]
  139. Irmak, S.; Dunford, N.T. Policosanol contents and compositions of wheat varieties. J. Agric. Food Chem. 2005, 53, 5583–5586. [Google Scholar] [CrossRef]
  140. Kabir, Y.; Kimura, S. Tissue distribution of (8-14C)-octacosanol in liver and muscle of rats after serial administration. Ann. Nutr. Metab. 1995, 39, 279–284. [Google Scholar] [CrossRef] [PubMed]
  141. Kim, H.; Park, S.; Han, D.S.; Park, T. Octacosanol supplementation increases running endurance time and improves biochemical parameters after exhaustion in trained rats. J. Med. Food 2003, 6, 345–351. [Google Scholar] [CrossRef]
  142. Kabir, Y.; Kimura, S. Biodistribution and metabolism of orally administered octacosanol in rats. Ann. Nutr. Metab. 1993, 37, 33–38. [Google Scholar] [CrossRef]
  143. Zhou, Y.; Cao, F.; Wu, Q.; Luo, Y.; Guo, T.; Han, S.; Huang, M.; Hu, Z.; Bai, J.; Luo, F.; et al. Dietary Supplementation of Octacosanol Improves Exercise-Induced Fatigue and Its Molecular Mechanism. J. Agric. Food Chem. 2021, 69, 7603–7618. [Google Scholar] [CrossRef] [PubMed]
  144. Frampton, J.; Murphy, K.G.; Frost, G.; Chambers, E.S. Higher dietary fibre intake is associated with increased skeletal muscle mass and strength in adults aged 40 years and older. J. Cachexia Sarcopenia Muscle 2021, 12, 2134–2144. [Google Scholar] [CrossRef] [PubMed]
  145. Kaviani, M.; Chilibeck, P.D.; Jochimi, J.; Gordon, J.; Zello, G.A. The Glycemic Index of Sport Nutrition Bars Affects Performance and Metabolism During Cycling and Next-Day Recovery. J. Hum. Kinet. 2019, 66, 69–79. [Google Scholar] [CrossRef] [PubMed]
  146. Nowotny, B.; Zahiragic, L.; Bierwagen, A.; Kabisch, S.; Groener, J.B.; Nowotny, P.J.; Fleitmann, A.K.; Herder, C.; Pacini, G.; Erlund, I.; et al. Low-energy diets differing in fibre, red meat and coffee intake equally improve insulin sensitivity in type 2 diabetes: A randomised feasibility trial. Diabetologia 2015, 58, 255–264. [Google Scholar] [CrossRef] [PubMed]
  147. Lee, J.Y.; Lee, S. Dietary Patterns Related to Appendicular Skeletal Muscle Mass: The Korea National Health and Nutrition Examination Survey 2008-2011. J. Am. Coll. Nutr. 2019, 38, 358–363. [Google Scholar] [CrossRef] [PubMed]
  148. Mafi, F.; Biglari, S.; Afousi, A.G.; Gaeini, A.A. Improvement in Skeletal Muscle Strength and Plasma Levels of Follistatin and Myostatin Induced by an 8-Week Resistance Training and Epicatechin Supplementation in Sarcopenic Older Adults. J. Aging Phys. Act. 2019, 27, 384–391. [Google Scholar] [CrossRef]
  149. Taub, P.R.; Ramirez-Sanchez, I.; Ciaraldi, T.P.; Perkins, G.; Murphy, A.N.; Naviaux, R.; Hogan, M.; Maisel, A.S.; Henry, R.R.; Ceballos, G.; et al. Alterations in Skeletal Muscle Indicators of Mitochondrial Structure and Biogenesis in Patients with Type 2 Diabetes and Heart Failure: Effects of Epicatechin Rich Cocoa. Clin. Transl. Sci. 2012, 5, 43–47. [Google Scholar] [CrossRef]
  150. Eggersdorfer, M.; Wyss, A. Carotenoids in human nutrition and health. Arch. Biochem. Biophys. 2018, 652, 18–26. [Google Scholar] [CrossRef]
  151. Villani, A.; Wright, H.; Slater, G.; Buckley, J. A randomised controlled intervention study investigating the efficacy of carotenoid-rich fruits and vegetables and extra-virgin olive oil on attenuating sarcopenic symptomology in overweight and obese older adults during energy intake restriction: Protocol paper. BMC Geriatr. 2018, 18, 2. [Google Scholar] [CrossRef] [Green Version]
  152. Semba, R.D.; Lauretani, F.; Ferrucci, L. Carotenoids as protection against sarcopenia in older adults. Arch. Biochem. Biophys. 2007, 458, 141–145. [Google Scholar] [CrossRef]
  153. Harari, A.; Coster, A.C.F.; Jenkins, A.; Xu, A.; Greenfield, J.R.; Harats, D.; Shaish, A.; Samocha-Bonet, D. Obesity and Insulin Resistance Are Inversely Associated with Serum and Adipose Tissue Carotenoid Concentrations in Adults. J. Nutr. 2020, 150, 38–46. [Google Scholar] [CrossRef]
  154. Long, J.-A.; Zhong, R.-H.; Chen, S.; Wang, F.; Luo, Y.; Lu, X.-T.; Yishake, D.; Chen, Y.-M.; Fang, A.-P.; Zhu, H.-L. Dietary betaine intake is associated with skeletal muscle mass change over 3 years in middle-aged adults: The Guangzhou Nutrition and Health Study. Br. J. Nutr. 2021, 125, 440–447. [Google Scholar] [CrossRef] [PubMed]
  155. Arazi, H.; Aboutalebi, S.; Taati, B.; Cholewa, J.M.; Candow, D.G. Effects of short-term betaine supplementation on muscle endurance and indices of endocrine function following acute high-intensity resistance exercise in young athletes. J. Int. Soc. Sports Nutr. 2022, 19, 1–16. [Google Scholar] [CrossRef] [PubMed]
  156. Cholewa, J.M.; Wyszczelska-Rokiel, M.; Glowacki, R.; Jakubowski, H.; Matthews, T.; Wood, R.; Craig, S.A.S.; Paolone, V. Effects of betaine on body composition, performance, and homocysteine thiolactone. J. Int. Soc. Sports Nutr. 2013, 10, 39. [Google Scholar] [CrossRef] [PubMed]
  157. Hah, Y.-S.; Lee, W.K.; Lee, S.; Kim, E.J.; Lee, J.H.; Lee, S.-J.; Ji, Y.H.; Kim, S.G.; Lee, H.-H.; Hong, S.Y.; et al. beta-Sitosterol Attenuates Dexamethasone-Induced Muscle Atrophy via Regulating FoxO1-Dependent Signaling in C2C12 Cell and Mice Model. Nutrients 2022, 14, 2894. [Google Scholar] [CrossRef]
Figure 1. Signaling pathways involved in the regulation of skeletal muscle diseases. → indicates promotion; T indicates inhibition.
Figure 1. Signaling pathways involved in the regulation of skeletal muscle diseases. → indicates promotion; T indicates inhibition.
Foods 11 02752 g001
Figure 2. Bioactive components of whole grains for improving skeletal muscle function.
Figure 2. Bioactive components of whole grains for improving skeletal muscle function.
Foods 11 02752 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Li, Q.; Yang, H.; Song, S.; Liu, J.; Wang, Z.; Wang, J. Bioactive Components in Whole Grains for the Regulation of Skeletal Muscle Function. Foods 2022, 11, 2752. https://doi.org/10.3390/foods11182752

AMA Style

Li Q, Yang H, Song S, Liu J, Wang Z, Wang J. Bioactive Components in Whole Grains for the Regulation of Skeletal Muscle Function. Foods. 2022; 11(18):2752. https://doi.org/10.3390/foods11182752

Chicago/Turabian Style

Li, Qing, Haihong Yang, Shuimiao Song, Jie Liu, Ziyuan Wang, and Jing Wang. 2022. "Bioactive Components in Whole Grains for the Regulation of Skeletal Muscle Function" Foods 11, no. 18: 2752. https://doi.org/10.3390/foods11182752

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop