Next Article in Journal
Exploring the Antioxidant Potential of Talisia esculenta Using In Vitro and In Vivo Approaches
Previous Article in Journal
Postpartum Depression Is Associated with Maternal Sociodemographic and Anthropometric Characteristics, Perinatal Outcomes, Breastfeeding Practices, and Mediterranean Diet Adherence
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances in Nutraceuticals for the Treatment of Sarcopenic Obesity

Department of Food and Nutrition, Hanyang University, Seoul 04763, Republic of Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2023, 15(17), 3854; https://doi.org/10.3390/nu15173854
Submission received: 5 August 2023 / Revised: 26 August 2023 / Accepted: 1 September 2023 / Published: 4 September 2023

Abstract

:
Sarcopenic obesity, low muscle mass, and high body fat are growing health concerns in the aging population. This review highlights the need for standardized criteria and explores nutraceuticals as potential therapeutic agents. Sarcopenic obesity is associated with insulin resistance, inflammation, hormonal changes, and reduced physical activity. These factors lead to impaired muscle activity, intramuscular fat accumulation, and reduced protein synthesis, resulting in muscle catabolism and increased fat mass. Myostatin and irisin are myokines that regulate muscle synthesis and energy expenditure, respectively. Nutritional supplementation with vitamin D and calcium is recommended for increasing muscle mass and reducing body fat content. Testosterone therapy decreases fat mass and improves muscle strength. Vitamin K, specifically menaquinone-4 (MK-4), improves mitochondrial function and reduces muscle damage. Irisin is a hormone secreted during exercise that enhances oxidative metabolism, prevents insulin resistance and obesity, and improves bone quality. Low-glycemic-index diets and green cardamom are potential methods for managing sarcopenic obesity. In conclusion, along with exercise and dietary support, nutraceuticals, such as vitamin D, calcium, vitamin K, and natural agonists of irisin or testosterone, can serve as promising future therapeutic alternatives.

1. Introduction

As the world population ages, the incidence of both obesity and metabolic dysfunction increases, resulting in an increase in frailty and sarcopenia [1]. Elderly individuals are particularly susceptible to sarcopenia, a condition characterized by reduced muscle mass and strength. In 2022, the worldwide prevalence of sarcopenia was estimated to be between 10% and 27% in individuals aged 68.5 years [2]. Simultaneously, obesity rates have steadily increased from 1980 to 2019, with a worldwide prevalence of 14.0% in 2019. Compared to 1980, this was an increase of 9.4% [3]. The coexistence of sarcopenia and obesity, known as “sarcopenic obesity” (SO), is a relatively new concept that describes the simultaneous increase in fat mass and decrease in muscle mass in individuals [4]. Summarizing data from 50 studies updated in PubMed, Embase, and the Web of Science, the worldwide prevalence of SO has been reported to be 11% [5].
Sarcopenia is a term derived from the combination of the Greek words “sarx,” meaning flesh, and “penia,” meaning reduction [6]. Although there are no established criteria for diagnosing SO, it is associated with various adverse health outcomes, including physical disability, increased risk of falls, and poor quality of life [7]. Therefore, it is crucial to establish standardized criteria for identifying and treating SO. The lack of bioactive compounds and nutraceuticals that could be used to treat SO remains an issue. This review investigated the feasible diagnostic criteria, underlying causes, and viable management options for SO. Additionally, this article addresses ongoing research on the application of nutraceuticals as a potential therapeutic approach for SO.

2. Definition and Diagnosis

SO is diagnosed when muscle loss and fat mass gain co-occur. The definition of SO is based on the individual definitions of sarcopenia and obesity; however, an accurate definition has not yet been established. Although various definitions of sarcopenia have been proposed, a consensus has yet to be reached. Various diagnostic criteria for SO have been proposed by organizations and researchers [7]. To explore how SO is currently diagnosed, the definitions and diagnoses of sarcopenia and obesity are considered separately. Individuals meeting both the sarcopenia and obesity criteria were diagnosed with SO.
In a cross-sectional study conducted in 1998, the diagnostic criteria for sarcopenia in a sample of 883 elderly individuals were included in the New Mexico Elder Health Survey (NMEHS). They defined sarcopenia as a decrease in the relative skeletal muscle index and calculated as appendicular skeletal muscle mass (ASM). It was divided by height squared (ASM/height2) of two or more standard deviations below the reference values for young and healthy individuals and measured by dual X-ray absorptiometry (DXA) (Table 1) [8]. The skeletal muscle index (SMI = skeletal muscle mass/body mass × 100) was measured using bioelectrical impedance analysis to define sarcopenia in elderly Americans [9]. The Health ABC Study proposed an alternative definition of sarcopenia using appendicular lean mass (ALM). The authors also validated whether the ASM/height2 index could appropriately measure sarcopenia. Individuals with a high body mass index (BMI) and obesity had increased fat mass and reduced muscle mass. However, most patients had high absolute muscle mass and did not exhibit sarcopenia. Thus, it was confirmed that SO may be underestimated by the ASM/height2 index [10].
Sarcopenia is currently defined based on muscle mass, muscle strength, and anthropometric measures, including mid-arm and calf circumference [7]. The European Working Group for the Study of Sarcopenia (EWGSOP) defines sarcopenia as low muscle mass and function (strength or performance) and has developed an algorithm using gait speed (<0.8 m/s) before the measurement of muscle mass or strength [11]. The International Working Group for the Study of Sarcopenia (IWGS) provided a consensus on the definition. It is a combination of low appendicular lean mass and poor physical functioning (gait speed ≤ 1 m/s) [12]. The Foundation for the National Institutes of Health (FNIH) Sarcopenia Project proposed a causal and indirect relationship between muscle mass and function based on the definition of sarcopenia. It suggested the assessment of low lean muscle mass using DXA and reduced muscle function using handgrip strength [13]. The Asian Working Group for Sarcopenia suggested a diagnostic algorithm based on the available evidence in Asia. They followed the EWGSOP approach to define sarcopenia, which included handgrip strength and gait speed during the initial screening [14]. In 2018, the renewed EWGSOP2 was targeted to increase the early detection and treatment of sarcopenia. Low muscle strength was adopted as the main determinant of sarcopenia since muscle strength is regarded as a better criterion than muscle mass in predicting adverse consequences. The EWGSOP2 focuses on low muscle strength, using grip strength as the main parameter of sarcopenia. They used low muscle quantity and quality to confirm the diagnosis of sarcopenia and proposed measures of physical performance to assess its severity [15]. Although definitions of sarcopenia have been proposed, the lack of consensus requires further research to establish a standardized definition.
Obesity is a chronic disease in which the accumulation of excessive body fat leads to metabolic diseases and increased mortality. The World Health Organization (WHO) defines obesity as a BMI of over 30 kg/m2 or a waist circumference of a minimum of 102 cm for men and 88 cm for women. East Asia has a lower BMI cut-off point for the definition of obesity (25 kg/m2) [16]. Although BMI is a useful indicator of body fat, it is preferable to assess body fat distribution to predict the risk of metabolic syndrome and cardiovascular disease [17]. The American Association of Clinical Endocrinology recommends using the WHO body fat thresholds for the diagnosis of obesity, with a minimum of 25% in men and 35% in women [18].
Table 1. Diagnostic criteria of sarcopenic obesity.
Table 1. Diagnostic criteria of sarcopenic obesity.
StudySarcopenia Diagnosis MethodMeasurement
(Cut-Off Points)
Obesity Diagnosis Method
(Cut-Off Points)
Baumgartner et al. [8]ASM divided by height squaredDXA (men < 7.26 kg/m2;
women < 5.45 kg/m2)
Body fat- men > 27%;
women > 38%
Newman et al.
[10]
ALM divided by height squaredDXA (men < 7.23 kg/m2;
women < 5.67 kg/m2)
BMI ≥ 30 kg/m2
Cruz-Jentoft [11],
EWGSOP
ASM divided by height squaredDXA (men ≤ 7.26 kg/m2; women ≤ 5.50 kg/m2) (Rosetta study)
DXA (men ≤ 7.25 kg/m2; women ≤ 5.67 kg/m2) (Health ABC study)
DXA (men ≤ 7.23 kg/m2; women ≤ 5.67 kg/m2) (Health ABC study)
BIA (men: severe ≤ 8.50 kg/m2, moderate 8.51–10.75 kg/m2.
women: severe ≤ 5.75 kg/m2; moderate 5.76–6.75 kg/m2) (NHANES III study)
NA
ResidualsDXA (ALM (fat mass divided by height), men: −2.29; women: −1.73)
SMI divided by height squaredBIA (men ≤ 8.87 kg/m2; women ≤ 6.42 kg/m2)
Muscle strengthHandgrip strength (men < 30 kg; women < 20 kg)
Muscle strength based on BMI categoryHandgrip strength
(men: BMI ≤ 24: ≤ 29 kg, BMI 24.1–26: ≤ 30 kg
BMI 26.1–28: ≤ 30 kg, BMI > 28: ≤ 32 kg;
women: BMI ≤ 23: ≤ 17 kg, BMI 23.1–26: ≤ 17.3 kg
BMI 26.1–29: ≤ 18 kg, BMI > 29: ≤ 21 kg)
Physical performanceSPPB (≤8-point score)
Gait speed over 6 m (<1 m/s) (Health ABC study)
Gait speed over 6 m (<1.175 m/s) (Health ABC study)
Gait speed over 4 m (<0.8 m/s) (InCHIANTI study)
LK Chen [14],
AWGS
ASM divided by height squaredDXA (men < 7.0 kg/m2; women < 5.4 kg/m2)
BIA (men < 7.0 kg/m2; women < 5.7 kg/m2)
NA
StrengthHandgrip strength (men < 26 kg; women < 18 kg)
Physical performanceGait speed over 6 m (< 0.8 m/s)
Cruz-Jentoft [15],
EWGSOP2
ASMDXA/BIA (men < 20 kg; women < 15 kg)NA
ASM divided by height squaredDXA/BIA (men < 7.0 kg/m2; women < 5.5 kg/m2)
StrengthGrip Strength (men: < 27 kg; women < 16 kg)
Chair stand > 15 s for five rises
Physical performanceGait speed (≤ 0.8 m/s)
SPPB (≤ 8-point score)
TUG (≥ 20 s)
400 m walk test (non-completion or ≥ 6 min for completion)

3. Pathophysiology and Complications

Although the mechanisms and pathologies of SO are uncertain, multiple mechanisms have emerged as possible causes. Insulin resistance (IR), which is highly associated with type 2 diabetes mellitus (T2DM), serves as a connection between sarcopenia and obesity and frequently appears in aged and obese populations [19]. IR is stimulated by inflammatory molecules that directly intervene in the crosstalk between cytokines and insulin receptor signaling pathways [20,21]. Insulin is responsible for protein anabolism in muscle and adipose catabolism, and resistance to such signals inhibits protein synthesis while inducing proteolysis and adipose tissue growth (Figure 1) [19]. IR increases the size and number of adipocytes. Overall, the inhibition of protein anabolism and proneness to fat synthesis are consequences of insulin resistance and diabetes.
Another possible cause of SO is the inflammatory response and secretion of cytokines due to low-grade inflammation triggered by increased IR. Insulin resistance induces inflammation, and obesity activates macrophages, which leads to inflammation and the secretion of adipokines [22]. Inflammation in adipocytes leads to increased leptin, chemerin, and resistin levels and decreased adiponectin [6]. The increase in leptin induces upregulation of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) [21]. However, the effects of leptin are mediated by adiponectin [23]. Insulin-like growth factor-1 (IGF-1), responsible for the inhibition of autophagy of myocytes via Forkhead box O (FOXO) and mammalian target of rapamycin (mTOR), is suppressed by IL-6 and TNF-α [24,25]. IGF-1 promotes the proliferation of satellite cells, leading to muscle hypertrophy [26]. This decline prevents muscle growth and weakens muscle action. IGF-1 release is also inhibited by an increase in the concentration of free fatty acids [27]. Additionally, inflammation and oxidative stress also lead to the disposition of ectopic fat in muscles. Intramuscular fat accumulation causes mitochondrial impairment and an imbalance in myokines, inducing lipid peroxidation and impairing muscle action [6]. Similar to the outcomes of IR, inflammatory actions intermittently damage muscles and precipitate the accumulation of lipids, which are the two crucial causes of SO.
In addition, SO is related to sex hormones such as estrogen and testosterone. When women experience menopause, body weight and fat mass increase, especially in the abdominal region; however, a loss in lean mass is observed [28]. Inflammatory responses are stimulated during this period [29]. This promotes an increase in waist circumference and muscle reduction. These changes result from a decline in estrogen levels during menopause. In men, testosterone is responsible for muscle regeneration, protein synthesis through increased amino acid utilization, and the stimulation of androgen receptor expression [30]. Decreasing testosterone levels in men aged 65 years triggers muscle loss and reduces muscle strength and performance [21]. The reduction in testosterone and estrogen production in men and women provokes a decline in muscle mass while promoting fat production, ultimately resulting in SO.
As individuals age, there is a general decrease in food intake, particularly protein intake, which causes a decrease in muscle mass and strength. In addition to malnutrition, older adults tend to spend more time indoors than engaging in outdoor exercise. Muscle dystrophy occurs because of a lack of exercise, ultimately leading to more frequent indoor activity. Consequently, a decrease in the lean mass and an increase in the fat mass were observed. More time spent indoors leads to less exposure to the sun and, ultimately, reduced vitamin D [6]. Vitamin D deficiency causes type II muscle fiber atrophy, enlarged interfibrillar spaces, fat infiltration, and fibrosis [25]. Since vitamin D is responsible for protein synthesis, less exposure to sunlight inevitably leads to depressed muscle generation.
Myokine actions can also evoke SO, in addition to vitamin D, insulin, and inflammatory actions. Myostatin and irisin are currently being studied for their mechanisms and potential therapeutic approaches in muscle-wasting disorders, and it is expected that a better understanding of their mechanisms will contribute to the development of treatment and prevention strategies. Briefly, myostatin, the first discovered myokine, is expressed in the skeletal muscle to negatively regulate muscle synthesis. When myostatin is expressed, IGF-1, Akt, and mTOR are inhibited, and the FOXO1 pathway is stimulated to suppress glucose transporter 4 (GLUT4) and AMP-activated protein kinase (AMPK) [31,32]. Subsequently, the uptake of glucose by skeletal muscles is inhibited, leading to an increase in IR [33]. Myostatin is upregulated in obese humans and reduced after exercise. During physical activity, peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) drives the expression of fibronectin type III domain-containing protein 5 (FNDC5), secreting irisin, the contradictory myokine to myostatin [34,35,36]. Further explanations of irisin are provided in a later section. The level of irisin decreases with age; alternatively, the level of myostatin increases and vice versa [37]. In summary, the secretion of myostatin not only causes muscle degradation but also induces the inhibition of irisin secretion, the myokine responsible for muscle production and fat reduction.
Sarcopenia and obesity are factors associated with poor quality of life, especially in adults. Functional capability, physical balance, and mortality are associated with muscle strength and body weight [38]. These effects are amplified when body size, specifically body weight, is substantial for poor muscle strength [20]. Higher muscle mass was positively correlated with greater physical performance. This means that when muscle mass declines, there is a greater risk of falls and fractures [39]. Osteoporosis and osteoarthritis are the two most common types of skeletal damage induced by SO. These are reciprocal procedures; thus, the occurrence of one event blocks another. Another study also compared the statistics of falls and fractures that occur in men and women. According to the New Mexico Elder Health Survey, compared by ALM/height2, men have a 2.5-fold greater likelihood of falling. However, no such association was observed among women [40]. Although there is no clear evidence, the rate of falls and fractures is expected to be related to visceral adipose tissue and ectopic fat [41]. Visceral fat and ectopic fat, which stimulate the pro-inflammatory system in the human body, are predicted to increase the likelihood of falls and fractures in obese individuals [42]. Patients diagnosed with SO often experience frequent falls and fractures, resulting in poor quality of life.
Increased body mass index (BMI), body weight, and reduced muscle mass are potential indicators of skeletal injury, including osteoarthritis and osteoporosis. Sarcopenia induces muscle weakness by loading more pressure on the hips and knees, two major body parts associated with osteoarthritis due to sarcopenia [43]. Additionally, increased body weight due to obesity places a greater burden on the lower limbs [44]. However, the pathological relationship between sarcopenia and osteoarthritis remains unclear [45]. Furthermore, the association between the two was independent of age, sex, and vitamin D levels [46]. Estrogen decline is a major regulator of bone metabolism and has been identified as a factor in SO [47]. Thus, a decline in estrogen levels and reduced skeletal strength lead to postmenopausal osteoporosis [48]. In summary, increased fat mass and decreased muscle mass among SO patients inevitably impose greater pressure and stress on the lower joints and bones, thereby contributing to the development of osteoarthritis and osteoporosis.
Additionally, SO is associated with increased mortality rates. A high waist circumference and low muscle strength are correlated with increased mortality [49]. Moreover, muscle strength and mass were independent of the mortality rate. A study conducted by Health, Aging, and Body Composition reported increased mortality resulting from less strength in the quadriceps [7]. Obese individuals were more prone to cancer, infection, steatosis, cirrhosis, etcetera [50]. The American Medical Association confirmed that mortality increased as the obesity grade increased. No association was observed between mortality and grade 1 obesity. However, aside from this grade, there was an increase in mortality in response to higher obesity levels [51]. A meta-analysis indicated that SO resulted in a higher mortality rate than in healthy individuals [49]. Low muscle strength and mass, along with obesity, expose individuals to a higher risk of various diseases such as cancer and cirrhosis, thereby elevating their proneness to mortality.
Sarcopenia and obesity are associated with metabolic disorders. Therefore, SO may have a greater impact on the metabolic syndrome (MetS) and may be associated with mortality rates, including T2DM, nonalcoholic fatty liver disease (NAFLD), dyslipidemia, hypertension, and cardiovascular disease (CVD), than sarcopenia or obesity alone [52]. The Korea National Health and Nutrition Examination Survey (KNHANES) evaluated sarcopenia, defined by muscle mass, with accompanying obesity (BMI ≥ 25 kg/m2). They reported that individuals with SO had an increased risk of dyslipidemia and a positive association with insulin resistance, as defined by Homeostatic Model Assessment (HOMA) scores and triglyceride levels [53]. A Korean SO Study (KSOS) cohort study reported that SO, defined by DXA-measured ALM/weight (%) and visceral fat, was associated with IR as assessed by the Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) score, inflammation, C-reactive protein level, and vitamin D deficiency [54]. In summary, not only does SO increase mortality but also cause MetS in surviving individuals, depressing their quality of life.

4. Irisin and Its Role in Sarcopenic Obesity

4.1. Pathology and Implications of Irisin for Sarcopenia

Irisin is secreted from muscles during exercise and inhibits myostatin secretion. Regular exercise promotes the secretion of FNDC5, a protein that induces irisin production [55]. Irisin, a myokine fragment of FNDC5, decreases fasting glucose levels and facilitates beta-cell function (Figure 2) [56]. In addition to regular exercise, irisin can be produced under environmental stresses such as caloric restriction, coldness, and oxidative stress [57]. Under these conditions, PGC-1α promotes nuclear respiratory factor-1 (NRF-1) and NRF-2 expression. Consequently, the mitochondrial transcription factor (TFAM) is promoted, resulting in mitochondrial biogenesis, fusion, and fission [58,59].
Although it is still unclear which type of exercise facilitates irisin secretion, multiple studies have verified that blood irisin levels increase instantly even after compulsory training, although the minimum increase is only 1.2-fold, depending on the intensity of the exercise (Table 2) [60,61]. Irisin secretion is not only a single-way procedure but also an interactive procedure, meaning that its secretion increases muscle biogenesis and is simultaneously increased by muscle growth [62]. In in vitro experiments, C2C12 myotubes treated with irisin showed increased TFAM and PGC-1α and significantly increased oxidative metabolism [59,63]. When applied to human myocytes and adipocytes, PGC-1α was upregulated, along with increased IGF-1 and decreased myostatin gene expression through the mitogen-activated protein kinase (MAPK) pathway. Simultaneously, muscle cell differentiation leads to increased mRNA expression of FNDC5 and irisin secretion [64]. Overall, the secretion of irisin and its precursors, FNDC5 and PGC-1α, is promoted under physical activities or other stressful conditions, and reciprocally, muscle growth also induces irisin production.
As mentioned earlier, other possible consequences of SO include osteoporosis and osteoarthritis. As muscle mass decreases, bone mass is reduced correspondingly [70]. To address this, irisin can be used to enhance bone marrow stromal cell differentiation and osteoblast phosphorylation. This prevents torsion, increases the bone perimeter, and enhances overall bone quality [63,71]. In addition, one of the major genes that deteriorates bone health is sclerostin, a major osteoblast differentiation-interrupting factor, via its conjunction with low-density lipoprotein (LDL) receptor protein 5/6 (LRP5/6) and frizzled coreceptors on the osteoblast surface to inhibit catenin signaling pathways [72]. However, not only is irisin eligible for reducing myostatin secretion, but serum sclerostin levels have also been reported to be inversely correlated with serum irisin levels [73]. In addition to promoting muscle prosperity, irisin mediates osteoporosis and osteoarthritis, which are possible outcomes of SO.

4.2. Pathophysiology and Implications of Irisin for Obesity

While inducing muscle hypertrophy, irisin can also induce an increase in uncoupling protein-1 (UCP-1) and energy expenditure in adipocytes, resulting in reduced lipid accumulation [73]. Serum irisin levels tended to be lower in patients diagnosed with obesity, independent of NAFLD, than in those that were not obese [74]. Moreover, although a positive correlation was observed between fat mass and myostatin levels, fat mass was negatively correlated with irisin levels [75]. Among the various treatments for obesity, exercise and physical activity are the most commonly recognized and effective solutions, along with caloric restriction [76,77]. Physical exercise promotes irisin secretion, eventually resulting in weight loss [78]. Irisin increases energy expenditure through increased mRNA expression of UCP-1 [79]. Consequently, triggered muscle shivering accelerates browning and thermogenesis regulation in white adipose tissue [24,80]. As a result, irisin can potentially treat various metabolic diseases such as T2DM, CVD, and NAFLD, which are well-known consequences of obesity.
In a study conducted in 2019, comprising two groups, a normal weight group and an obese group, a negative correlation was observed between irisin levels and BMI, waist circumference, fasting glucose level, insulin, HOMA-IR, Homeostatic Model Assessment of cell function (HOMA-B2), and the estimation of β-cell function in diabetic patients in the obese group [56]. In a similar experiment, obese patients with a BMI of an average of 36.9 kg/m2 were placed under irisin intervention, which improved their fat percentage and significantly reduced fasting insulin levels and HOMA-IR [65]. In in vivo studies, irisin was initially shown to be effective in browning white adipose tissue (WAT) in mice and treating obesity [63,66]. Fourteen to sixteen-week-old C57BL/6J mice were experimented with to determine whether swimming affected serum irisin levels. The group subjected to swimming as an exercise showed an increase in the levels of PGC-1α and FNDC5, which are stimulators of irisin secretion. Browning markers, including UCP-1, also increased in the group that swam [67]. Overall, obesity itself and its consequences are modified by irisin supplementation.

5. Approaches for the Treatment and Management

Although various attempts have been made to develop new drugs, there are currently no approved pharmacological treatments for SO [6]. Therefore, SO is managed through lifestyle therapies such as exercise, diet, and nutritional supplements. Bariatric surgery, targeted drug therapy, and advanced exercise are being actively researched for the treatment of SO [7]. Approaches involving nutraceuticals and dietary supplements have also been emphasized. Nutraceuticals with anti-inflammatory properties and potential improvements in mitochondrial function have been proposed as emerging treatment options for SO. Additionally, active research on nutraceuticals, such as irisin, as well as new dietary approaches, has demonstrated positive effects on both obesity and muscle loss [81].

5.1. Lifestyle Intervention

SO presents a unique challenge as it involves the interplay between sarcopenia and obesity. The recommended approach for treating SO involves lifestyle interventions aimed at reducing weight and fat mass while increasing muscle mass and strength, which may include modifications to diet, physical activity, or specific exercises. Exercise is a highly effective approach for managing cardiovascular and respiratory diseases, diabetes, and various cancers and has been proposed as a preventive and therapeutic strategy for obesity [82,83]. It can improve muscle mass and strength in older adults with sarcopenia [84]. Although the optimal exercise regimen has yet to be determined, combining resistance training and aerobic exercise is recommended [67]. Resistance exercise is considered an effective therapeutic strategy for treating muscle atrophy because it promotes muscle hypertrophy and muscle protein synthesis, thereby enhancing muscle strength, quality, and physical performance [85]. Aerobic exercise can improve not only fiber size and strength but also overall muscle size and strength [86]. Additionally, it increases mitochondrial biogenesis and mitophagy, enhancing mitochondrial turnover and protecting against sarcopenia [87,88]. Physical exercise boosts mitochondrial activity to enhance the muscle-to-fat ratio, and customized exercise programs are recommended for older adults.
To optimize SO management, it is crucial to broaden the scope beyond exercise alone and consider the importance of nutrition and functional ingredients. An important aim of nutritional supplementation in patients with SO is to increase muscle mass while reducing body weight, particularly fat mass [89]. A hypocaloric diet is an integral approach for reducing energy intake and facilitating weight loss and is ideally supplemented with vitamin D and minerals, particularly calcium [7]. Vitamin D is a vital micronutrient for fat and muscle metabolism [90]. It is critical to proper fat storage in the WAT, and its deficiency leads to ectopic fat deposition in other tissues, resulting in inflammation and insulin resistance [91].
Calcitriol, the active form of vitamin D, negatively affects parathyroid hormone (PTH) synthesis. PTH synthesis is inhibited when circulating calcitriol levels increase. Because PTH weakens muscle strength, vitamin D prevents the muscle from losing function [92,93]. According to a short-term longitudinal study conducted in 2020, a 65% increase in vitamin D was concealed 60 times after the vitamin D receptor (VDR) gene was expressed. In the same study, participants showed a significant decrease in total body fat (p = 0.001) and an increase in gynoid lean mass (p = 0.007) (Table 3) [94]. Furthermore, VDR overexpression is associated with skeletal muscle hypertrophy. In addition to muscle hypertrophy and skeletal muscle repair, vitamin D is reported to repair mitochondrial function and balance its activities to prevent oxidative stress, lipid peroxidation, and DNA damage [91,95]. Protection against IL-6-induced inflammation is a clinical effect of vitamin D [96]. Briefly, vitamin D can enhance muscle activity by improving mitochondrial activity and actuating defensive actions against inflammation.
Vitamin K, a fat-soluble protein-producing vitamin, is required for blood clotting and bone structure [97]. Although there is conflict regarding whether vitamin K can treat fractures and bone mineral density (BMD), vitamin K has the potential to increase serum vitamin concentration and produce bone resorption markers [7]. Menaquinone-4 (MK-4) or menatetrenone, one of the nine forms of vitamin K2, improves mitochondrial function by boosting mitochondrial electron transfer, resulting in more efficient ATP production [98]. Among all cell types in the body, skeletal muscle cells have the most abundant mitochondria. Mitochondrial imbalance weakens muscle strength and promotes muscle degeneration, ultimately leading to sarcopenia [99]. In an in vitro study, the release of lactate dehydrogenase (LDH), an enzyme used as a marker of muscle damage and sarcopenia, was reduced in muscle cells treated with MK-4 [98]. In a 2017 cohort study, menaquinone-7 (MK-7) was administered to postmenopausal women aged 55–65 to verify whether MK-7 affects body weight. In cells that responded well to osteocalcin carboxylation, MK-7 treatment significantly increased adiponectin levels and decreased abdominal fat mass and visceral adipose tissue [100]. Thus, vitamin K can deteriorate the factors that cause muscle damage and simultaneously reduce fat accumulation.
Among the essential minerals that counteract sarcopenia, including magnesium, selenium, and calcium, calcium plays the largest role in preventing and treating sarcopenia [101]. Calcium is necessary to maintain calcium kinetics and promote muscle contraction [102]. Muscle contraction is responsible for joint bending, stability, and posture, which are likely to decline in adults. Because of its myogenic regulation, calcium maintenance is important for retaining the contradictory actions of muscles [103]. The fourth KNHANES, conducted in 2009, revealed a relationship between calcium intake and muscle mass. Muscle mass decreases correspondingly with lower calcium intake [104]. Moreover, calcium and vitamin D play protective roles against sarcopenia and loss of muscle mass. Calcium intake also increases BMD, which repairs osteoporosis and osteoarthritis, which are consequences of SO [105]. A boost in muscle strength and bone mass are major calcium-induced consequences in the treatment of skeletal damage and muscle repair.
SO exerts greater pressure on the lower limbs and can ultimately trigger knee osteoporosis [106]. However, testosterone can be used to manage these problems. Decreased muscle mass and function due to testosterone loss can be reversed by testosterone therapy [107]. Testosterone therapy applied to older adults showed decreased fat mass in one study and improved hamstring and quadriceps strength in another [99]. Testosterone levels are also highly correlated with muscle function. This is a major factor in allowing the body to adapt to training and resistance exercise [30]. One study showed a dose-dependent increase in leg press strength when testosterone levels increased in older adults [89]. In addition to the functional aspects, muscle composition is affected by testosterone secretion [108]. Testosterone is likely to be responsible for muscle growth [109]. Secreted testosterone facilitates protein synthesis, which leads to muscle growth and increased muscle mass [110]. However, whether testosterone can be supplied through dietary action remains questionable. Although testosterone is not a component of food, some foods promote the production and secretion of testosterone [111]. Multiple diet types were tested to determine whether they affected the testosterone concentration. The NHANES, conducted in 2020, was designed to determine whether a plant-based diet index was associated with serum testosterone levels. It was concluded that these two factors are not associated [112]. Conversely, in another study in 2020, the ketogenic diet (KD) and Western diet (WD) were compared to determine which had a greater effect on testosterone levels. In KD, total testosterone significantly increased by 118 ng/dL but decreased by 36 ng/dL in WD [113]. Through these diets, an increase in testosterone levels strengthens the joints and muscles of the lower body and reduces the possibility of osteoporosis.
Fatty acids are proficient in regulating metabolism and body functions [114]. Many organizations, such as the WHO and Food and Agriculture Organization, have recommended polyunsaturated fatty acids (PUFA), especially n-3 PUFA [115]. Multiple fatty acids have been recognized for their ability to modify muscle and fat mass in the human body. Docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and azelaic acid (AZA) are fatty acids that can be used to treat the distressing conditions of SO.
DHA, a ⍵-3 fatty acid commonly found in fish and milk, can delay skeletal muscle degradation related to obesity-induced inflammation [116]. When administered to C57BL/6J mice, DHA reduced inflammatory markers, and lipid accumulation was modified at the beginning of adipocytes [117]. The expression of genes related to mitochondrial biogenesis, PGC-1α, TFAM, and NRF-1, is also elevated in the presence of DHA in C2C12 muscle cells, proving DHA’s validity in both muscle genesis and lipid degradation [118]. DHA is adequate for generating proteins; however, it attenuates lipid accumulation by reducing inflammation.
EPA, a ⍵-3 fatty acid found in fish and nuts, can increase muscle function and reduce muscle damage [119]. When rats were treated with a 1 g/kg ratio, TNF-α and atrogin-1 levels were diminished, whereas myoblast determination protein (MyoD), a myogenic factor, increased [120]. In addition to boosting muscle, EPA is used to treat obesity. Mice fed a high-fat diet with EPA showed significantly reduced body weight, adiposity, adipocyte size, and macrophage infiltration into the adipose tissue. This group showed modifications in mitochondrial function [121]. Increased muscle and mitochondrial activity, along with improved adipocyte content, are outcomes that EPA is capable of bringing about.
By the same token, AZA, a ⍵-9 fatty acid mostly obtained from oatmeal and barley, can ameliorate muscle mass and fat capacity. During muscle hypertrophy, AZA promotes mitochondrial biogenesis and alleviates mitochondrial dysfunction. As a result, mitochondrial improvement reinforces muscle function and prevents muscle aging and contraction. Regarding obesity, AZA can attenuate fat mass as its consumption leads to a decline in lipid accumulation through the hydrolysis of triglycerides in adipose tissues. This disintegrates adipocytes, resulting in weight loss and body fat reduction [122]. Similar to the two fatty acids mentioned earlier, AZA can treat SO via mitochondrial biogenesis and adipose tissue collapse.
Table 3. The intervention of various nutrients on SO, muscle growth, and adipose tissue.
Table 3. The intervention of various nutrients on SO, muscle growth, and adipose tissue.
ReferenceIntervention
(Exposure, Dose, Duration)
ConditionSubjects
(Sample Size, Gender, Age)
Markers
Medeiros et al. [94]Cholecalciferol 2000 IU
60 days
Twins
Not on vitamin supplementation
CG
n = 45
Same sex
Age: 18–45
SG
n = 45
Same sex
Age: 18–45
Total body fat ↓
Gynoid lean mass ↑
VDR mRNA expression ↑
Labudzynskyi et al. [96]Vitamin D3 800 IU/kg
6 weeks
T1DMNegative CG mice
C56B1/J6
n = 8
21 ± 3 g
SG mice
C56B1/J6
n = 8
21 ± 3 g
IL-6 mRNA expression ↓
Skinner et al. [107]Testosterone replacement treatmentHypogonadismPlacebo
n = 1168
Age: ≥ 45
SG
n = 1213
Age: ≥ 45
Fat free mass ↑
Total body strength ↑
Wilson et al. [113]Ketogenic diet, western diet
10 weeks
-n = 25
Age: college-aged
Serum testosterone level ↑ in KD
Rønning et al. [98]MK-4 1 µM, 10 µM, 20 µM, 50 µM
6 days
Bovine primary skeletal muscle cells cultivated in 2% FBS with Ultroser G serumCG
n = 3000 cells/well
MK-4 10 µM
n = 3000 cells/well
Cell proliferation ↑
Gap closure ↑
Knapen et al. [100]MK-7 180 mcg/day
3 years
Postmenopausal womenSG
n = 107
Female
Age: 55–65
Placebo
n = 107
Female
Age: 55–65
Adiponectin ↑
Abdominal fat ↓
Visceral adipose tissue area ↓
Félix-Soriano et al. [117]DHA fish oil 15% HFD 3 times/day
12 months
Fed HFD for 4 monthsHFD mice
n = 10
Age: 6 months
HFD + DHA mice
n = 6
Age: 6 months
Visceral white adipose tissue ↓
Subcutaneous white adipose tissue ↓
Body weight ↓
TFAM ↑
Beige adipose tissue markers ↑
LeMieux et al. [121]EPA 10% HFD
11 weeks
-HFD mice
n = 8–10
HFD + EPA mice
n = 8–10
Body weight ↓
Adiposity ↓
Adipocyte size ↓
Osella et al. [66]Low glycemic index diet
6 months
Metabolic syndromeCG
n = 80
SG
n = 55
Serum irisin concentration ↑
Fat free mass ↑
Estell et al. [71]Irisin 2–20 ng/mL
4 h–7 days
OsteoblastsCGSGOsteoblasts/well ↑
Zafar et al. [69]Low glycemic index diet
≥1 week
Insulin resistance, T1DM, T2DMn = 54Glycated hemoglobin ↓
Fasting glucose ↓
BMI ↓
Total cholesterol ↓
LDL ↓

5.2. Emerging Therapy

5.2.1. Efficacy of Nutraceuticals

Polyphenols with anti-inflammatory and antioxidant properties have been proposed as potential nutraceuticals for treating sarcopenia by enhancing mitochondrial function [123]. Oligonol, a low-molecular-weight polyphenol derived from lychee, possesses anti-inflammatory and anti-obesity properties [124,125]. Recent studies have shown that oligonol regulates protein degradation related to mitochondrial function and improves skeletal muscle quality [126,127]. In a study conducted using an ovariectomized rat model, oligonol supplementation revealed the molecular mechanisms underlying its effects on body composition, protein turnover, and mitochondrial quality [128]. In this study, significant changes in body weight and accumulated fat were observed without any muscle loss. Oligonol treatment decreases sterol regulatory element-binding protein 1 expression. In another study, oligonol supplementation was shown to regulate body weight and improve serum lipid profiles in obese women [129]. Oligonol was found to increase AMPKα activity and enhance peroxisome proliferator-activator receptor α expression, thereby inhibiting lipid accumulation within the muscle [126]. Oligonol activates phospho-mTOR and its related pathways and stimulates the expression of FOXO and MuRF1, thereby increasing protein turnover. Oligonol also influences the expression of PGC-1α and NRF2, which are associated with mitochondrial biogenesis. Oligonol has the potential to serve as a treatment for SO as it inhibits lipid accumulation without muscle loss and regulates protein turnover and mitochondrial function [130].
Curcumin, also known as diferuloylmethane, is a major polyphenolic component that imparts a yellow color to turmeric [128]. The health benefits of turmeric have been attributed to curcumin. Curcumin possesses anticancer, antioxidant, anti-inflammatory, antidepressant, and anti-aging properties. Curcumin has been reported to have beneficial effects on neurological disorders, neuromuscular diseases, and osteoarthritis [131]. Several studies have revealed that impaired expression of nuclear factor erythroid-2 related factor-2 (NFE2LE) during aging is associated with oxidative stress and muscle degeneration [132]. Curcumin also activates NFE2LE [133]. It has been demonstrated that curcumin increases the expression and activation of NFE2LE by promoting the release of NFE2LE from kelch-like ECH-associated protein 1 [134]. Upregulation of NFE2LE in the skeletal muscle is induced by curcumin supplementation [135]. In one experiment, long-term administration of curcumin was conducted in rats, followed by evaluation of muscle mass and function. When 32-month-old male F344xBN rats were fed a diet containing 0.2% curcumin for 4 months, they exhibited greater plantaris mass and increased force production compared to the control group. Additionally, rats that consume curcumin have higher nuclear fraction levels of NFE2LE and lower oxidative macromolecular damage [136]. In summary, curcumin has the potential to be a therapeutic agent for SO owing to its anti-aging effects, such as antioxidant and anti-inflammatory properties, and its ability to activate NFE2L2.
Together, green tea extract (GTE) and other substances can treat sarcopenia and obesity. GTE facilitates muscle function and induces weight loss. GTE not only suppresses muscle loss and improves muscle function but can also increase satellite cell proliferation and stimulate differentiation of the plantaris and soleus muscles after hindlimb suspension [137]. Similarly, 15-day supplementation with 500 mg GTE daily reduced muscle damage markers after exercise, leading to muscle recovery [138]. In addition to enhancing muscle structure, GTE plays an important role in weight management. After endurance training, a group supplemented with GTE exhibited increased serum irisin concentrations and significant reductions in body weight, BMI, body fat percentage, and visceral fat [139]. Moreover, when GTE was administered to obese women with a BMI > 27 kg/m2 and a weight circumference of 80 cm or above for 12 weeks, a consistent decrease in total cholesterol and LDL plasma levels was observed with no side effects. These results suggest that they may be associated with the inhibition of ghrelin secretion, leading to increased adiponectin levels [140]. Ultimately, increased serum irisin levels and muscle mass and decreased markers of obesity were observed after supplementation with GTE.
Epigallocatechin gallate (EGCG), commonly found in green tea, is known for its ability to translocate GLUT4 and reinforce muscle function. Not only does GLUT4 translocation fix IR, but it also boosts glycogen storage in muscles, sustaining muscle performance [141]. ECGC also acts as an antioxidant by reversing the increase in reactive oxygen species. Such antioxidant actions, GLUT4 translocation, and activation of the AMPK pathway regulate glucose metabolism in skeletal muscles and diminish fatty acid synthesis, reducing body weight during muscle development [142]. In an experiment supplementing sarcopenic rats with 200 mg/kg body weight ECGC, the expression of anabolic factors, such as IGF-1, increased significantly, and the muscle was preserved [143]. Myogenin and MyoD levels were increased, whereas myostatin levels declined when aged mice were supplemented with ECGC. Moreover, the RNA expression of mitochondrial metabolism-related molecules, such as mitochondrial cytochrome b and mitochondrial cytochrome c oxidase II, III, and IV, increased when old mice were treated with ECGC along with treadmill runs for 10 weeks [144]. Catechin and epicatechin are flavonols found in green tea extract [145]. These two myostatin inhibitors have pharmacotherapeutic effects against SO [146]. When two groups of males were treated with epicatechin or a placebo after resistance exercise for 8 weeks, the group supplemented with epicatechin showed significantly greater myostatin inhibition than the placebo group (p ≤ 0.05) [147]. Overall, EGCG, catechin, and epicatechin were the bioactive compounds in GTE that demonstrated a superior effect in the treatment of SO.
Green cardamom, a perennial plant in the ginger family native to southern India, has attracted attention as a potential treatment for SO. Green cardamom has therapeutic effects on MetS, including diabetes, hyperlipidemia, obesity, and high blood pressure [148]. Green cardamom improves several blood factors such as lipids, inflammatory markers, liver enzymes, and irisin (Table 4) [149]. This study was conducted at the National Iranian Oil Company Central Hospital in Tehran to test the effect of green cardamom on irisin secretion. Subjects supplemented with three capsules of 500 mg green cardamom daily for 3 months showed a significant increase in serum irisin, HDLc, quantitative insulin sensitivity check index, and decreased fasting blood insulin level, triglycerides, LDLc, HOMA-IR, and fatty liver grade (p < 0.05) [150]. Green cardamom can prevent MetS-related obesity and enhance irisin production, thereby potentially treating SO.
A-terpineol (TPN), an alcoholic compound found in essential oils extracted from green cardamom, reduces inflammation and insulin resistance [151]. Mice treated with different concentrations of TPN (25, 50, and 100 mg/kg) showed decreased inflammatory responses and diseases caused by TNF-α and prostaglandin E₂ [152]. In another study, mice fed a high-fat diet showed counteractions against insulin resistance when treated with 50 mg of TPN. The same study evaluated whether TPN can potentially reduce proinflammatory cytokines, such as IL-6 and TNF-α, that lead to obesity [153]. Linalool, another compound in green cardamom, reduces hyperalgesia and non-inflammatory muscle pain [154]. Limonene, a cyclic monoterpene found in green cardamom, stimulates osteoblast differentiation and nullifies the effects of p38 inhibitors. P38 is a type of MAPK responsible for osteoblast differentiation and muscle regulation; thus, when inhibited, it prevents muscle recovery [155]. Limonene also acts as an antioxidant, reducing lipid peroxidation and superoxide dismutase levels after muscle injury [156]. In summary, TPN and limonene, the major nutraceuticals in green cardamom, inhibit inflammatory cytokines by treating obesity and facilitating muscle recovery.
Garlic is a commonly used spice well known as an effective antioxidant that simultaneously diminishes bacteria and parasites and lowers blood pressure and cholesterol levels [157]. In a study in which subjects were administered two tablets consisting of 400 mg of garlic powder, a significant decrease in body weight and fat mass was observed compared with the group that received placebo tablets (p < 0.05) [158]. In another study, subjects with NAFLD were supplemented with four tablets of 400 mg garlic powder and compared with the control group. A significant reduction in waist circumference (p = 0.001), body fat percentage (p < 0.001), fasting glucose level (p = 0.01), and insulin resistance (p < 0.001) were observed, along with a significant increase in skeletal muscle mass (p = 0.002) [159]. Garlic has the ability not only to increase muscle mass but also to lower blood pressure, cholesterol levels, and obesity.
To explore the compounds in garlic that contribute to the therapeutic effects of SO, s-allyl cysteine (SAC), a compound regularly found in garlic, has been highlighted as a potential defender against muscle atrophy [160]. C2C12 myotubes were exposed to 100 µM H₂O₂ and supplemented with 400 µM SAC to examine whether SAC exerted an anti-atrophic effect. SAC reduces myostatin secretion and mediates the degradation of muscle-specific proteins, proving that it is an effective anti-atrophic compound [161]. In another experiment, C2C12 was treated with 100 ng/mL TNF-α. Groups present in SAC and without SAC were compared to detect that the group supplemented with SAC restricted TNF-α-induced proteolysis and protected myotubes against inflammatory molecules, such as IL-6 and TNF-like weak inducers of apoptosis [162]. SAC protects garlic against muscle atrophy by inhibiting myostatin production and diminishing inflammatory responses.
Table 4. Studies of potential nutraceuticals and their results related to SO.
Table 4. Studies of potential nutraceuticals and their results related to SO.
ReferenceSubstance (Compound)Intervention
(Exposure, Dose, Duration)
ConditionSubjects
(Sample Size, Gender, Mean Age)
Markers
Daneshi-Maskooni et al. [149]Green Cardamom2 cardamom 500 mg capsules 3 times/day
3 months
Overweight or obese, NAFLDPlacebo
n = 44
Age: 30–60
Supplement group
n = 43
Age: 30–60
TNF-α ↓
IL-6 ↓
ALT ↓
Degree of fatty liver ↓
Daneshi-Maskooni et al. [150]2 cardamom 500 mg capsules 3 times/day
3 months
Overweight or obese, NAFLDPlacebo
n = 44
Age: 30–60
Supplement group
n = 43
Age: 30–60
Irisin ↑
HDL-c ↑
TG ↓
LDL-c ↓
HOMA-IR ↓
Degree of fatty liver ↓
Oliveira et al. [152]Green Cardamomα-terpineolα-terpineol 25, 50, 100 mg/kg
180 min
Hypernoinception induced by carrageenanMale Swiss mice
n = 6 per group
Age: 2–3 months
Mass: 28–32 g
TNF-α ↓
Prostaglandin E₂ ↓
Sousa et al. [153]A-terpineol 25, 50, 100 mg/kg
6 weeks
Obese, high-fat and hypercaloric dietSqrague-Dawley rats
n = 6 per group
Age: 21 days
Mass: ≃150 g
TNF-α ↓
IL-1β ↓
Weight gain ↓
Degree of fatty liver ↓
Nascimento et al. [154]LinaloolLinalool 25 mg/kg
Alternate days for 27 days
Hyperalgesia,
injected with 20 μL pH 4 acidic saline
Male Swiss mice
Mass: 25–30 g
Non-inflammatory pain ↓
Soundharrajan et al.
[155]
LimoneneLimonene 2.5, 5, 10 μM
6 days
Seeded 5 × 10⁴ cells/well,
10% FBS in DMEM,
37 °C, 5% CO₂
C2C12 skeletal cell
80–90% confluency
Calcium deposition ↑
Myogenin ↑
MyoD ↑
p38 MAPK signalling pathway ↑
Santos et al. [156]Limonene 5%
96 h
Gastrocnemius muscle injured by 0.459 kg metal bar press with 0.811 JMale Wistar ratsThiobarbituric acid reactive substances ↓
Superoxide dimutase ↓
Muslce injured group
n = 6
Mass: 250–280 g
Supplemented group
n = 6
Mass: 250–280 g
Silva et al. [138]Green TeaGTE 500 mg/day
15 days
Exerecise-induced muscle sorenessPlacebo
n = 10
Supplemented group
n = 10
Muscle recovery ↑
Bagheri et al. [139]GTE 500 mg/day
8 weeks
Under endurance training:
Circuit training, fast walking, jogging 3 times/week
Placebo
n = 15
Supplemented group
n = 15
IL-6 ↓
Adiponectin ↑
Irisin ↑
Body weight ↓
BMI ↓
Body fat percentage ↓
Visceral fat area ↓
Chen et al. [140]Green TeaEGCGEGCG 856.8 mg/day
12 weeks
BMI ≥ 27 kg/m2, waist circumference ≥ 80 cmPlacebo
n = 39
Age: 20–60
Supplemented group
n = 38
Age: 20–60
Body weight ↓
BMI ↓
Waist circumference ↓
TC ↓
LDL-c ↓
Ghrelin ↓
Adiponectin ↑
Meador et al. [143]EGCG 200 mg/kg
8 weeks
SarcopeniaSprague-Dawley ratsIGF-1 ↑
Control group
Age: 20 months
Supplemented group
Age: 20 months
Mafi et al. [147]EpicatechinEpicatechin along with resistance trainingSarcopeniaPlacebo
n = 15
Age: 68.63 ± 2.86
Supplemented group
n = 15
Age: 68.63 ± 2.86
Myostain ↓
Soleimani et al. [158]Garlic2 garlic powder 400 mg tablets/day
15 weeks
NAFLDPlacebo
n = 45
Age: 20–79
Supplemented group
n = 45
Age: 20–70
Body weight ↓
Body fat mass ↓
Sangouni et al. [159]Garlic4 garlic powder 400 mg tablets/day
12 weeks
NAFLDControl group
n = 43
Age: ≥ 18
Supplemented group
n = 45
Age: ≥ 18
Waist circumference ↓
Body fat percent ↓
Fasting glucose level ↓
Insulin resistance ↓
Skeleta muscle mass ↑
Gupta et al. [161]GarlicSACSAC 200 μM
48 h
Atrophic effect by H₂O₂
DMEM, FBS, 2% horse serum,
1 μg/mL ciprofloxacin, 1.25 μg/mL amphotericin B
C2C12 muscle cell
80–90% confluency
TWEAK ↓
IL-6 ↓
Myostatin ↓
Muscle denervation ↓
Dutt et al. [162]SAC 0.01 mM
72 h
Treated with TNF-α 100 ng/mL
Seeded 2 × 10⁶ cells/well,
DMEM with 20% FBS,
5 μg/mL ciprofloxacin, 2.5 μg/mL amphotericin B,
37 °C, 5% CO₂
C2C12 muscle cell
70–80% confluency
TNF-α ↓
IL-6 ↓
IL-1β ↓
TWEAK ↓
Chang et al. [127]Oligonol200 mg/kg oligonol
8 weeks
Senescence-accelerated SAMP8 Micephosphorylation of AKT/mTOR/p70sk6 ↑
MuRF-1/MAFbx ↓
PGC-1α/Tfam ↓
Mfn2/Opa1 ↓
cytochrome c ↓
Receno et al. [136]Curcumin0.2% curcumin
4 months
AgedF344xBN rats
ad libitum control (CON; n = 18)
0.2% curcumin (CUR; n = 18)
pair-fed (PAIR; n = 18)
Age: 32 months
Food intake ↓
plantaris mass ↑
force production ↑
nuclear fraction levels of Nrf2 ↑
oxidative macromolecule damage ↓

5.2.2. Dietary Interventions to Modulate Irisin Secretion

In addition to lifestyle interventions, multiple dietary methods can also enhance irisin production. Downregulation of FNDC5 was observed when mice were fed a high-fat diet, whereas a high-carbohydrate and high-protein diet showed the opposite result. A high-protein diet maintained FNDC5 and irisin levels and increased brown adipose tissue in mice (Table 2) [163]. Although controversial, some studies have reported that meat consumption is negatively correlated with blood irisin levels [68]. However, increased intake of vegetables, cheese, and processed meat effectively increases irisin secretion [66].
The most common form of diet that manages SO is a low-glycemic-index diet (LGID). Several studies have been conducted to determine whether LGID effectively improves irisin levels. In one study of subjects with MetS, the LGID was compared with the Mediterranean diet (MD) and low-glycemic index Mediterranean diet (LGIMD). Subjects with LGID showed higher irisin levels than those with MD and LGIMD [66]. In another study, LGID led to weight loss; however, a greater reduction was observed in those with normal blood glucose levels than in those with higher blood glucose levels. Body fat was significantly reduced in those with normal glucose tolerance, but those with impaired glucose tolerance, or T2DM, did not show a decrease. In addition, LGID lowers fasting glucose and glycated hemoglobin levels. LGID was also able to reduce BMI, total cholesterol (TC), and low-density lipoprotein cholesterol (LDLc), but did not enhance fasting insulin or high-density lipoprotein cholesterol (HDLc) [69,164]. In addition to a high-protein diet and increased vegetable intake, LGID mice showed increased irisin secretion and reduced levels of obesity markers such as fasting glucose and cholesterol.

6. Conclusions

Aging populations have emerged as a social issue in many countries, increasing the prevalence of sarcopenia due to the age-related decline in muscle mass and strength. Additionally, there is a simultaneous increase in the prevalence of SO when sarcopenia coexists with obesity. SO can have adverse consequences such as physical disability, poor quality of life, metabolic syndrome, and mortality, negatively affecting individuals and society. Although the definition and pathology of SO have continued to develop and be suggested, more research is essential to obtain a clear pathology and a consensus definition. Various treatments have been suggested for SO. The most important factors are increased skeletal muscle mass and decreased body fat mass. Therefore, exercise has been proposed as a fundamental treatment. However, implementing an optimal diet is also important for maximizing recovery from SO. As mentioned in this paper, several dietary methods can have positive effects, such as calcium supplementation, vitamins D and K, a ketogenic diet, and a high-protein diet. Moreover, irisin, a myokine released during exercise or under specific stressful conditions, is competent in ameliorating muscle and fat content while canceling the secretion of myostatin, a myokine that reverses irisin. Nutraceuticals such as green cardamom, which can inhibit myostatin secretion, promote irisin secretion, or treat obesity, may also be a possible aid in managing SO. Although the treatment of SO through dietary effects and exercise remains questionable, attempts to treat it using nutraceuticals offer a potential therapeutic approach by targeting various mechanisms of action.

Author Contributions

Conceptualization and G.-w.G.; methodology, G.-w.G.; formal analysis, Y.-C.K. and S.-W.K.; investigation, Y.-C.K. and S.-W.K.; data curation, Y.-C.K. and S.-W.K.; writing—original draft preparation, Y.-C.K. and S.-W.K.; writing—review and editing, H.K. (Hannah Kim), S.K., H.K. (Hayoon Kim) and G.-w.G.; visualization, Y.-C.K. and S.-W.K.; supervision, G.-w.G.; project administration, G.-w.G.; funding acquisition, G.-w.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (Ministry of Science and ICT). (No. 2022R1A2C1006193).

Data Availability Statement

The data presented in this study are available within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jura, M.; Kozak, L.P. Obesity and related consequences to ageing. Age 2016, 38, 23. [Google Scholar] [CrossRef] [PubMed]
  2. Petermann-Rocha, F.; Balntzi, V.; Gray, S.R.; Lara, J.; Ho, F.K.; Pell, J.P.; Celis-Morales, C. Global prevalence of sarcopenia and severe sarcopenia: A systematic review and meta-analysis. J. Cachexia Sarcopenia Muscle 2022, 13, 86–99. [Google Scholar] [CrossRef] [PubMed]
  3. Boutari, C.; Mantzoros, C.S. A 2022 update on the epidemiology of obesity and a call to action: As its twin COVID-19 pandemic appears to be receding, the obesity and dysmetabolism pandemic continues to rage on. Metabolism 2022, 133, 155217. [Google Scholar] [CrossRef]
  4. Silveira, E.A.; da Silva Filho, R.R.; Spexoto, M.C.B.; Haghighatdoost, F.; Sarrafzadegan, N.; de Oliveira, C. The Role of Sarcopenic Obesity in Cancer and Cardiovascular Disease: A Synthesis of the Evidence on Pathophysiological Aspects and Clinical Implications. Int. J. Mol. Sci. 2021, 22, 4339. [Google Scholar] [CrossRef] [PubMed]
  5. Gao, Q.; Mei, F.; Shang, Y.; Hu, K.; Chen, F.; Zhao, L.; Ma, B. Global prevalence of sarcopenic obesity in older adults: A systematic review and meta-analysis. Clin. Nutr. 2021, 40, 4633–4641. [Google Scholar] [CrossRef]
  6. Polyzos, S.A.; Margioris, A.N. Sarcopenic obesity. Hormones 2018, 17, 321–331. [Google Scholar] [CrossRef] [PubMed]
  7. Batsis, J.A.; Villareal, D.T. Sarcopenic obesity in older adults: Aetiology, epidemiology and treatment strategies. Nat. Rev. Endocrinol. 2018, 14, 513–537. [Google Scholar] [CrossRef]
  8. Baumgartner, R.N.; Koehler, K.M.; Gallagher, D.; Romero, L.; Heymsfield, S.B.; Ross, R.R.; Garry, P.J.; Lindeman, R.D. Epidemiology of sarcopenia among the elderly in New Mexico. Am. J. Epidemiol. 1998, 147, 755–763. [Google Scholar] [CrossRef]
  9. Janssen, I.; Heymsfield, S.B.; Ross, R. Low relative skeletal muscle mass (sarcopenia) in older persons is associated with functional impairment and physical disability. J. Am. Geriatr. Soc. 2002, 50, 889–896. [Google Scholar] [CrossRef]
  10. Newman, A.B.; Kupelian, V.; Visser, M.; Simonsick, E.; Goodpaster, B.; Nevitt, M.; Kritchevsky, S.B.; Tylavsky, F.A.; Rubin, S.M.; Harris, T.B. Sarcopenia: Alternative definitions and associations with lower extremity function. J. Am. Geriatr. Soc. 2003, 51, 1602–1609. [Google Scholar] [CrossRef]
  11. Cruz-Jentoft, A.J.; Baeyens, J.P.; Bauer, J.M.; Boirie, Y.; Cederholm, T.; Landi, F.; Martin, F.C.; Michel, J.P.; Rolland, Y.; Schneider, S.M.; et al. Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing 2010, 39, 412–423. [Google Scholar] [CrossRef] [PubMed]
  12. Fielding, R.A.; Vellas, B.; Evans, W.J.; Bhasin, S.; Morley, J.E.; Newman, A.B.; Abellan van Kan, G.; Andrieu, S.; Bauer, J.; Breuille, D.; et al. Sarcopenia: An undiagnosed condition in older adults. Current consensus definition: Prevalence, etiology, and consequences. International working group on sarcopenia. J. Am. Med. Dir. Assoc. 2011, 12, 249–256. [Google Scholar] [CrossRef] [PubMed]
  13. Studenski, S.A.; Peters, K.W.; Alley, D.E.; Cawthon, P.M.; McLean, R.R.; Harris, T.B.; Ferrucci, L.; Guralnik, J.M.; Fragala, M.S.; Kenny, A.M.; et al. The FNIH sarcopenia project: Rationale, study description, conference recommendations, and final estimates. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69, 547–558. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, L.K.; Liu, L.K.; Woo, J.; Assantachai, P.; Auyeung, T.W.; Bahyah, K.S.; Chou, M.Y.; Chen, L.Y.; Hsu, P.S.; Krairit, O.; et al. Sarcopenia in Asia: Consensus report of the Asian Working Group for Sarcopenia. J. Am. Med. Dir. Assoc. 2014, 15, 95–101. [Google Scholar] [CrossRef]
  15. Cruz-Jentoft, A.J.; Bahat, G.; Bauer, J.; Boirie, Y.; Bruyère, O.; Cederholm, T.; Cooper, C.; Landi, F.; Rolland, Y.; Sayer, A.A.; et al. Sarcopenia: Revised European consensus on definition and diagnosis. Age Ageing 2019, 48, 16–31. [Google Scholar] [CrossRef]
  16. World Health Organization Regional Office for the Western Pacific; Regional Office for the Western Pacific. The Asia-Pacific Perspective: Redefining Obesity and Its Treatment; Health Communications Australia: Sydney, Australia, 2000. [Google Scholar]
  17. Després, J.P. Body fat distribution and risk of cardiovascular disease: An update. Circulation 2012, 126, 1301–1313. [Google Scholar] [CrossRef]
  18. Garvey, W.T.; Mechanick, J.I.; Brett, E.M.; Garber, A.J.; Hurley, D.L.; Jastreboff, A.M.; Nadolsky, K.; Pessah-Pollack, R.; Plodkowski, R. American Association of Clinical Endocrinologists and American College of Endocrinology Comprehensive Clinical Practice Guidelines for Medical Care of Patients with Obesity. Endocr. Pract. 2016, 22 (Suppl. S3), 1–203. [Google Scholar] [CrossRef]
  19. Cleasby, M.E.; Jamieson, P.M.; Atherton, P.J. Insulin resistance and sarcopenia: Mechanistic links between common co-morbidities. J. Endocrinol. 2016, 229, R67–R81. [Google Scholar] [CrossRef]
  20. Stenholm, S.; Harris, T.B.; Rantanen, T.; Visser, M.; Kritchevsky, S.B.; Ferrucci, L. Sarcopenic obesity: Definition, cause and consequences. Curr. Opin. Clin. Nutr. Metab. Care 2008, 11, 693–700. [Google Scholar] [CrossRef]
  21. Kim, T.N.; Choi, K.M. Sarcopenia: Definition, epidemiology, and pathophysiology. J. Bone Metab. 2013, 20, 1–10. [Google Scholar] [CrossRef]
  22. de Luca, C.; Olefsky, J.M. Inflammation and insulin resistance. FEBS Lett. 2008, 582, 97–105. [Google Scholar] [CrossRef] [PubMed]
  23. Matsubara, M.; Maruoka, S.; Katayose, S. Inverse relationship between plasma adiponectin and leptin concentrations in normal-weight and obese women. Eur. J. Endocrinol. 2002, 147, 173–180. [Google Scholar] [CrossRef] [PubMed]
  24. Ceglia, L. Vitamin D and its role in skeletal muscle. Curr. Opin. Clin. Nutr. Metab. Care 2009, 12, 628–633. [Google Scholar] [CrossRef]
  25. Maalouf, G.E.; El Khoury, D. Exercise-Induced Irisin, the Fat Browning Myokine, as a Potential Anticancer Agent. J. Obes. 2019, 2019, 6561726. [Google Scholar] [CrossRef]
  26. Machida, S.; Booth, F.W. Insulin-like growth factor 1 and muscle growth: Implication for satellite cell proliferation. Proc. Nutr. Soc. 2004, 63, 337–340. [Google Scholar] [CrossRef]
  27. Aguirre, G.A.; De Ita, J.R.; de la Garza, R.G.; Castilla-Cortazar, I. Insulin-like growth factor-1 deficiency and metabolic syndrome. J. Transl. Med. 2016, 14, 3. [Google Scholar] [CrossRef] [PubMed]
  28. Maltais, M.L.; Desroches, J.; Dionne, I.J. Changes in muscle mass and strength after menopause. J. Musculoskelet. Neuronal Interact. 2009, 9, 186–197. [Google Scholar]
  29. McCarthy, M.; Raval, A.P. The peri-menopause in a woman’s life: A systemic inflammatory phase that enables later neurodegenerative disease. J. Neuroinflamm. 2020, 17, 317. [Google Scholar] [CrossRef]
  30. Vingren, J.L.; Kraemer, W.J.; Ratamess, N.A.; Anderson, J.M.; Volek, J.S.; Maresh, C.M. Testosterone physiology in resistance exercise and training: The up-stream regulatory elements. Sports Med. 2010, 40, 1037–1053. [Google Scholar] [CrossRef]
  31. Trendelenburg, A.U.; Meyer, A.; Rohner, D.; Boyle, J.; Hatakeyama, S.; Glass, D.J. Myostatin reduces Akt/TORC1/p70S6K signaling, inhibiting myoblast differentiation and myotube size. Am. J. Physiol. Cell Physiol. 2009, 296, C1258–C1270. [Google Scholar] [CrossRef]
  32. Hitachi, K.; Nakatani, M.; Tsuchida, K. Myostatin signaling regulates Akt activity via the regulation of miR-486 expression. Int. J. Biochem. Cell Biol. 2014, 47, 93–103. [Google Scholar] [CrossRef] [PubMed]
  33. Amor, M.; Itariu, B.K.; Moreno-Viedma, V.; Keindl, M.; Jurets, A.; Prager, G.; Langer, F.; Grablowitz, V.; Zeyda, M.; Stulnig, T.M. Serum Myostatin is Upregulated in Obesity and Correlates with Insulin Resistance in Humans. Exp. Clin. Endocrinol. Diabetes 2019, 127, 550–556. [Google Scholar] [CrossRef] [PubMed]
  34. Safdar, A.; Little, J.P.; Stokl, A.J.; Hettinga, B.P.; Akhtar, M.; Tarnopolsky, M.A. Exercise increases mitochondrial PGC-1alpha content and promotes nuclear-mitochondrial cross-talk to coordinate mitochondrial biogenesis. J. Biol. Chem. 2011, 286, 10605–10617. [Google Scholar] [CrossRef]
  35. Seo, D.Y.; Kwak, H.B.; Lee, S.R.; Cho, Y.S.; Song, I.S.; Kim, N.; Bang, H.S.; Rhee, B.D.; Ko, K.S.; Park, B.J.; et al. Effects of aged garlic extract and endurance exercise on skeletal muscle FNDC-5 and circulating irisin in high-fat-diet rat models. Nutr. Res. Pract. 2014, 8, 177–182. [Google Scholar] [CrossRef]
  36. Jandova, T.; Buendia-Romero, A.; Polanska, H.; Hola, V.; Rihova, M.; Vetrovsky, T.; Courel-Ibanez, J.; Steffl, M. Long-Term Effect of Exercise on Irisin Blood Levels-Systematic Review and Meta-Analysis. Healthcare 2021, 9, 1438. [Google Scholar] [CrossRef] [PubMed]
  37. Garcia-Fontana, B.; Reyes-Garcia, R.; Morales-Santana, S.; Avila-Rubio, V.; Munoz-Garach, A.; Rozas-Moreno, P.; Munoz-Torres, M. Relationship between myostatin and irisin in type 2 diabetes mellitus: A compensatory mechanism to an unfavourable metabolic state? Endocrine 2016, 52, 54–62. [Google Scholar] [CrossRef] [PubMed]
  38. Cauley, J.A. Estrogen and bone health in men and women. Steroids 2015, 99, 11–15. [Google Scholar] [CrossRef] [PubMed]
  39. Szulc, P.; Beck, T.J.; Marchand, F.; Delmas, P.D. Low skeletal muscle mass is associated with poor structural parameters of bone and impaired balance in elderly men—The MINOS study. J. Bone Miner. Res. 2005, 20, 721–729. [Google Scholar] [CrossRef] [PubMed]
  40. Scott, D.; Daly, R.M.; Sanders, K.M.; Ebeling, P.R. Fall and Fracture Risk in Sarcopenia and Dynapenia with and without Obesity: The Role of Lifestyle Interventions. Curr. Osteoporos. Rep. 2015, 13, 235–244. [Google Scholar] [CrossRef] [PubMed]
  41. Liu, L.; Feng, J.; Zhang, G.; Yuan, X.; Li, F.; Yang, T.; Hao, S.; Huang, D.; Hsue, C.; Lou, Q. Visceral adipose tissue is more strongly associated with insulin resistance than subcutaneous adipose tissue in Chinese subjects with pre-diabetes. Curr. Med. Res. Opin. 2018, 34, 123–129. [Google Scholar] [CrossRef] [PubMed]
  42. Fontana, L.; Eagon, J.C.; Trujillo, M.E.; Scherer, P.E.; Klein, S. Visceral fat adipokine secretion is associated with systemic inflammation in obese humans. Diabetes 2007, 56, 1010–1013. [Google Scholar] [CrossRef] [PubMed]
  43. Katz, J.N.; Arant, K.R.; Loeser, R.F. Diagnosis and Treatment of Hip and Knee Osteoarthritis: A Review. JAMA 2021, 325, 568–578. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, H.; Wang, N.; Wang, Y.; Li, H. Association between sarcopenia and osteoarthritis: A protocol for meta-analysis. PLoS ONE 2022, 17, e0272284. [Google Scholar] [CrossRef]
  45. Pegreffi, F.; Balestra, A.; De Lucia, O.; Smith, L.; Barbagallo, M.; Veronese, N. Prevalence of Sarcopenia in Knee Osteoarthritis: A Systematic Review and Meta-Analysis. J. Clin. Med. 2023, 12, 1532. [Google Scholar] [CrossRef] [PubMed]
  46. Pickering, M.E.; Chapurlat, R. Where Two Common Conditions of Aging Meet: Osteoarthritis and Sarcopenia. Calcif. Tissue Int. 2020, 107, 203–211. [Google Scholar] [CrossRef] [PubMed]
  47. Khosla, S.; Monroe, D.G. Regulation of Bone Metabolism by Sex Steroids. Cold Spring Harb. Perspect. Med. 2018, 8, a031211. [Google Scholar] [CrossRef] [PubMed]
  48. Vasarhelyi, E.M.; MacDonald, S.J. The influence of obesity on total joint arthroplasty. J. Bone Jt. Surg. Br. 2012, 94, 100–102. [Google Scholar] [CrossRef] [PubMed]
  49. Tian, S.; Xu, Y. Association of sarcopenic obesity with the risk of all-cause mortality: A meta-analysis of prospective cohort studies. Geriatr. Gerontol. Int. 2016, 16, 155–166. [Google Scholar] [CrossRef] [PubMed]
  50. Barazzoni, R.; Bischoff, S.C.; Boirie, Y.; Busetto, L.; Cederholm, T.; Dicker, D.; Toplak, H.; Van Gossum, A.; Yumuk, V.; Vettor, R. Sarcopenic obesity: Time to meet the challenge. Clin. Nutr. 2018, 37, 1787–1793. [Google Scholar] [CrossRef]
  51. Flegal, K.M.; Kit, B.K.; Orpana, H.; Graubard, B.I. Association of all-cause mortality with overweight and obesity using standard body mass index categories: A systematic review and meta-analysis. JAMA 2013, 309, 71–82. [Google Scholar] [CrossRef] [PubMed]
  52. Lim, S.; Kim, J.H.; Yoon, J.W.; Kang, S.M.; Choi, S.H.; Park, Y.J.; Kim, K.W.; Lim, J.Y.; Park, K.S.; Jang, H.C. Sarcopenic obesity: Prevalence and association with metabolic syndrome in the Korean Longitudinal Study on Health and Aging (KLoSHA). Diabetes Care 2010, 33, 1652–1654. [Google Scholar] [CrossRef]
  53. Baek, S.J.; Nam, G.E.; Han, K.D.; Choi, S.W.; Jung, S.W.; Bok, A.R.; Kim, Y.H.; Lee, K.S.; Han, B.D.; Kim, D.H. Sarcopenia and sarcopenic obesity and their association with dyslipidemia in Korean elderly men: The 2008–2010 Korea National Health and Nutrition Examination Survey. J. Endocrinol. Investig. 2014, 37, 247–260. [Google Scholar] [CrossRef] [PubMed]
  54. Kim, T.N.; Park, M.S.; Lim, K.I.; Choi, H.Y.; Yang, S.J.; Yoo, H.J.; Kang, H.J.; Song, W.; Choi, H.; Baik, S.H.; et al. Relationships between sarcopenic obesity and insulin resistance, inflammation, and vitamin D status: The Korean Sarcopenic Obesity Study. Clin. Endocrinol. 2013, 78, 525–532. [Google Scholar] [CrossRef]
  55. Waseem, R.; Shamsi, A.; Mohammad, T.; Hassan, M.I.; Kazim, S.N.; Chaudhary, A.A.; Rudayni, H.A.; Al-Zharani, M.; Ahmad, F.; Islam, A. FNDC5/Irisin: Physiology and Pathophysiology. Molecules 2022, 27, 1118. [Google Scholar] [CrossRef]
  56. Rashid, F.A.; Abbas, H.J.; Naser, N.A.; Addai Ali, H. Effect of Long-Term Moderate Physical Exercise on Irisin between Normal Weight and Obese Men. Sci. World J. 2020, 2020, 1897027. [Google Scholar] [CrossRef]
  57. Ulupinar, S.; Ozbay, S.; Gencoglu, C.; Altinkaynak, K.; Sebin, E.; Oymak, B. Exercise in the cold causes greater irisin release but may not be enough for adropin. Chin. J. Physiol. 2021, 64, 129–134. [Google Scholar] [CrossRef] [PubMed]
  58. Jornayvaz, F.R.; Shulman, G.I. Regulation of mitochondrial biogenesis. Essays Biochem. 2010, 47, 69–84. [Google Scholar] [CrossRef] [PubMed]
  59. Vaughan, R.A.; Gannon, N.P.; Barberena, M.A.; Garcia-Smith, R.; Bisoffi, M.; Mermier, C.M.; Conn, C.A.; Trujillo, K.A. Characterization of the metabolic effects of irisin on skeletal muscle in vitro. Diabetes Obes. Metab. 2014, 16, 711–718. [Google Scholar] [CrossRef]
  60. Norheim, F.; Langleite, T.M.; Hjorth, M.; Holen, T.; Kielland, A.; Stadheim, H.K.; Gulseth, H.L.; Birkeland, K.I.; Jensen, J.; Drevon, C.A. The effects of acute and chronic exercise on PGC-1alpha, irisin and browning of subcutaneous adipose tissue in humans. FEBS J. 2014, 281, 739–749. [Google Scholar] [CrossRef] [PubMed]
  61. Tsuchiya, Y.; Ando, D.; Goto, K.; Kiuchi, M.; Yamakita, M.; Koyama, K. High-intensity exercise causes greater irisin response compared with low-intensity exercise under similar energy consumption. Tohoku J. Exp. Med. 2014, 233, 135–140. [Google Scholar] [CrossRef] [PubMed]
  62. Lavi, G.; Horwitz, A.; Einstein, O.; Zipori, R.; Gross, O.; Birk, R. Fndc5/irisin is regulated by myogenesis stage, irisin, muscle type and training. Am. J. Transl. Res. 2022, 14, 7063–7079. [Google Scholar] [PubMed]
  63. Colaianni, G.; Cinti, S.; Colucci, S.; Grano, M. Irisin and musculoskeletal health. Ann. N. Y. Acad. Sci. 2017, 1402, 5–9. [Google Scholar] [CrossRef] [PubMed]
  64. Huh, J.Y.; Dincer, F.; Mesfum, E.; Mantzoros, C.S. Irisin stimulates muscle growth-related genes and regulates adipocyte differentiation and metabolism in humans. Int. J. Obes. 2014, 38, 1538–1544. [Google Scholar] [CrossRef] [PubMed]
  65. Fukushima, Y.; Kurose, S.; Shinno, H.; Thi Thu, H.C.; Takao, N.; Tsutsumi, H.; Hasegawa, T.; Nakajima, T.; Kimura, Y. Effects of Body Weight Reduction on Serum Irisin and Metabolic Parameters in Obese Subjects. Diabetes Metab. J. 2016, 40, 386–395. [Google Scholar] [CrossRef]
  66. Osella, A.R.; Colaianni, G.; Correale, M.; Pesole, P.L.; Bruno, I.; Buongiorno, C.; Deflorio, V.; Leone, C.M.; Colucci, S.C.; Grano, M.; et al. Irisin Serum Levels in Metabolic Syndrome Patients Treated with Three Different Diets: A Post-Hoc Analysis from a Randomized Controlled Clinical Trial. Nutrients 2018, 10, 844. [Google Scholar] [CrossRef] [PubMed]
  67. Cho, E.; Jeong, D.Y.; Kim, J.G.; Lee, S. The Acute Effects of Swimming Exercise on PGC-1alpha-FNDC5/Irisin-UCP1 Expression in Male C57BL/6J Mice. Metabolites 2021, 11, 111. [Google Scholar] [CrossRef]
  68. Ko, B.J.; Park, K.H.; Shin, S.; Zaichenko, L.; Davis, C.R.; Crowell, J.A.; Joung, H.; Mantzoros, C.S. Diet quality and diet patterns in relation to circulating cardiometabolic biomarkers. Clin. Nutr. 2016, 35, 484–490. [Google Scholar] [CrossRef]
  69. Zafar, M.I.; Mills, K.E.; Zheng, J.; Regmi, A.; Hu, S.Q.; Gou, L.; Chen, L.L. Low-glycemic index diets as an intervention for diabetes: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2019, 110, 891–902. [Google Scholar] [CrossRef]
  70. Reginster, J.Y.; Beaudart, C.; Buckinx, F.; Bruyere, O. Osteoporosis and sarcopenia: Two diseases or one? Curr. Opin. Clin. Nutr. Metab. Care 2016, 19, 31–36. [Google Scholar] [CrossRef] [PubMed]
  71. Estell, E.G.; Le, P.T.; Vegting, Y.; Kim, H.; Wrann, C.; Bouxsein, M.L.; Nagano, K.; Baron, R.; Spiegelman, B.M.; Rosen, C.J. Irisin directly stimulates osteoclastogenesis and bone resorption in vitro and in vivo. eLife 2020, 9, e58172. [Google Scholar] [CrossRef]
  72. Lewiecki, E.M. Role of sclerostin in bone and cartilage and its potential as a therapeutic target in bone diseases. Ther. Adv. Musculoskelet. Dis. 2014, 6, 48–57. [Google Scholar] [CrossRef] [PubMed]
  73. Colaianni, G.; Sanesi, L.; Storlino, G.; Brunetti, G.; Colucci, S.; Grano, M. Irisin and Bone: From Preclinical Studies to the Evaluation of Its Circulating Levels in Different Populations of Human Subjects. Cells 2019, 8, 451. [Google Scholar] [CrossRef]
  74. Ulualan, G.; Kiraz, Z.K.; Kirel, B. Relation of serum irisin levels to obesity and non-alcoholic fatty liver disease. Turk. J. Pediatr. 2022, 64, 246–254. [Google Scholar] [CrossRef] [PubMed]
  75. Oguz, A.; Sahin, M.; Tuzun, D.; Kurutas, E.B.; Ulgen, C.; Bozkus, O.; Gul, K. Irisin is a predictor of sarcopenic obesity in type 2 diabetes mellitus: A cross-sectional study. Medicine 2021, 100, e26529. [Google Scholar] [CrossRef] [PubMed]
  76. Thompson, W.G.; Cook, D.A.; Clark, M.M.; Bardia, A.; Levine, J.A. Treatment of obesity. Mayo Clin. Proc. 2007, 82, 93–101; quiz 101–102. [Google Scholar] [CrossRef] [PubMed]
  77. Ruban, A.; Stoenchev, K.; Ashrafian, H.; Teare, J. Current treatments for obesity. Clin. Med. 2019, 19, 205–212. [Google Scholar] [CrossRef]
  78. Bernal Rivas, C.; Llamunao Tropa, A.; Reyes Barria, A.; Halabi, D.; Pavicic, F.; Ehrenfeld, P.; Martinez Huenchullan, S. Effects of exercise on irisin in subjects with overweight or obesity. A systematic review of clinical studies. Nutr. Hosp. 2022, 39, 1389–1396. [Google Scholar] [CrossRef] [PubMed]
  79. Mahgoub, M.O.; D’Souza, C.; Al Darmaki, R.; Baniyas, M.; Adeghate, E. An update on the role of irisin in the regulation of endocrine and metabolic functions. Peptides 2018, 104, 15–23. [Google Scholar] [CrossRef]
  80. Choi, S.S.; Kim, E.S.; Jung, J.E.; Marciano, D.P.; Jo, A.; Koo, J.Y.; Choi, S.Y.; Yang, Y.R.; Jang, H.J.; Kim, E.K.; et al. PPARgamma Antagonist Gleevec Improves Insulin Sensitivity and Promotes the Browning of White Adipose Tissue. Diabetes 2016, 65, 829–839. [Google Scholar] [CrossRef] [PubMed]
  81. Polyzos, S.A.; Anastasilakis, A.D.; Efstathiadou, Z.A.; Makras, P.; Perakakis, N.; Kountouras, J.; Mantzoros, C.S. Irisin in metabolic diseases. Endocrine 2018, 59, 260–274. [Google Scholar] [CrossRef] [PubMed]
  82. Reiner, M.; Niermann, C.; Jekauc, D.; Woll, A. Long-term health benefits of physical activity--a systematic review of longitudinal studies. BMC Public. Health 2013, 13, 813. [Google Scholar] [CrossRef]
  83. Vissers, D.; Hens, W.; Taeymans, J.; Baeyens, J.P.; Poortmans, J.; Van Gaal, L. The effect of exercise on visceral adipose tissue in overweight adults: A systematic review and meta-analysis. PLoS ONE 2013, 8, e56415. [Google Scholar] [CrossRef]
  84. Montero-Fernández, N.; Serra-Rexach, J.A. Role of exercise on sarcopenia in the elderly. Eur. J. Phys. Rehabil. Med. 2013, 49, 131–143. [Google Scholar]
  85. Johnston, A.P.; De Lisio, M.; Parise, G. Resistance training, sarcopenia, and the mitochondrial theory of aging. Appl. Physiol. Nutr. Metab. 2008, 33, 191–199. [Google Scholar] [CrossRef]
  86. Harber, M.P.; Konopka, A.R.; Undem, M.K.; Hinkley, J.M.; Minchev, K.; Kaminsky, L.A.; Trappe, T.A.; Trappe, S. Aerobic exercise training induces skeletal muscle hypertrophy and age-dependent adaptations in myofiber function in young and older men. J. Appl. Physiol. 2012, 113, 1495–1504. [Google Scholar] [CrossRef]
  87. Wilkinson, S.B.; Phillips, S.M.; Atherton, P.J.; Patel, R.; Yarasheski, K.E.; Tarnopolsky, M.A.; Rennie, M.J. Differential effects of resistance and endurance exercise in the fed state on signalling molecule phosphorylation and protein synthesis in human muscle. J. Physiol. 2008, 586, 3701–3717. [Google Scholar] [CrossRef]
  88. Scalzo, R.L.; Peltonen, G.L.; Binns, S.E.; Shankaran, M.; Giordano, G.R.; Hartley, D.A.; Klochak, A.L.; Lonac, M.C.; Paris, H.L.; Szallar, S.E.; et al. Greater muscle protein synthesis and mitochondrial biogenesis in males compared with females during sprint interval training. FASEB J. 2014, 28, 2705–2714. [Google Scholar] [CrossRef]
  89. Bouchonville, M.F.; Villareal, D.T. Sarcopenic obesity: How do we treat it? Curr. Opin. Endocrinol. Diabetes Obes. 2013, 20, 412–419. [Google Scholar] [CrossRef]
  90. Cipriani, C.; Pepe, J.; Piemonte, S.; Colangelo, L.; Cilli, M.; Minisola, S. Vitamin d and its relationship with obesity and muscle. Int. J. Endocrinol. 2014, 2014, 841248. [Google Scholar] [CrossRef]
  91. Di Filippo, L.; De Lorenzo, R.; Giustina, A.; Rovere-Querini, P.; Conte, C. Vitamin D in Osteosarcopenic Obesity. Nutrients 2022, 14, 1816. [Google Scholar] [CrossRef]
  92. de Brito Galvao, J.F.; Nagode, L.A.; Schenck, P.A.; Chew, D.J. Calcitriol, calcidiol, parathyroid hormone, and fibroblast growth factor-23 interactions in chronic kidney disease. J. Vet. Emerg. Crit. Care 2013, 23, 134–162. [Google Scholar] [CrossRef] [PubMed]
  93. Khundmiri, S.J.; Murray, R.D.; Lederer, E. PTH and Vitamin D. Compr. Physiol. 2016, 6, 561–601. [Google Scholar] [CrossRef] [PubMed]
  94. Medeiros, J.F.P.; de Oliveira Borges, M.V.; Soares, A.A.; Dos Santos, J.C.; de Oliveira, A.B.B.; da Costa, C.H.B.; Cruz, M.S.; Bortolin, R.H.; de Freitas, R.C.C.; Dantas, P.M.S.; et al. The impact of vitamin D supplementation on VDR gene expression and body composition in monozygotic twins: Randomized controlled trial. Sci. Rep. 2020, 10, 11943. [Google Scholar] [CrossRef] [PubMed]
  95. Wimalawansa, S.J. Vitamin D Deficiency: Effects on Oxidative Stress, Epigenetics, Gene Regulation, and Aging. Biology 2019, 8, 30. [Google Scholar] [CrossRef]
  96. Labudzynskyi, D.; Shymanskyy, I.; Veliky, M. Role of vitamin D3 in regulation of interleukin-6 and osteopontin expression in liver of diabetic mice. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 2916–2919. [Google Scholar]
  97. Weber, P. Vitamin K and bone health. Nutrition 2001, 17, 880–887. [Google Scholar] [CrossRef]
  98. Ronning, S.B.; Pedersen, M.E.; Berg, R.S.; Kirkhus, B.; Rodbotten, R. Vitamin K2 improves proliferation and migration of bovine skeletal muscle cells in vitro. PLoS ONE 2018, 13, e0195432. [Google Scholar] [CrossRef]
  99. Ferri, E.; Marzetti, E.; Calvani, R.; Picca, A.; Cesari, M.; Arosio, B. Role of Age-Related Mitochondrial Dysfunction in Sarcopenia. Int. J. Mol. Sci. 2020, 21, 5236. [Google Scholar] [CrossRef]
  100. Knapen, M.H.J.; Jardon, K.M.; Vermeer, C. Vitamin K-induced effects on body fat and weight: Results from a 3-year vitamin K2 intervention study. Eur. J. Clin. Nutr. 2018, 72, 136–141. [Google Scholar] [CrossRef]
  101. Tieland, M.; Trouwborst, I.; Clark, B.C. Skeletal muscle performance and ageing. J. Cachexia Sarcopenia Muscle 2018, 9, 3–19. [Google Scholar] [CrossRef]
  102. Kirk, B.; Zanker, J.; Duque, G. Osteosarcopenia: Epidemiology, diagnosis, and treatment-facts and numbers. J. Cachexia Sarcopenia Muscle 2020, 11, 609–618. [Google Scholar] [CrossRef]
  103. Coletti, C.; Acosta, G.F.; Keslacy, S.; Coletti, D. Exercise-mediated reinnervation of skeletal muscle in elderly people: An update. Eur. J. Transl. Myol. 2022, 32, 10416. [Google Scholar] [CrossRef] [PubMed]
  104. van Dronkelaar, C.; van Velzen, A.; Abdelrazek, M.; van der Steen, A.; Weijs, P.J.M.; Tieland, M. Minerals and Sarcopenia; The Role of Calcium, Iron, Magnesium, Phosphorus, Potassium, Selenium, Sodium, and Zinc on Muscle Mass, Muscle Strength, and Physical Performance in Older Adults: A Systematic Review. J. Am. Med. Dir. Assoc. 2018, 19, 6–11.e13. [Google Scholar] [CrossRef] [PubMed]
  105. Papadopoulou, S.K.; Papadimitriou, K.; Voulgaridou, G.; Georgaki, E.; Tsotidou, E.; Zantidou, O.; Papandreou, D. Exercise and Nutrition Impact on Osteoporosis and Sarcopenia—The Incidence of Osteosarcopenia: A Narrative Review. Nutrients 2021, 13, 4499. [Google Scholar] [CrossRef]
  106. Jin, W.S.; Choi, E.J.; Lee, S.Y.; Bae, E.J.; Lee, T.H.; Park, J. Relationships among Obesity, Sarcopenia, and Osteoarthritis in the Elderly. J. Obes. Metab. Syndr. 2017, 26, 36–44. [Google Scholar] [CrossRef] [PubMed]
  107. Skinner, J.W.; Otzel, D.M.; Bowser, A.; Nargi, D.; Agarwal, S.; Peterson, M.D.; Zou, B.; Borst, S.E.; Yarrow, J.F. Muscular responses to testosterone replacement vary by administration route: A systematic review and meta-analysis. J. Cachexia Sarcopenia Muscle 2018, 9, 465–481. [Google Scholar] [CrossRef] [PubMed]
  108. Snyder, P.J.; Peachey, H.; Hannoush, P.; Berlin, J.A.; Loh, L.; Holmes, J.H.; Dlewati, A.; Staley, J.; Santanna, J.; Kapoor, S.C.; et al. Effect of testosterone treatment on bone mineral density in men over 65 years of age. J. Clin. Endocrinol. Metab. 1999, 84, 1966–1972. [Google Scholar] [CrossRef] [PubMed]
  109. Griggs, R.C.; Kingston, W.; Jozefowicz, R.F.; Herr, B.E.; Forbes, G.; Halliday, D. Effect of testosterone on muscle mass and muscle protein synthesis. J. Appl. Physiol. 1989, 66, 498–503. [Google Scholar] [CrossRef]
  110. Herbst, K.L.; Bhasin, S. Testosterone action on skeletal muscle. Curr. Opin. Clin. Nutr. Metab. Care 2004, 7, 271–277. [Google Scholar] [CrossRef]
  111. Zamir, A.; Ben-Zeev, T.; Hoffman, J.R. Manipulation of Dietary Intake on Changes in Circulating Testosterone Concentrations. Nutrients 2021, 13, 3375. [Google Scholar] [CrossRef]
  112. Kuchakulla, M.; Narasimman, M.; Soni, Y.; Leong, J.Y.; Patel, P.; Ramasamy, R. A systematic review and evidence-based analysis of ingredients in popular male testosterone and erectile dysfunction supplements. Int. J. Impot. Res. 2021, 33, 311–317. [Google Scholar] [CrossRef]
  113. Wilson, J.M.; Lowery, R.P.; Roberts, M.D.; Sharp, M.H.; Joy, J.M.; Shields, K.A.; Partl, J.M.; Volek, J.S.; D’Agostino, D.P. Effects of Ketogenic Dieting on Body Composition, Strength, Power, and Hormonal Profiles in Resistance Training Men. J. Strength. Cond. Res. 2020, 34, 3463–3474. [Google Scholar] [CrossRef] [PubMed]
  114. Calder, P.C. Functional Roles of Fatty Acids and Their Effects on Human Health. JPEN J. Parenter. Enter. Nutr. 2015, 39, 18S–32S. [Google Scholar] [CrossRef] [PubMed]
  115. Roche, H.M. Unsaturated fatty acids. Proc. Nutr. Soc. 1999, 58, 397–401. [Google Scholar] [CrossRef]
  116. Soni, N.; Ross, A.B.; Scheers, N.; Nookaew, I.; Gabrielsson, B.G.; Sandberg, A.S. The Omega-3 Fatty Acids EPA and DHA, as a Part of a Murine High-Fat Diet, Reduced Lipid Accumulation in Brown and White Adipose Tissues. Int. J. Mol. Sci. 2019, 20, 5895. [Google Scholar] [CrossRef] [PubMed]
  117. Felix-Soriano, E.; Sainz, N.; Fernandez-Galilea, M.; Gil-Iturbe, E.; Celay, J.; Martinez-Climent, J.A.; Moreno-Aliaga, M.J. Chronic docosahexaenoic acid supplementation improves metabolic plasticity in subcutaneous adipose tissue of aged obese female mice. J. Nutr. Biochem. 2023, 111, 109153. [Google Scholar] [CrossRef]
  118. Lee, M.S.; Shin, Y.; Moon, S.; Kim, S.; Kim, Y. Effects of Eicosapentaenoic Acid and Docosahexaenoic Acid on Mitochondrial DNA Replication and PGC-1alpha Gene Expression in C(2)C(12) Muscle Cells. Prev. Nutr. Food Sci. 2016, 21, 317–322. [Google Scholar] [CrossRef]
  119. Ochi, E.; Tsuchiya, Y. Eicosapentaenoic Acid (EPA) and Docosahexaenoic Acid (DHA) in Muscle Damage and Function. Nutrients 2018, 10, 552. [Google Scholar] [CrossRef]
  120. Castillero, E.; Martin, A.I.; Lopez-Menduina, M.; Villanua, M.A.; Lopez-Calderon, A. Eicosapentaenoic acid attenuates arthritis-induced muscle wasting acting on atrogin-1 and on myogenic regulatory factors. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2009, 297, R1322–R1331. [Google Scholar] [CrossRef]
  121. LeMieux, M.J.; Kalupahana, N.S.; Scoggin, S.; Moustaid-Moussa, N. Eicosapentaenoic acid reduces adipocyte hypertrophy and inflammation in diet-induced obese mice in an adiposity-independent manner. J. Nutr. 2015, 145, 411–417. [Google Scholar] [CrossRef]
  122. Thach, T.T.; Wu, C.; Hwang, K.Y.; Lee, S.J. Azelaic Acid Induces Mitochondrial Biogenesis in Skeletal Muscle by Activation of Olfactory Receptor 544. Front. Physiol. 2020, 11, 329. [Google Scholar] [CrossRef]
  123. Salucci, S.; Falcieri, E. Polyphenols and their potential role in preventing skeletal muscle atrophy. Nutr. Res. 2020, 74, 10–22. [Google Scholar] [CrossRef]
  124. Noh, J.S.; Park, C.H.; Yokozawa, T. Treatment with oligonol, a low-molecular polyphenol derived from lychee fruit, attenuates diabetes-induced hepatic damage through regulation of oxidative stress and lipid metabolism. Br. J. Nutr. 2011, 106, 1013–1022. [Google Scholar] [CrossRef]
  125. Park, J.Y.; Kim, Y.; Im, J.A.; Lee, H. Oligonol suppresses lipid accumulation and improves insulin resistance in a palmitate-induced in HepG2 hepatocytes as a cellular steatosis model. BMC Complement. Altern. Med. 2015, 15, 185. [Google Scholar] [CrossRef]
  126. Liu, H.W.; Chen, Y.J.; Chang, Y.C.; Chang, S.J. Oligonol, a Low-Molecular Weight Polyphenol Derived from Lychee, Alleviates Muscle Loss in Diabetes by Suppressing Atrogin-1 and MuRF1. Nutrients 2017, 9, 1040. [Google Scholar] [CrossRef]
  127. Chang, Y.C.; Chen, Y.T.; Liu, H.W.; Chan, Y.C.; Liu, M.Y.; Hu, S.H.; Tseng, W.T.; Wu, H.L.; Wang, M.F.; Chang, S.J. Oligonol Alleviates Sarcopenia by Regulation of Signaling Pathways Involved in Protein Turnover and Mitochondrial Quality. Mol. Nutr. Food Res. 2019, 63, e1801102. [Google Scholar] [CrossRef]
  128. Hewlings, S.J.; Kalman, D.S. Curcumin: A Review of Its Effects on Human Health. Foods 2017, 6, 92. [Google Scholar] [CrossRef]
  129. Bahijri, S.M.; Ajabnoor, G.; Hegazy, G.A.; Alsheikh, L.; Moumena, M.Z.; Bashanfar, B.M.; Alzahrani, A.H. Supplementation with Oligonol, Prevents Weight Gain and Improves Lipid Profile in Overweight and Obese Saudi Females. Curr. Nutr. Food Sci. 2018, 14, 164–170. [Google Scholar] [CrossRef]
  130. Kim, J.H.; Lee, H.; Kim, J.M.; Lee, B.J.; Kim, I.J.; Pak, K.; Jeon, Y.K.; Kim, K. Effect of oligonol, a lychee-derived polyphenol, on skeletal muscle in ovariectomized rats by regulating body composition, protein turnover, and mitochondrial quality signaling. Food Sci. Nutr. 2022, 10, 1184–1194. [Google Scholar] [CrossRef]
  131. Bagherniya, M.; Mahdavi, A.; Shokri-Mashhadi, N.; Banach, M.; Von Haehling, S.; Johnston, T.P.; Sahebkar, A. The beneficial therapeutic effects of plant-derived natural products for the treatment of sarcopenia. J. Cachexia Sarcopenia Muscle 2022, 13, 2772–2790. [Google Scholar] [CrossRef]
  132. Safdar, A.; de Beer, J.; Tarnopolsky, M.A. Dysfunctional Nrf2-Keap1 redox signaling in skeletal muscle of the sedentary old. Free Radic. Biol. Med. 2010, 49, 1487–1493. [Google Scholar] [CrossRef] [PubMed]
  133. Balogun, E.; Hoque, M.; Gong, P.; Killeen, E.; Green, C.J.; Foresti, R.; Alam, J.; Motterlini, R. Curcumin activates the haem oxygenase-1 gene via regulation of Nrf2 and the antioxidant-responsive element. Biochem. J. 2003, 371, 887–895. [Google Scholar] [CrossRef] [PubMed]
  134. Shen, G.; Xu, C.; Hu, R.; Jain, M.R.; Gopalkrishnan, A.; Nair, S.; Huang, M.T.; Chan, J.Y.; Kong, A.N. Modulation of nuclear factor E2-related factor 2-mediated gene expression in mice liver and small intestine by cancer chemopreventive agent curcumin. Mol. Cancer Ther. 2006, 5, 39–51. [Google Scholar] [CrossRef]
  135. Sahin, K.; Pala, R.; Tuzcu, M.; Ozdemir, O.; Orhan, C.; Sahin, N.; Juturu, V. Curcumin prevents muscle damage by regulating NF-kappaB and Nrf2 pathways and improves performance: An in vivo model. J. Inflamm. Res. 2016, 9, 147–154. [Google Scholar] [CrossRef]
  136. Receno, C.N.; Liang, C.; Korol, D.L.; Atalay, M.; Heffernan, K.S.; Brutsaert, T.D.; DeRuisseau, K.C. Effects of Prolonged Dietary Curcumin Exposure on Skeletal Muscle Biochemical and Functional Responses of Aged Male Rats. Int. J. Mol. Sci. 2019, 20, 1178. [Google Scholar] [CrossRef] [PubMed]
  137. Alway, S.E.; Bennett, B.T.; Wilson, J.C.; Sperringer, J.; Mohamed, J.S.; Edens, N.K.; Pereira, S.L. Green tea extract attenuates muscle loss and improves muscle function during disuse, but fails to improve muscle recovery following unloading in aged rats. J. Appl. Physiol. 2015, 118, 319–330. [Google Scholar] [CrossRef] [PubMed]
  138. da Silva, W.; Machado, A.S.; Souza, M.A.; Mello-Carpes, P.B.; Carpes, F.P. Effect of green tea extract supplementation on exercise-induced delayed onset muscle soreness and muscular damage. Physiol. Behav. 2018, 194, 77–82. [Google Scholar] [CrossRef] [PubMed]
  139. Bagheri, R.; Rashidlamir, A.; Ashtary-Larky, D.; Wong, A.; Grubbs, B.; Motevalli, M.S.; Baker, J.S.; Laher, I.; Zouhal, H. Effects of green tea extract supplementation and endurance training on irisin, pro-inflammatory cytokines, and adiponectin concentrations in overweight middle-aged men. Eur. J. Appl. Physiol. 2020, 120, 915–923. [Google Scholar] [CrossRef]
  140. Chen, I.J.; Liu, C.Y.; Chiu, J.P.; Hsu, C.H. Therapeutic effect of high-dose green tea extract on weight reduction: A randomized, double-blind, placebo-controlled clinical trial. Clin. Nutr. 2016, 35, 592–599. [Google Scholar] [CrossRef]
  141. Ueda-Wakagi, M.; Hayashibara, K.; Nagano, T.; Ikeda, M.; Yuan, S.; Ueda, S.; Shirai, Y.; Yoshida, K.I.; Ashida, H. Epigallocatechin gallate induces GLUT4 translocation in skeletal muscle through both PI3K- and AMPK-dependent pathways. Food Funct. 2018, 9, 4223–4233. [Google Scholar] [CrossRef]
  142. Casanova, E.; Salvado, J.; Crescenti, A.; Gibert-Ramos, A. Epigallocatechin Gallate Modulates Muscle Homeostasis in Type 2 Diabetes and Obesity by Targeting Energetic and Redox Pathways: A Narrative Review. Int. J. Mol. Sci. 2019, 20, 532. [Google Scholar] [CrossRef] [PubMed]
  143. 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]
  144. Luk, H.Y.; Appell, C.; Chyu, M.C.; Chen, C.H.; Wang, C.Y.; Yang, R.S.; Shen, C.L. Impacts of Green Tea on Joint and Skeletal Muscle Health: Prospects of Translational Nutrition. Antioxidants 2020, 9, 1050. [Google Scholar] [CrossRef]
  145. Musial, C.; Kuban-Jankowska, A.; Gorska-Ponikowska, M. Beneficial Properties of Green Tea Catechins. Int. J. Mol. Sci. 2020, 21, 1744. [Google Scholar] [CrossRef] [PubMed]
  146. Sabarathinam, S.; Rajappan Chandra, S.K.; Satheesh, S. Network pharmacology based pharmacokinetic assessment and evaluation of the therapeutic potential of catechin derivatives as a potential myostatin inhibitor: A special view on Sarcopenic Obesity. Nat. Prod. Res. 2023, 1–5. [Google Scholar] [CrossRef] [PubMed]
  147. Mafi, F.; Biglari, S.; Ghardashi Afousi, A.; 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] [PubMed]
  148. Yahyazadeh, R.; Ghasemzadeh Rahbardar, M.; Razavi, B.M.; Karimi, G.; Hosseinzadeh, H. The effect of Elettaria cardamomum (cardamom) on the metabolic syndrome: Narrative review. Iran. J. Basic. Med. Sci. 2021, 24, 1462–1469. [Google Scholar] [CrossRef]
  149. Daneshi-Maskooni, M.; Keshavarz, S.A.; Qorbani, M.; Mansouri, S.; Alavian, S.M.; Badri-Fariman, M.; Jazayeri-Tehrani, S.A.; Sotoudeh, G. Green cardamom supplementation improves serum irisin, glucose indices, and lipid profiles in overweight or obese non-alcoholic fatty liver disease patients: A double-blind randomized placebo-controlled clinical trial. BMC Complement. Altern. Med. 2019, 19, 59. [Google Scholar] [CrossRef]
  150. Daneshi-Maskooni, M.; Keshavarz, S.A.; Mansouri, S.; Qorbani, M.; Alavian, S.M.; Badri-Fariman, M.; Jazayeri-Tehrani, S.A.; Sotoudeh, G. The effects of green cardamom on blood glucose indices, lipids, inflammatory factors, paraxonase-1, sirtuin-1, and irisin in patients with nonalcoholic fatty liver disease and obesity: Study protocol for a randomized controlled trial. Trials 2017, 18, 260. [Google Scholar] [CrossRef]
  151. Held, S.; Schieberle, P.; Somoza, V. Characterization of alpha-terpineol as an anti-inflammatory component of orange juice by in vitro studies using oral buccal cells. J. Agric. Food Chem. 2007, 55, 8040–8046. [Google Scholar] [CrossRef]
  152. de Oliveira, M.G.; Marques, R.B.; de Santana, M.F.; Santos, A.B.; Brito, F.A.; Barreto, E.O.; De Sousa, D.P.; Almeida, F.R.; Badaue-Passos, D., Jr.; Antoniolli, A.R.; et al. alpha-terpineol reduces mechanical hypernociception and inflammatory response. Basic. Clin. Pharmacol. Toxicol. 2012, 111, 120–125. [Google Scholar] [CrossRef] [PubMed]
  153. Sousa, G.M.; Cazarin, C.B.B.; Marostica Junior, M.R.; Lamas, C.A.; Quitete, V.; Pastore, G.M.; Bicas, J.L. The effect of alpha-terpineol enantiomers on biomarkers of rats fed a high-fat diet. Heliyon 2020, 6, e03752. [Google Scholar] [CrossRef] [PubMed]
  154. Nascimento, S.S.; Camargo, E.A.; DeSantana, J.M.; Araujo, A.A.; Menezes, P.P.; Lucca-Junior, W.; Albuquerque-Junior, R.L.; Bonjardim, L.R.; Quintans-Junior, L.J. Linalool and linalool complexed in beta-cyclodextrin produce anti-hyperalgesic activity and increase Fos protein expression in animal model for fibromyalgia. Naunyn Schmiedebergs Arch. Pharmacol. 2014, 387, 935–942. [Google Scholar] [CrossRef] [PubMed]
  155. Soundharrajan, I.; Kim, D.H.; Srisesharam, S.; Kuppusamy, P.; Sivanesan, R.; Choi, K.C. Limonene promotes osteoblast differentiation and 2-deoxy-d-glucose uptake through p38MAPK and Akt signaling pathways in C2C12 skeletal muscle cells. Phytomedicine 2018, 45, 41–48. [Google Scholar] [CrossRef]
  156. Santos, M.M.B.; Filho, L.F.S.; De Souza, J.B.; Filho, J.; Mesquita, T.R.R.; Santos, M.S.; De Vasconcelos, C.M.L.; Lauton-Santos, S.; De Oliveira, E.D. Topical application of (S)-(-)-limonene is as effective as phonophoresis for improving oxidative parameters of injured skeletal muscle in rats. Naunyn Schmiedebergs Arch. Pharmacol. 2020, 393, 2293–2300. [Google Scholar] [CrossRef]
  157. Tesfaye, A. Revealing the Therapeutic Uses of Garlic (Allium sativum) and Its Potential for Drug Discovery. Sci. World J. 2021, 2021, 8817288. [Google Scholar] [CrossRef]
  158. Soleimani, D.; Paknahad, Z.; Askari, G.; Iraj, B.; Feizi, A. Effect of garlic powder consumption on body composition in patients with nonalcoholic fatty liver disease: A randomized, double-blind, placebo-controlled trial. Adv. Biomed. Res. 2016, 5, 2. [Google Scholar] [CrossRef]
  159. Sangouni, A.A.; Mohammad Hosseini Azar, M.R.; Alizadeh, M. Effects of garlic powder supplementation on insulin resistance, oxidative stress, and body composition in patients with non-alcoholic fatty liver disease: A randomized controlled clinical trial. Complement. Ther. Med. 2020, 51, 102428. [Google Scholar] [CrossRef]
  160. Avula, P.R.; Asdaq, S.M.; Asad, M. Effect of aged garlic extract and s-allyl cysteine and their interaction with atenolol during isoproterenol induced myocardial toxicity in rats. Indian J. Pharmacol. 2014, 46, 94–99. [Google Scholar] [CrossRef]
  161. Gupta, P.; Dutt, V.; Kaur, N.; Kalra, P.; Gupta, S.; Dua, A.; Dabur, R.; Saini, V.; Mittal, A. S-allyl cysteine: A potential compound against skeletal muscle atrophy. Biochim. Biophys. Acta Gen. Subj. 2020, 1864, 129676. [Google Scholar] [CrossRef]
  162. Dutt, V.; Saini, V.; Gupta, P.; Kaur, N.; Bala, M.; Gujar, R.; Grewal, A.; Gupta, S.; Dua, A.; Mittal, A. S-allyl cysteine inhibits TNFalpha-induced skeletal muscle wasting through suppressing proteolysis and expression of inflammatory molecules. Biochim. Biophys. Acta Gen. Subj. 2018, 1862, 895–906. [Google Scholar] [CrossRef] [PubMed]
  163. de Macedo, S.M.; Lelis, D.F.; Mendes, K.L.; Fraga, C.A.C.; Brandi, I.V.; Feltenberger, J.D.; Farias, L.C.; Guimaraes, A.L.S.; de Paula, A.M.B.; Santos, S.H.S. Effects of Dietary Macronutrient Composition on FNDC5 and Irisin in Mice Skeletal Muscle. Metab. Syndr. Relat. Disord. 2017, 15, 161–169. [Google Scholar] [CrossRef] [PubMed]
  164. Zafar, M.I.; Mills, K.E.; Zheng, J.; Peng, M.M.; Ye, X.; Chen, L.L. Low glycaemic index diets as an intervention for obesity: A systematic review and meta-analysis. Obes. Rev. 2019, 20, 290–315. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanism of sarcopenic obesity (SO) and its association with endogenous metabolites.
Figure 1. Mechanism of sarcopenic obesity (SO) and its association with endogenous metabolites.
Nutrients 15 03854 g001
Figure 2. Mechanism of irisin and its intervention with myostatin and body components.
Figure 2. Mechanism of irisin and its intervention with myostatin and body components.
Nutrients 15 03854 g002
Table 2. Interaction between irisin and sarcopenic or obese patients and causes and potential consequences of irisin secretion.
Table 2. Interaction between irisin and sarcopenic or obese patients and causes and potential consequences of irisin secretion.
ReferenceIntervention
(Exposure, Dose, Duration, Follow-Up)
ConditionSubjects
(Sample Size, Gender, Age)
Results
Norheim et al. [60]Endurance and strength training
12 weeks
Inactive, hyperglycaemic,
overweight, pre-diabetic
Normal weight
n = 13
Age: 40–65
Obese group
n = 13
Age: 40–65
Irisin ↑
PGC-1α ↑
FNDC5 ↑
Tsuchiya et al. [61]High-intensity exercise
40 min
Healthy, sedentaryn = 6
Age: 22.5 ± 1.1
Height: 174.8 ± 2.8 cm
Weight: 67.1 ± 2.2 kg
BMI: 22.1 ± 1.1
Irisin ↑
LDH ↓
Vaughan et al. [59]Irisin 5 nM
24 h
5 × 10⁵ cells/well,
DMEM containing 4500 mg/L glucose,
10% FBS, 100 U/mL penicillin/streptomycin,
37 °C, 5% CO₂
C2C12 muscle cellMitochondrial content ↑
NRF-1 ↑
TFAM ↑
GLUT4 ↑
UCP-3 ↑
Huh et al., (2014) [64]Irisin 10 nM, 50 nM
12 days
Muscle collected from healthy subjects3T3L1 adipocyteIrisin ↑
FNDC5 ↑
IGF-1 ↑
PGC-1α ↑
UCP-1 ↑
Rashid et al. [56]Moderate exercise
6 months
ObesityNormal group
n = 30
Age: 20–43
BMI: <25 kg/m2
Obese group
n = 30
Age: 20–43
BMI: ≥30 kg/m2
Irisin ↑
BMI ↓
Waist circumference ↓
Fasting glucose level ↓
Fasting insulin level ↓
HOMA-IR ↓
HOMA-B2 ↓
Fukushima et al. [65]Diet, exercise, behavioral therapy
6 months
Obesityn = 22
5 males, 17 females
Age: 46.1 ± 16.0
BMI: 36.9 ± 5.0 kg/m2
Irisin ↑
BMI ↓
Body fat percentage ↓
Subcutaneous fat area ↓
Triglycerides ↓
HOMA-IR ↓
Fasting glucose level ↓
Osella et al. [66]Low glycaemic index diet, Mediterranean diet, Low glycaemic index Mediterranean diet
24 weeks
Metabolic syndromeControl group
n = 80
LGID group
n = 55
MD group
n = 51
LGIMD group
n = 45
Irisin ↑
Saturated fatty acids ↓
Fat free mass ↑
BMI ↓
Cho et al. [67]Swimming
90 min
Acclimitized to swimmingC57BL/6JFNDC5 ↑
Control group
n = 10
Age: 14–16 weeks
Swimming exercise group
n = 10
Age: 14–16 weeks
Mâcedo et al. [68]High protein diet
60 days
-Standard diet mice
n = 7
High protein diet mice
n = 7
Brown adipose tissue ↑
Zafar et al. [69]Low glycemic index diet
≥1 week
Insulin resistance, T1DM, T2DMn = 54Glycated hemoglobin ↓
Fasting glucose ↓
BMI ↓
Total cholesterol ↓
LDL ↓
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kim, Y.-C.; Ki, S.-W.; Kim, H.; Kang, S.; Kim, H.; Go, G.-w. Recent Advances in Nutraceuticals for the Treatment of Sarcopenic Obesity. Nutrients 2023, 15, 3854. https://doi.org/10.3390/nu15173854

AMA Style

Kim Y-C, Ki S-W, Kim H, Kang S, Kim H, Go G-w. Recent Advances in Nutraceuticals for the Treatment of Sarcopenic Obesity. Nutrients. 2023; 15(17):3854. https://doi.org/10.3390/nu15173854

Chicago/Turabian Style

Kim, Young-Chan, Sang-Woo Ki, Hannah Kim, Sumin Kang, Hayoon Kim, and Gwang-woong Go. 2023. "Recent Advances in Nutraceuticals for the Treatment of Sarcopenic Obesity" Nutrients 15, no. 17: 3854. https://doi.org/10.3390/nu15173854

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