BMB Reports 2021; 54(12): 581-591  https://doi.org/10.5483/BMBRep.2021.54.12.151
Molecular targets of exercise mimetics and their natural activators
Young Jin Jang1 & Sanguine Byun2,*
1Major of Food Science & Technology, Seoul Women’s University, Seoul 01797, 2Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
Correspondence to: Tel: +82-2-2123-5896; Fax: +82-2-362-7265; E-mail: sanguine@yonsei.ac.kr
Received: September 23, 2021; Revised: November 15, 2021; Accepted: November 24, 2021; Published online: December 31, 2021.
© Korean Society for Biochemistry and Molecular Biology. All rights reserved.

cc This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
ABSTRACT
Physical exercise can be effective in preventing or ameliorating various diseases, including diabetes, cardiovascular diseases, neurodegenerative diseases, and cancer. However, not everyone may be able to participate in exercise due to illnesses, age-related frailty, or difficulty in long-term behavior change. An alternative option is to utilize pharmacological interventions that mimic the positive effects of exercise training. Recent studies have identified signaling pathways associated with the benefits of physical activity and discovered exercise mimetics that can partially simulate the systemic impact of exercise. This review describes the molecular targets for exercise mimetics and their effect on skeletal muscle and other tissues. We will also discuss the potential advantages of using natural products as a multitargeting agent for mimicking the health-promoting effects of exercise.
Keywords: Exercise mimetics, Molecular targets, Myokines, Natural products
INTRODUCTION

The health benefits of exercise have been well-established. Exercise is closely related to health conditions of bone, immune system, brain, and reproductive system as well as skeletal and cardiovascular systems (1). Physical exercise has been shown to have a positive impact on a wide range of diseases including obesity, metabolic diseases, cardiovascular disease, cancer, neurodegenerative disease, and osteoporosis (2, 3). Exercise also has anti-depressant effects and improves immune function, and therefore may contribute as a defense strategy against infectious diseases such as COVID-19 (4, 5). Nevertheless, exercising on a regular basis may not be an option for everyone. Therefore, exercise mimetics, pharmacologic therapeutics that mimic the health benefit effects of exercise, have been proposed as an alternative option (1). Exercise mimetics may, to some extent, generate health benefits without performing actual exercise. Recent studies have identified pathways that are activated during physical exercise and found critical signaling molecules that contribute to the health-promoting effects of exercise. In this review, we will discuss the potential targets of exercise mimetics and the need for developing exercise mimetics from natural sources.

SKELETAL MUSCLE ADAPTATION AND HEALTH BENEFITS OF EXERCISE

Exercise promotes skeletal muscle adaptation and these adaptive changes are the basis for the health benefits of exercise (6). Endurance exercise and resistance exercise induce different adaptive changes to the skeletal muscle (7). The major adaptive changes of endurance exercise include increase in mitochondrial density, oxidative function, and capillarization (7). It is also well-known that endurance exercise promotes transformation of glycolytic muscle fibers to oxidative muscle fibers (2). Oxidative muscle fibers are rich in mitochondria compared with glycolytic muscle fibers, have higher myoglobin content, and are more densely vascularized (2). They also perform increased fatty acid oxidation due to the increased levels of lipid-metabolizing enzymes, which provide extra energy for performance and reduce the dependence on glucose (8). This results in increased lactate tolerance and endurance capacity (8). On the other hand, resistance exercise leads to increased muscle strength and power as a result of neuromuscular adaptation (9). Resistance exercise promotes development of glycolytic muscle fibers and directly increases the size of muscle fibers (9). The enlargement of muscle fibers is attributed to upregulation of protein synthesis and selective hypertrophy of fast twitch fibers (10). Although endurance exercise and resistance exercise both provide health benefits, there can be some differences in the particular effect each type produces. For instance, endurance exercise is known to be more effective in reducing cardiovascular risks, while resistance training can be more effective in maintaining muscle mass and physical function. Combination of endurance exercise and resistance exercise have been reported to be more potent in reducing insulin resistance and functional limitation in abdominally obese adults, compared to either modality alone (7).

Exercise has a positive effect not only on skeletal muscles, but also on various organs and tissues including the heart, brain, adipose tissue, liver, blood vessels, and bones (11). Therefore, the effect of exercise goes beyond improving muscle function and strength, leading to other health-promoting effects on cardiovascular function, memory, immunity, metabolism, and aging (12-14). While the impact of physical training or exercise mimetics on multiple organs are well-documented, the underlying molecular mechanism is still unraveling (15). In this regard, myokines have been suggested as an important factor to explain the multiple benefits of exercise (16). Myokines are peptides synthesized and released by myocytes in response to muscular contraction (16). Myokines are implicated in the autocrine regulation of muscle function as well as in paracrine and endocrine regulation of other tissues and organs including adipose tissue, liver, and brain (16). Secretome profiling of primary human skeletal muscle cells revealed 305 myokines (17). While the role of each myokine is still under investigation, certain myokines appear to have a physiological effect on other parts of the body leading to favorable health outcomes, and thus represent a promising target for exercise mimetics. In addition, studies have found specific genes expressed in multiple tissues that mimic the diverse effects of exercise when activated. Thus, modulating the activity or expression of these genes could potentially simulate certain aspects of physical training. Next, we will describe some of the potential targets of exercise mimetics.

MOLECULAR TARGETS OF EXERCISE MIMETICS

Irisin

Irisin is a hormone-like myokine induced by exercise, and is also expressed in small amounts in bone, brain, and other tissues (18, 19). The peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) is a critical regulator of exercise-induced skeletal muscle adaptation (20). And exercise-driven upregulation of PGC-1α in muscle promotes the synthesis of fibronectin domain-containing protein 5 (FNDC5), which is subsequently cleaved to generate irisin (18, 21). The level of irisin positively correlates with muscle mass and muscle strength (19) and injection of irisin rescues denervation-induced loss of skeletal muscle mass by enhancing satellite cell activation and reducing protein degradation (22). Also, upregulation of the PGC-1α/FNDC5/irisin pathway has been suggested to be responsible for the exercise-mediated accelerated recovery of myopathy through increasing mitochondrial fission and mitophagy (23).

Irisin acts as a link between muscle and other tissue and organs, and has positive effects on obesity, insulin resistance, type 2 diabetes, brain, and bone health (24). Irisin attenuated LPS-induced inflammation in mature adipocytes (25). Exercise has been known to have major impacts on adipose tissue browning and fat metabolism (26). The conversion of white to brown adipose tissue mediated by exercise has been reported to be through inducing irisin which stimulates the expression UCP-1, the master regulator of brown adipose tissue (27). The benefit of physical exercise on bone mineral density is widely-accepted, and irisin has been reported to play an active role between skeletal muscles and bones (19). Irisin promotes cortical bone mass and strength as well as osteoblast differentiation through regulating expression of bone-specific genes and up-stream signaling pathways (24). In addition, exercise increases the hippocampal expression of FNDC5, the precursor of irisin, in mice, in a PGC-1α-dependent manner (28). Irisin stimulates neurogenesis, synaptic plasticity, and cognitive function by upregulating the expression of brain-derived neurotrophic factor (BDNF), demonstrating that irisin may act as a link between exercise and brain function (29).

Brain-derived neurotrophic factor (BDNF)

BDNF is a polypeptide belonging to the neurotrophin family. It regulates neuronal proliferation, differentiation, maturation, and plasticity in neurogenesis (30). Varying intensity of exercise has been reported to increase BDNF mRNA expression in the hippocampus of mice (31, 32). BDNF has been known to play a crucial role in exercise-induced neurogenesis, synaptic plasticity, and improved cognition. Interestingly, plasma concentration of BDNF is also increased by exercise (33). Notably, BDNF is increased in human skeletal muscle after exercise as well as in electrically stimulated muscle cells (34). Induction of BDNF through exercise and its multifaceted effect on the various organs suggests BDNF as a myokine. Running induces upregulation of BDNF in skeletal muscle and is involved in exercise-induced skeletal muscle regeneration (35). BDNF decreases the atrophy of skeletal muscle following exercise and is mediated via AMPK phosphorylation (36). BDNF acts in an autocrine or paracrine fashion with strong effects on peripheral metabolism, including fat oxidation, and subsequent effects on the size of adipose tissue (37). BDNF is also effective against insulin intolerance and has been shown to play an important role in angiogenesis, cardiovascular development, and cardioprotection (38). Furthermore, circulating BDNF levels are decreased in patients with obesity, type 2 diabetes, cardiovascular disease, depression, and Alzheimer’s disease (34).

Interleukin-6 (IL-6)

IL-6 was originally identified as a proinflammatory cytokine, synthesized by the liver and expressed in monocytes and macrophages, contributing to immune responses (1). However, IL-6 is also produced and released by skeletal muscle after prolonged exercise and may function as a myokine, independent from controlling inflammatory responses (39). It is well known that the level of circulating plasma IL-6 as well as expression of IL-6 receptor in skeletal muscle are upregulated after exercise (40, 41). By contrast, the plasma TNF-α level was not increased by exercise and only slightly increased in extremely strenuous exercise conditions such as marathons (40). IL-6 production in muscle is independent of nuclear factor-κB activation, and thus differs from the mechanism observed in immune cells (42). IL-6 has beneficial effects on muscle formation and growth (39). IL-6 knockout mice showed impaired hypertrophic muscle growth, which is attributed to blunted accretion of myonuclei (39). Moreover, several studies suggest that IL-6 acts as a myokine in other organs. Exercise decreases visceral adipose tissues and this effect of exercise is abrogated by IL-6 blockade (43). IL-6 contributes to hepatic glucose production during exercise (44). IL-6 also enhances fat oxidation in skeletal muscle via AMPK activation and increases lipolysis in skeletal muscle with little effect on adipose tissue (39). Additionally, glucose uptake and fatty acid oxidation by IL-6 in skeletal myotube were abolished by an AMPK-dominant negative construct, further suggesting a connection between exercise, AMPK, IL-6, and metabolism (45). Adult IL-6 knockout mice show impaired neurogenesis suggesting that lack of IL-6 might be detrimental to neurogenesis in the adult brain (46). Collectively, induction of IL-6 appears to contribute to the metabolic and neurogenic effects generated by physical exercise.

AMP-activated protein kinase (AMPK)

AMPK is the master regulator of metabolism sensing energy supplies (47). AMPK is activated in skeletal muscle during exercise in response to increased binding of AMP and decreased binding of ATP (48). Transgenic mice carrying inactive muscle-specific AMPK showed reduced exercise capacity and impaired glucose tolerance and insulin response (49). AMPK activation is required for exercise-induced mitochondrial biogenesis via PGC-1α (47). Many studies showed that the AMPK activator, 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) mimics the effects of exercise. AICAR consumption alone enhanced running endurance by 44% and metabolic genes in sedentary mice (50). AICAR increases the levels of glucose transporter type 4 (GLUT4) and mitochondrial enzyme in skeletal muscle (51). AICAR also increases angiogenesis and vascularization by inducing VEGF-A expression, which in turn facilitates stable supply of oxygen and nutrients similar to exercise (52). AICAR was used as a “next-generation” performance-enhancing drug in the Olympic Spanish Cycling Team, and a sports doctor was arrested for doping (2).

AICAR also has a positive effect on other organs. AICAR reduces circulating levels of triglyceride and blood pressure and promotes hepatic fat consumption (53). Further, AICAR inhibits inflammatory response and cytokine levels. AICAR inhibits NF-κB DNA binding and cytokine expression in human macrophages (54). Notably, AICAR treatment improved spatial memory and neurogenesis in spite of the poor permeability through the blood-brain barrier, suggesting that the positive effect of AICAR in the brain is probably due to the indirect effect of AMPK activation in other organs (52, 55). AICAR improved cognition and motor function in mice, but it was abolished in mice carrying mutant muscle-specific AMPKα2 (56). These results suggest the importance of muscle AMPK activation on the effects of AICAR in brain. Although it was a transient effect, AICAR also enhanced hippocampus cell number and BDNF protein levels in mice (57).

Peroxisome proliferator-activated receptor δ (PPARδ)

PPARs are a family of nuclear hormone receptors that sense metabolic status and are involved in lipid metabolism (58). There are three isoforms, PPARα, β/δ, and γ, and PPARδ is the predominant form in skeletal muscle (59). Selective PPARδ agonist GW501516 increased the number of oxidative myofibers and the level of running endurance in adult mice (50). Exercise-induced performance improvement was attenuated in PPARδ-deficient mice (8). These effects are attributed to PPARδ-induced suppression of glucose catabolism; glucose sparing delays hypoglycemia and extends running time (8). PPARδ overexpression increases AMPK activity, and PPARδ activity is also stimulated by AMPK (60). PPARδ appears to interact with AMPK and synergistically regulates exercise endurance genes (50). In line with this, GW501516 has been listed as an illegal drug by the World Anti-Doping Agency similar to AICAR (52).

PPARδ also plays a critical role in metabolic diseases. Constitutive PPARδ activation in mouse adipocytes resulted in reduced fat composition and prevented high-fat diet-induced obesity (61). GW501516 also induces fatty acid oxidation and ameliorates obesity and insulin resistance in mice (62). In obese monkeys, GW501516 attenuated dyslipidemia, lowering triglyceride and LDL-c levels while increasing HDL-c (63). Cardiomyocyte-restricted PPARδ knockout decreased the rate of fatty acid oxidation, resulting in lipid accumulation in the heart (64).

GW501516 has a positive effect on the brain, although it hardly crosses the blood-brain barrier. Administration of GW501516 improves hippocampal neurogenesis and spatial memory (55). These results suggest that the positive effect of GW501516 on the brain is likely due to the indirect exercise mimetic effects (52). GW501516 was developed because of its possible beneficial effects on metabolic diseases and cardiovascular diseases, but its carcinogenic properties were identified in animal studies (52). The discovery of safer small molecules that can increase PPAR activity can be a strategy to develop exercise mimetics.

Estrogen-related receptor γ (ERRγ)

ERRγ is a member belonging to the nuclear receptor super-family and plays a key role in regulating skeletal muscle adaptation to exercise through regulating mitochondria biogenesis, angiogenesis, and oxidative muscle remodeling (65-67). Transgenic mice expressing ERRγ in skeletal muscle exhibit red muscles, larger mitochondria, and improved oxidative capacity and vascularization (68, 69). ERRγ is highly expressed in oxidative and vascularized muscle and is induced by endurance exercise (65). While ERRγ-induced oxidative muscle transformation and vascularization is independent of PGC-1α (68), exercise and ERRγ individually and cooperatively attenuate muscle damage in PGC-1α knockout mice (67). ERRγ is recognized as a promising target of exercise mimetics because of its role in direct regulation of oxidative muscle remodeling (2). Further, overexpression of ERRγ attenuates the symptoms of Duchenne muscular dystrophy and muscle damage (70). These results suggested that genetic activation of ERRγ led to exercise-like phenotype in skeletal muscle with positive effects towards muscular disease (47). However, only a few studies reported the effects of ERRγ agonist on skeletal muscle or muscular disease. ERRγ agonist GSK4716 increases genes involved in mitochondrial biogenesis, fatty acid oxidation, and TCA cycle in mouse myotubes (69).

However, studies on activating ERRγ in other organs have not always met with positive results. ERRγ was reported to block hepatic insulin signaling via transcriptionally regulating LIPIN1 expression (71). Inverse agonist of ERRγ also ameliorates chronic alcohol-induced liver injury in mice (72). Also, treatment with an inverse agonist of ERRγ resulted in antimicrobial effect and improved host survival (73). However, the systemic effect of ERRγ activation in various organs or diseases requires further examination.

THE NEED FOR DEVELOPING EXERCISE MIMETICS FROM NATURAL PRODUCTS

Exercise mimetics should have physiological effects in various tissues or organs in order to mimic the pleiotropic effects of physical exercise. Modulating the activity or expression of a single gene may not be sufficient to generate the multiple effects observed in exercise. Also, as physical exercise induces broad-ranging effects on various types of cells, tissues, and organs, it is highly unlikely that a single pharmacological agent can mimic the complex and wide-ranging effects. However, the combination of compounds affecting two different exercise-mediated targets has been shown to elicit synergistic effects in terms of mimicking the response to exercise (50). Hence, multi-targeting pharmacological agents have a greater potential to simulate the effect of exercise rather than single-targeting compounds. In this regard, exercise mimetics may be more effective if designed as a polypill, for polypills could target multiple pathways to closely simulate the complexity of the exercise response. Some natural bioactive compounds have been shown to display multi-targeting effects (74, 75). While compounds with less selectivity are generally not favored in the conventional drug discovery concept, certain compounds with the right combination of multi-targets may be useful in the case of exercise mimetics. In this context, natural extracts containing various compounds or multi-targeting compounds could have benefits for a potential exercise mimetic.

Further, the constant activation of metabolic pathways of by exercise mimetics can induce a chronic catabolic state, with potentially deleterious outcomes (15). It is likely that exercise mimetics would be applied for a long period for the purpose for maintaining health and preventing diseases, and since natural products are safer, they may more suitable than drugs for long-term consumption. Considering the side effects induced by the use of single-targeting drugs, natural products may be preferred as exercise mimetics. Several natural products have been identi-fied to increase skeletal muscle mass, strength, and function. However, the effects on various organs and the relationship between skeletal muscle and other organs should be investigated to develop exercise mimetics. Table 1 lists natural com-pounds used as exercise mimetics base on in vivo studies. The exercise mimetic effects observed in muscle (e.g. increased skeletal muscle mass, strength, and exercise capacity) and in other tissues/organs are separately described.

CANDIDATES FOR NATURAL EXERCISE MIMETICS

Resveratrol, a stilbene-structured compound naturally occurring in plants, increased oxidative muscle fibers by regulating the AMPK-PGC-1α pathway, and enhanced grip strength, and exercise capacity in high-fat diet-induced obese mouse model (52, 76). Notably, resveratrol increased serum BDNF concentration, a myokine increased by exercise, and it is possible that the positive effects on muscle are mediated by activating AMPK as BDNF contributes to anti-atrophic effect of exercise via the AMPK-PGC-1α pathway (36, 77). Ursolic acid, a natural triterpene compound found in various fruits and vegetables, induced exercise mimetic effects in various animal models (Table 1) (78). It also increased serum irisin levels and maximal muscle strength in a clinical study, suggesting that ursolic acid may exert other health beneficial effects in humans (79). Apigenin, a natural flavone abundant in various plants such as parsley and celery, increases serum irisin and FNDC5 mRNA expression in skeletal muscle (80). Apigenin also restored isoflurane-induced BDNF suppression in aged rat hippocampus and high-fat diet-induced downregulation of AMPK phosphorylation in skeletal muscle (81, 82). These may explain some of the health benefits of apigenin including improved cognitive function, insulin resistance, and the suppression of inflammation. Daidzein, a natural isoflavone found in soybean, suppresses cisplatin-induced muscle atrophy by regulating the Glut4/AMPK/FoxO pathway (83). Since it is unknown whether daidzein regulates AMPK in other tissues, it is not clear whether the health effects on other tissues are mediated via AMPK activation of skeletal muscle although soy isoflavone increased AMPK activity in visceral fat and 3T3-L1 cells (84). Quercetin is a natural flavonoid occur-ring in vegetables, fruits, tea, and wine (85). The target of quer-cetin has not been identified in relation with exercise mimetic effects, but quercetin increases BDNF level in the rat brain, which partially recapitulates exercise effects (32, 86). Tomatidine is abundant in green tomatoes but is typically reduced by 99% following ripening to red tomato (87). The exercise target of tomatidine is unknown, but it stimulates protein synthesis by increasing mTORC1 activity in mouse skeletal muscle and improves skeletal muscle function (87). Tomatidine also attenuates inflammation and nonalcoholic fatty liver disease and extends health span (88-90). Seaweeds Codium fragile and Undaria pinnatifida extracts improve running endurance and skeletal muscle mass by upregulating PPARδ and ERRγ, AMPK and ERRγ, respectively (6, 91). γ-Oryzanol, containing a mixture of triterpene alcohols and sterol ferulates found in rice bran oil, is a well-known antioxidant used by body builders and athletes to boost strength and increase muscle gain (92). It improves muscle function by upregulating PPARδ and ERRγ activity in skeletal muscle (92). Hydrangea serrata tea has an approximately 1000-fold higher sweetness than sugar and therefore has been used as a sugar substitute by diabetic patients. It also increases exercise endurance and muscle mass by enhancing PPARδ expression in the skeletal muscle (93). All of these exercise mimetics have been reported to exhibit health benefits beyond improving muscle function, suggesting the potential for development as a natural exercise mimetic. A more comprehensive investigation is further needed to fully understand the health-promoting effect in connection with exercise.

ACKNOWLEDGEMENTS

This research was supported by a research grant from Seoul Women’s University (2020-0452) and the Yonsei University Research Fund of 2020-22-0073.

CONFLICTS OF INTEREST

The authors have no conflicting interests.

TABLE

Candidates of exercise mimetics from natural sources

Name Model Feeding period Effect on muscle Target Other physiological effects Ref
1 Resveratrol Male KM mice 21 days 400 mg/kg for 12 weeks
  • Oxidative muscle fiber↑

AMPK
  • Spatial memory↑

  • Neurogenesis↑

  • Serum BDNF↑

  • Blood glucose, body weight↓

  • Immune system↑

(52, 76, 77, 94)
High-fat diet-induced obesity model 4 g/kg of food (400 mpk) for 16 weeks
  • Grip strength↑

  • Rotarod activity↑

2 Ursolic acid High-fat diet-induced obesity model 0.14% ursolic acid for 6 weeks
  • Grip strength↑

  • Skeletal muscle mass↑

  • Running endurance in treadmill↑

  • Tumors↓

  • Fasting glucose↓

  • Anti-obesity

  • Bone formation↑

  • Memory impairment↓

  • Inflammation↓

(78, 79, 95)
Fasting (24 hr) induced muscle atrophy model 25 mg/ml ursolic acid twice injection for 24 hr
  • Skeletal muscle mass↑

10 months old male C57BL/6 200 mg/kg, twice a day for 7 days
  • Type2a, slow-twitch fiber, myoglobin ↑

AMPK
22 months old male C57BL/6 0.27% ursolic acid for 2 months
  • Grip strength↑

  • Skeletal muscle mass↑

  • Specific force↑

Korean healthy men 450 mg/day for 8 weeks
  • Maximal muscle strength

Irisin
3 Apigenin High-fat diet-induced obesity model (9 weeks) 0.1% apigenin diet for 8 weeks
  • Muscle atrophy↓

  • Running endurance in treadmill↑

AMPK
  • Cognitive function by regulating BDNF signaling ↑

  • Reverse depression by upregulating BDNF

  • Blood glucose, serum lipid, insulin resistance index↓

  • Tumor growth↓

  • Inflammation↓

(80-82, 96-99)
6 weeks old male C57BL/6 0.2, 0.4% apigenin diet for 7 weeks
  • Running endurance in treadmill↑

  • Skeletal muscle mass↑

Irisin
Sciatic nerve denervation-induced muscle loss model 1% apigenin diet for 2 weeks
  • Muscle atrophy↓

16 months old male C57BL/6 25, 50, 100 mg/kg/day for 9 months
  • Frailty index↑

  • Grip strength↑

  • Running endurance in treadmill↑

  • Muscle atrophy↓

4 Daidzein Cisplatin induced muscle atrophy model 20, 80 mg/kg daidzein for 12 days
  • Skeletal muscle mass↑

  • Grip strength↑

AMPK
  • Inflammation↓

  • Breast cancer↓

  • Plasma lipid profile↑

  • Fasting blood glucose ↓

  • Insulin resistance↑

  • Obesity↓

  • Spatial learning, memory↑

  • BDNF level↑

(83, 99-103)
8 week old female mice 0.1% daidzein for 1 week
  • Skeletal muscle mass↑

5 Quercetin High-fat diet-induced obesity model 0.05%, 0.1% quercetin for 9 weeks
  • Skeletal muscle mass↑

  • Inflammation↓

  • Insulin sensitivity↑

  • Cognitive function↑

  • BDNF expression ↑

  • Healthspan↑

  • Obesity↓

(85, 86, 104-112)
Dexamethasone induced muscle atrophy model 0.15, 0.45% quercetin glycoside in drinking water for 7 days
  • Skeletal muscle mass↑

24 week old male C57BL/6 mice 1.5, 3.0 g/L quercetin glucoside in drinking water for 24 weeks
  • Grip strength↑

  • Rotarod time↑

  • Skeletal muscle mass↑

8 week old male ICR mice 12.5, 24 mg/kg for 7 days
  • Running endurance in treadmill↑

  • Voluntary wheel running ↑

26 male badminton players 1000 mg per day for 2 months
  • Endurance exercise performance↑

6 Tomatidine 7 week old male C57BL/6 0.05% tomatidine for 5 weeks
  • Skeletal muscle mass↑

  • Specific force↑

  • Grip strength↑

  • Inflammation↓

  • Nonalcoholic fatty liver disease↓

  • Lifespan, healthspan↑

(87-90)
Fasting-induced muscle atrophy model 25 mg/kg tomatidine at the beginning of the fast and 12 h later
  • Skeletal muscle mass↑

  • Specific force↑

Limb immobilization induced muscle atrophy model 25 mg/kg tomatidine every 12 h for 8 days
  • Skeletal muscle mass↑

7 Codium fragile extract 19 week old male C57BL/6 mice 0.1% Codium fragile extract diet for 10 weeks
  • Running endurance in treadmill↑

  • Skeletal muscle mass↑

PPARδ
ERRγ
  • Arterial thrombosis↓

  • Inflammatory cytokine↓

  • Anti-cancer immunity

  • Immune enhancing

  • Anti-obesity

(91, 113-117)
8 Undaria pinnatifida extract 12 week old male C57BL/6 0.25% U pinnatifida extracts for 8 weeks
  • Running endurance in treadmill↑

  • Skeletal muscle mass↑

AMPK
ERRγ
  • Growth and metastasis of cancer

  • Anti-obesity

  • Presynaptic Plasticity↑

  • Recover immunity

  • Insulin resistance↓

  • Inflammatory cytokine↓

(6, 118-122)
9 γ-Oryzanol 74 week old male C57BL/6 0.02% γ-Oryzanol diet for 13 weeks
  • Running endurance in treadmill↑

  • Grip strength↑

PPARδ
ERRγ
  • Improve cognitive function

  • Antidepressant-like effect

  • Insulin resistance↓

  • Inflammation↓

  • Anti-obesity

  • Immune response↑

(92, 123-128)
32 health young men (18-32 yr) 600 mg/day γ-Oryzanol and resistance training for 9 weeks
  • Skeletal muscle strength↑

10 Hydrangea serrata tea 12 weeks old male C57BL/6 0.25%, 0.5% H. serrata extract for 8 weeks
  • Running endurance in treadmill↑

  • Skeletal muscle mass↑

PPARδ
  • Anti-obesity

  • Inflammation↓

  • Total cholesterol and low-density lipoprotein, insulin↓

(93, 129, 130)

REFERENCES
  1. Gubert C and Hannan AJ (2021) Exercise mimetics: harnessing the therapeutic effects of physical activity. Nat Rev Drug Discov 20, 862-879
    Pubmed CrossRef
  2. Fan W and Evans RM (2017) Exercise mimetics: impact on health and performance. Cell Metab 25, 242-247
    Pubmed KoreaMed CrossRef
  3. O'Gorman DJ, Karlsson HK, McQuaid S et al (2006) Exercise training increases insulin-stimulated glucose disposal and GLUT4 (SLC2A4) protein content in patients with type 2 diabetes. Diabetologia 49, 2983-2992
    Pubmed CrossRef
  4. Ernst C, Olson AK, Pinel JP, Lam RW and Christie BR (2006) Antidepressant effects of exercise: evidence for an adult-neurogenesis hypothesis?. J Psychiatry Neurosci 31, 84-92
    Pubmed KoreaMed
  5. Wang M, Baker JS, Quan W, Shen S, Fekete G and Gu Y (2020) A preventive role of exercise across the Coronavirus 2 (SARS-CoV-2) pandemic. Front Physiol 11, 572718
    Pubmed KoreaMed CrossRef
  6. Ahn J, Ha TY, Ahn J et al (2020) Undaria pinnatifida extract feeding increases exercise endurance and skeletal muscle mass by promoting oxidative muscle remodeling in mice. FASEB J 34, 8068-8081
    Pubmed CrossRef
  7. Egan B and Zierath JR (2013) Exercise metabolism and the molecular regulation of skeletal muscle adaptation. Cell Metab 17, 162-184
    Pubmed CrossRef
  8. Fan W, Waizenegger W, Lin CS et al (2017) PPARdelta promotes running endurance by preserving glucose. Cell Metab 25, 1186-1193
    Pubmed KoreaMed CrossRef
  9. Hughes DC, Ellefsen S and Baar K (2018) Adaptations to endurance and strength training. Cold Spring Harb Perspect Med 8, a029769
    Pubmed KoreaMed CrossRef
  10. Bandy WD, Lovelace-Chandler V and McKitrick-Bandy B (1990) Adaptation of skeletal muscle to resistance training. J Orthop Sports Phys Ther 12, 248-255
    Pubmed CrossRef
  11. Warburton DE, Nicol CW and Bredin SS (2006) Health benefits of physical activity: the evidence. CMAJ 174, 801-809
    Pubmed KoreaMed CrossRef
  12. Mercken EM, Carboneau BA, Krzysik-Walker SM and de Cabo R (2012) Of mice and men: the benefits of caloric restriction, exercise, and mimetics. Ageing Res Rev 11, 390-398
    Pubmed KoreaMed CrossRef
  13. Li S and Laher I (2017) Exercise mimetics: running without a road map. Clin Pharmacol Ther 101, 188-190
    Pubmed CrossRef
  14. Carey AL and Kingwell BA (2009) Novel pharmaco-logical approaches to combat obesity and insulin resistance: targeting skeletal muscle with 'exercise mimetics'. Diabeto-logia 52, 2015-2026
    Pubmed CrossRef
  15. Hawley JA, Joyner MJ and Green DJ (2021) Mimicking exercise: what matters most and where to next?. J Physiol 599, 791-802
    Pubmed KoreaMed CrossRef
  16. Lee JH and Jun HS (2019) Role of myokines in regulating skeletal muscle mass and function. Front Physiol 10, 42
    Pubmed KoreaMed CrossRef
  17. Hartwig S, Raschke S, Knebel B et al (2014) Secretome profiling of primary human skeletal muscle cells. Biochim Biophys Acta 1844, 1011-1017
    Pubmed CrossRef
  18. Chen N, Li Q, Liu J and Jia S (2016) Irisin, an exercise-induced myokine as a metabolic regulator: an updated narrative review. Diabetes Metab Res Rev 32, 51-59
    Pubmed CrossRef
  19. Liu L, Guo J, Chen X, Tong X, Xu J and Zou J (2021) The role of irisin in exercise-mediated bone health. Front Cell Dev Biol 9, 668759
    Pubmed KoreaMed CrossRef
  20. Lira VA, Benton CR, Yan Z and Bonen A (2010) PGC-1alpha regulation by exercise training and its influences on muscle function and insulin sensitivity. Am J Physiol Endocrinol Metab 299, E145-E161
    Pubmed KoreaMed CrossRef
  21. Bostrom P, Wu J, Jedrychowski MP et al (2012) A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 481, 463-468
    Pubmed KoreaMed CrossRef
  22. Reza MM, Subramaniyam N, Sim CM et al (2017) Irisin is a pro-myogenic factor that induces skeletal muscle hypertrophy and rescues denervation-induced atrophy. Nat Commun 8, 1104
    Pubmed KoreaMed CrossRef
  23. He W, Wang P, Chen Q and Li C (2020) Exercise enhances mitochondrial fission and mitophagy to improve myopathy following critical limb ischemia in elderly mice via the PGC1a/FNDC5/irisin pathway. Skelet Muscle 10, 25
    Pubmed KoreaMed CrossRef
  24. Korta P, Pochec E and Mazur-Bialy A (2019) Irisin as a multifunctional protein: implications for health and certain diseases. Medicina (Kaunas) 55, 485
    Pubmed KoreaMed CrossRef
  25. Mazur-Bialy AI, Bilski J, Pochec E and Brzozowski T (2017) New insight into the direct anti-inflammatory activity of a myokine irisin against proinflammatory activation of adipocytes. Implication for exercise in obesity. J Physiol Pharmacol 68, 243-251
    Pubmed
  26. Otero-Diaz B, Rodriguez-Flores M, Sanchez-Munoz V et al (2018) Exercise induces white adipose tissue browning across the weight spectrum in humans. Front Physiol 9, 1781
    Pubmed KoreaMed CrossRef
  27. Castillo-Quan JI (2012) From white to brown fat through the PGC-1alpha-dependent myokine irisin: implications for diabetes and obesity. Dis Model Mech 5, 293-295
    Pubmed KoreaMed CrossRef
  28. Wrann CD, White JP, Salogiannnis J et al (2013) Exercise induces hippocampal BDNF through a PGC-1alpha/FNDC5 pathway. Cell Metab 18, 649-659
    Pubmed KoreaMed CrossRef
  29. Islam MR, Valaris S, Young MF et al (2021) Exercise hormone irisin is a critical regulator of cognitive function. Nat Metab 3, 1058-1070
    Pubmed CrossRef
  30. Liu PZ and Nusslock R (2018) Exercise-mediated neurogenesis in the Hippocampus via BDNF. Front Neurosci 12, 52
    Pubmed KoreaMed CrossRef
  31. Gomez-Pinilla F, Ying Z, Roy RR, Molteni R and Edgerton VR (2002) Voluntary exercise induces a BDNF-mediated mechanism that promotes neuroplasticity. J Neurophysiol 88, 2187-2195
    Pubmed CrossRef
  32. Soya H, Nakamura T, Deocaris CC et al (2007) BDNF induction with mild exercise in the rat hippocampus. Biochem Biophys Res Commun 358, 961-967
    Pubmed CrossRef
  33. Nilsson J, Ekblom O, Ekblom M et al (2020) Acute increases in brain-derived neurotrophic factor in plasma following physical exercise relates to subsequent learning in older adults. Sci Rep 10, 4395
    Pubmed KoreaMed CrossRef
  34. Bishop-Bailey D (2013) Mechanisms governing the health and performance benefits of exercise. Br J Pharmacol 170, 1153-1166
    Pubmed KoreaMed CrossRef
  35. Yu T, Chang Y, Gao XL, Li H and Zhao P (2017) Dynamic expression and the role of BDNF in exercise-induced skeletal muscle regeneration. Int J Sports Med 38, 959-966
    Pubmed CrossRef
  36. Zhang Z, Wang B and Fei A (2019) BDNF contributes to the skeletal muscle anti-atrophic effect of exercise training through AMPK-PGC1alpha signaling in heart failure mice. Arch Med Sci 15, 214-222
    Pubmed KoreaMed CrossRef
  37. So B, Kim HJ, Kim J and Song W (2014) Exercise-induced myokines in health and metabolic diseases. Integr Med Res 3, 172-179
    Pubmed KoreaMed CrossRef
  38. Motamedi S, Karimi I and Jafari F (2017) The interrelationship of metabolic syndrome and neurodegenerative diseases with focus on brain-derived neurotrophic factor (BDNF): kill two birds with one stone. Metab Brain Dis 32, 651-665
    Pubmed CrossRef
  39. Munoz-Canoves P, Scheele C, Pedersen BK and Serrano AL (2013) Interleukin-6 myokine signaling in skeletal muscle: a double-edged sword?. FEBS J 280, 4131-4148
    Pubmed KoreaMed CrossRef
  40. Pedersen BK, Steensberg A, Fischer C et al (2003) Searching for the exercise factor: is IL-6 a candidate?. J Muscle Res Cell Motil 24, 113-119
    Pubmed CrossRef
  41. Pedersen BK and Fischer CP (2007) Beneficial health effects of exercise--the role of IL-6 as a myokine. Trends Pharmacol Sci 28, 152-156
    Pubmed CrossRef
  42. Pedersen BK and Febbraio MA (2008) Muscle as an endocrine organ: focus on muscle-derived interleukin-6. Physiol Rev 88, 1379-1406
    Pubmed CrossRef
  43. Wedell-Neergaard AS, Lang Lehrskov L, Christensen RH et al (2019) Exercise-induced changes in visceral adipose tissue mass are regulated by IL-6 signaling: a randomized controlled trial. Cell Metab 29, 844-855
    Pubmed CrossRef
  44. Febbraio MA, Hiscock N, Sacchetti M, Fischer CP and Pedersen BK (2004) Interleukin-6 is a novel factor mediating glucose homeostasis during skeletal muscle contraction. Diabetes 53, 1643-1648
    Pubmed CrossRef
  45. Carey AL, Steinberg GR, Macaulay SL et al (2006) Interleukin-6 increases insulin-stimulated glucose disposal in humans and glucose uptake and fatty acid oxidation in vitro via AMP-activated protein kinase. Diabetes 55, 2688-2697
    Pubmed CrossRef
  46. Bowen KK, Dempsey RJ and Vemuganti R (2011) Adult interleukin-6 knockout mice show compromised neurogenesis. Neuroreport 22, 126-130
    Pubmed CrossRef
  47. Fan W, Atkins AR, Yu RT, Downes M and Evans RM (2013) Road to exercise mimetics: targeting nuclear receptors in skeletal muscle. J Mol Endocrinol 51, T87-T100
    Pubmed KoreaMed CrossRef
  48. Richter EA and Ruderman NB (2009) AMPK and the biochemistry of exercise: implications for human health and disease. Biochem J 418, 261-275
    Pubmed KoreaMed CrossRef
  49. Fujii N, Seifert MM, Kane EM et al (2007) Role of AMP-activated protein kinase in exercise capacity, whole body glucose homeostasis, and glucose transport in skeletal muscle -insight from analysis of a transgenic mouse model. Diabetes Res Clin Pract 77 Suppl 1, S92-S98
    Pubmed CrossRef
  50. Narkar VA, Downes M, Yu RT et al (2008) AMPK and PPARdelta agonists are exercise mimetics. Cell 134, 405-415
    Pubmed KoreaMed CrossRef
  51. Winder WW, Holmes BF, Rubink DS, Jensen EB, Chen M and Holloszy JO (2000) Activation of AMP-activated protein kinase increases mitochondrial enzymes in skeletal muscle. J Appl Physiol (1985) 88, 2219-2226
    Pubmed CrossRef
  52. Guerrieri D, Moon HY and van Praag H (2017) Exercise in a pill: the latest on exercise-mimetics. Brain Plast 2, 153-169
    Pubmed KoreaMed CrossRef
  53. Buhl ES, Jessen N, Pold R et al (2002) Long-term AICAR administration reduces metabolic disturbances and lowers blood pressure in rats displaying features of the insulin resistance syndrome. Diabetes 51, 2199-2206
    Pubmed CrossRef
  54. Kirchner J, Brune B and Namgaladze D (2018) AICAR inhibits NFkappaB DNA binding independently of AMPK to attenuate LPS-triggered inflammatory responses in human macrophages. Sci Rep 8, 7801
    Pubmed KoreaMed CrossRef
  55. Kobilo T, Yuan C and van Praag H (2011) Endurance factors improve hippocampal neurogenesis and spatial memory in mice. Learn Mem 18, 103-107
    Pubmed KoreaMed CrossRef
  56. Kobilo T, Guerrieri D, Zhang Y, Collica SC, Becker KG and van Praag H (2014) AMPK agonist AICAR improves cognition and motor coordination in young and aged mice. Learn Mem 21, 119-126
    Pubmed KoreaMed CrossRef
  57. Guerrieri D and van Praag H (2015) Exercise-mimetic AICAR transiently benefits brain function. Oncotarget 6, 18293-18313
    Pubmed KoreaMed CrossRef
  58. Zizola C, Kennel PJ, Akashi H et al (2015) Activation of PPARdelta signaling improves skeletal muscle oxidative metabolism and endurance function in an animal model of ischemic left ventricular dysfunction. Am J Physiol Heart Circ Physiol 308, H1078-H1085
    Pubmed KoreaMed CrossRef
  59. Braissant O, Foufelle F, Scotto C, Dauca M and Wahli W (1996) Differential expression of peroxisome proliferator-activated receptors (PPARs): tissue distribution of PPAR-alpha, -beta, and -gamma in the adult rat. Endocrinology 137, 354-366
    Pubmed CrossRef
  60. Greene NP, Fluckey JD, Lambert BS, Greene ES, Riechman SE and Crouse SF (2012) Regulators of blood lipids and lipoproteins? PPARdelta and AMPK, induced by exercise, are correlated with lipids and lipoproteins in overweight/obese men and women. Am J Physiol Endocrinol Metab 303, E1212-E1221
    Pubmed CrossRef
  61. Reilly SM and Lee CH (2008) PPAR delta as a therapeutic target in metabolic disease. FEBS Lett 582, 26-31
    Pubmed KoreaMed CrossRef
  62. Tanaka T, Yamamoto J, Iwasaki S et al (2003) Activation of peroxisome proliferator-activated receptor delta induces fatty acid beta-oxidation in skeletal muscle and attenuates metabolic syndrome. Proc Natl Acad Sci U S A 100, 15924-15929
    Pubmed KoreaMed CrossRef
  63. Oliver WR Jr, Shenk JL, Snaith MR et al (2001) A selective peroxisome proliferator-activated receptor delta agonist promotes reverse cholesterol transport. Proc Natl Acad Sci U S A 98, 5306-5311
    Pubmed KoreaMed CrossRef
  64. Cheng L, Ding G, Qin Q et al (2004) Cardiomyocyte-restricted peroxisome proliferator-activated receptor-delta deletion perturbs myocardial fatty acid oxidation and leads to cardiomyopathy. Nat Med 10, 1245-1250
    Pubmed CrossRef
  65. Badin PM, Vila IK, Sopariwala DH et al (2016) Exercise-like effects by Estrogen-related receptor-gamma in muscle do not prevent insulin resistance in db/db mice. Sci Rep 6, 26442
    Pubmed KoreaMed CrossRef
  66. Matsakas A, Macharia R, Otto A et al (2012) Exercise training attenuates the hypermuscular phenotype and restores skeletal muscle function in the myostatin null mouse. Exp Physiol 97, 125-140
    Pubmed CrossRef
  67. Fan W, He N, Lin CS et al (2018) ERRgamma promotes angiogenesis, mitochondrial biogenesis, and oxidative remodeling in PGC1alpha/beta-deficient muscle. Cell Rep 22, 2521-2529
    Pubmed KoreaMed CrossRef
  68. Narkar VA, Fan W, Downes M et al (2011) Exercise and PGC-1alpha-independent synchronization of type I muscle metabolism and vasculature by ERRgamma. Cell Metab 13, 283-293
    Pubmed KoreaMed CrossRef
  69. Rangwala SM, Wang X, Calvo JA et al (2010) Estrogen-related receptor gamma is a key regulator of muscle mitochondrial activity and oxidative capacity. J Biol Chem 285, 22619-22629
    Pubmed KoreaMed CrossRef
  70. Matsakas A, Yadav V, Lorca S and Narkar V (2013) Muscle ERRgamma mitigates Duchenne muscular dystrophy via metabolic and angiogenic reprogramming. FASEB J 27, 4004-4016
    Pubmed CrossRef
  71. Kim DK, Kim JR, Koh M et al (2011) Estrogen-related receptor gamma (ERRgamma) is a novel transcriptional regulator of phosphatidic acid phosphatase, LIPIN1, and inhibits hepatic insulin signaling. J Biol Chem 286, 38035-38042
    Pubmed KoreaMed CrossRef
  72. Kim DK, Kim YH, Jang HH et al (2013) Estrogen-related receptor gamma controls hepatic CB1 receptor-mediated CYP2E1 expression and oxidative liver injury by alcohol. Gut 62, 1044-1054
    Pubmed KoreaMed CrossRef
  73. Kim DK, Jeong JH, Lee JM et al (2014) Inverse agonist of estrogen-related receptor gamma controls Salmonella typhimurium infection by modulating host iron homeostasis. Nat Med 20, 419-424
    Pubmed CrossRef
  74. Byun S, Lim S, Mun JY et al (2015) Identification of a dual inhibitor of janus kinase 2 (JAK2) and p70 ribosomal S6 kinase1 (S6K1) pathways. J Biol Chem 290, 23553-23562
    Pubmed KoreaMed CrossRef
  75. Shin EJ, Lee JS, Hong S, Lim TG and Byun S (2019) Quercetin directly targets JAK2 and PKCdelta and prevents UV-induced photoaging in human skin. Int J Mol Sci 20, 5262
    Pubmed KoreaMed CrossRef
  76. Jiang Q, Cheng X, Cui Y et al (2019) Resveratrol regulates skeletal muscle fibers switching through the AdipoR1-AMPK-PGC-1alpha pathway. Food Funct 10, 3334-3343
    Pubmed CrossRef
  77. Wicinski M, Malinowski B, Weclewicz MM, Grzesk E and Grzesk G (2017) Resveratrol increases serum BDNF concentrations and reduces vascular smooth muscle cells contractility via a NOS-3-independent mechanism. Biomed Res Int 2017, 9202954
    Pubmed KoreaMed CrossRef
  78. Seo DY, Lee SR, Heo JW et al (2018) Ursolic acid in health and disease. Korean J Physiol Pharmacol 22, 235-248
    Pubmed KoreaMed CrossRef
  79. Bang HS, Seo DY, Chung YM et al (2014) Ursolic Acid-induced elevation of serum irisin augments muscle strength during resistance training in men. Korean J Physiol Pharmacol 18, 441-446
    Pubmed KoreaMed CrossRef
  80. Jang YJ, Son HJ, Choi YM, Ahn J, Jung CH and Ha TY (2017) Apigenin enhances skeletal muscle hypertrophy and myoblast differentiation by regulating Prmt7. Oncotarget 8, 78300-78311
    Pubmed KoreaMed CrossRef
  81. Chen L, Xie W, Xie W, Zhuang W, Jiang C and Liu N (2017) Apigenin attenuates isoflurane-induced cognitive dysfunction via epigenetic regulation and neuroinflammation in aged rats. Arch Gerontol Geriatr 73, 29-36
    Pubmed CrossRef
  82. Choi WH, Son HJ, Jang YJ, Ahn J, Jung CH and Ha TY (2017) Apigenin ameliorates the obesity-induced skeletal muscle atrophy by attenuating mitochondrial dysfunction in the muscle of obese mice. Mol Nutr Food Res 61, 1700218
    Pubmed CrossRef
  83. Zhang H, Chi M, Chen L et al (2021) Daidzein alleviates cisplatin-induced muscle atrophy by regulating Glut4/AMPK/FoxO pathway. Phytother Res 35, 4363-4376
    Pubmed CrossRef
  84. Tan J, Huang C, Luo Q et al (2019) Soy isoflavones ameliorate fatty acid metabolism of visceral adipose tissue by increasing the AMPK activity in male rats with diet-induced obesity (DIO). Molecules 24, 2809
    Pubmed KoreaMed CrossRef
  85. Zhao Y, Chen B, Shen J et al (2017) The beneficial effects of quercetin, curcumin, and resveratrol in obesity. Oxid Med Cell Longev 2017, 1459497
    Pubmed KoreaMed CrossRef
  86. Rahvar M, Owji AA and Mashayekhi FJ (2018) Effect of quercetin on the brain-derived neurotrophic factor gene expression in the rat brain. Bratisl Lek Listy 119, 28-31
    Pubmed CrossRef
  87. Dyle MC, Ebert SM, Cook DP et al (2014) Systems-based discovery of tomatidine as a natural small molecule inhibitor of skeletal muscle atrophy. J Biol Chem 289, 14913-14924
    Pubmed KoreaMed CrossRef
  88. Kuo CY, Huang WC, Liou CJ, Chen LC, Shen JJ and Kuo ML (2017) Tomatidine attenuates airway hyperresponsiveness and inflammation by suppressing Th2 cytokines in a mouse model of asthma. Mediators Inflamm 2017, 5261803
    Pubmed KoreaMed CrossRef
  89. Wu SJ, Huang WC, Yu MC et al (2021) Tomatidine ameliorates obesity-induced nonalcoholic fatty liver disease in mice. J Nutr Biochem 91, 108602
    Pubmed CrossRef
  90. Fang EF, Waltz TB, Kassahun H et al (2017) Tomatidine enhances lifespan and healthspan in C. elegans through mitophagy induction via the SKN-1/Nrf2 pathway. Sci Rep 7, 46208
    Pubmed KoreaMed CrossRef
  91. Ahn J, Kim MJ, Yoo A et al (2021) Identifying Codium fragile extract components and their effects on muscle weight and exercise endurance. Food Chem 353, 129463
    Pubmed CrossRef
  92. Ahn J, Son HJ, Seo HD et al (2021) Gamma-oryzanol improves exercise endurance and muscle strength by upregulating PPARdelta and ERRgamma activity in aged mice. Mol Nutr Food Res 65, e2000652
    Pubmed CrossRef
  93. Jang YJ, Ahn J, Son HJ, Jung CH, Ahn J and Ha TY (2019) Hydrangea serrata tea enhances running endurance and skeletal muscle mass. Mol Nutr Food Res 63, e1801149
    Pubmed CrossRef
  94. Malaguarnera L (2019) Influence of resveratrol on the immune response. Nutrients 11, 946
    Pubmed KoreaMed CrossRef
  95. Kunkel SD, Suneja M, Ebert SM et al (2011) mRNA expression signatures of human skeletal muscle atrophy identify a natural compound that increases muscle mass. Cell Metab 13, 627-638
    Pubmed KoreaMed CrossRef
  96. Choi WH, Jang YJ, Son HJ, Ahn J, Jung CH and Ha TY (2018) Apigenin inhibits sciatic nerve denervation-induced muscle atrophy. Muscle Nerve 58, 314-318
    Pubmed CrossRef
  97. Wang D, Yang Y, Zou X, Zhang J, Zheng Z and Wang Z (2020) Antioxidant apigenin relieves age-related muscle atrophy by inhibiting oxidative stress and hyperactive mitophagy and apoptosis in skeletal muscle of mice. J Gerontol A Biol Sci Med Sci 75, 2081-2088
    Pubmed KoreaMed CrossRef
  98. Salehi B, Venditti A, Sharifi-Rad M et al (2019) The therapeutic potential of apigenin. Int J Mol Sci 20, 1305
    Pubmed KoreaMed CrossRef
  99. Ahmed T, Javed S, Tariq A et al (2017) Daidzein and its effects on brain. Curr Med Chem 24, 365-375
    Pubmed CrossRef
  100. Ogawa M, Kitano T, Kawata N et al (2017) Daidzein down-regulates ubiquitin-specific protease 19 expression through estrogen receptor beta and increases skeletal muscle mass in young female mice. J Nutr Biochem 49, 63-70
    Pubmed CrossRef
  101. Sakamoto Y, Naka A, Ohara N, Kondo K and Iida K (2014) Daidzein regulates proinflammatory adipokines thereby improving obesity-related inflammation through PPARgamma. Mol Nutr Food Res 58, 718-726
    Pubmed CrossRef
  102. Liu X, Suzuki N, Santosh Laxmi YR, Okamoto Y and Shibutani S (2012) Anti-breast cancer potential of daidzein in rodents. Life Sci 91, 415-419
    Pubmed CrossRef
  103. Das D, Sarkar S, Bordoloi J, Wann SB, Kalita J and Manna P (2018) Daidzein, its effects on impaired glucose and lipid metabolism and vascular inflammation associated with type 2 diabetes. Biofactors 44, 407-417
    Pubmed CrossRef
  104. Le NH, Kim CS, Park T et al (2014) Quercetin protects against obesity-induced skeletal muscle inflammation and atrophy. Mediators Inflamm 2014, 834294
    Pubmed KoreaMed CrossRef
  105. Anhe GF, Okamoto MM, Kinote A et al (2012) Quercetin decreases inflammatory response and increases insulin action in skeletal muscle of ob/ob mice and in L6 myotubes. Eur J Pharmacol 689, 285-293
    Pubmed CrossRef
  106. Otsuka Y, Egawa K, Kanzaki N, Izumo T, Rogi T and Shibata H (2019) Quercetin glycosides prevent dexamethasone-induced muscle atrophy in mice. Biochem Biophys Rep 18, 100618
    Pubmed KoreaMed CrossRef
  107. Ghafouri-Fard S, Shabestari FA, Vaezi S et al (2021) Emerging impact of quercetin in the treatment of prostate cancer. Biomed Pharmacother 138, 111548
    Pubmed CrossRef
  108. Geng L, Liu Z, Wang S et al (2019) Low-dose quercetin positively regulates mouse healthspan. Protein Cell 10, 770-775
    Pubmed KoreaMed CrossRef
  109. Kanzaki N, Takemoto D, Ono Y et al (2019) Quercetin glycosides improve motor perfor-mance and muscle weight in adult mice. J Nutr Food Sci 9, 760
    CrossRef
  110. Davis JM, Murphy EA, Carmichael MD and Davis B (2009) Quercetin increases brain and muscle mitochondrial biogenesis and exercise tolerance. Am J Physiol Regul Integr Comp Physiol 296, R1071-R1077
    Pubmed CrossRef
  111. Daneshvar P, Hariri M, Ghiasvand R et al (2013) Effect of eight weeks of quercetin supplementation on exercise performance, muscle damage and body muscle in male badminton players. Int J Prev Med 4, S53-S57
    Pubmed KoreaMed
  112. Dajas F, Abin-Carriquiry JA, Arredondo F et al (2015) Quercetin in brain diseases: Potential and limits. Neuro-chem Int 89, 140-148
    Pubmed CrossRef
  113. Kim TI, Kim YJ and Kim K (2021) Extract of seaweed Codium fragile inhibits integrin alphaIIbbeta3-induced outside-in signaling and arterial thrombosis. Front Pharmacol 12, 685948
    Pubmed KoreaMed CrossRef
  114. Yang Y, Lim J, Li C, Lee S and Hong S (2021) Effects of sulfated polysaccharides isolated from Codium fragile on inflammatory cytokine gene expression and Edwardsiella tarda infection in rockfish, Sebastes schlegelii. Fish Shellfish Immunol 112, 125-134
    Pubmed CrossRef
  115. Park HB, Hwang J, Zhang W et al (2020) Polysaccharide from Codium fragile induces anti-cancer immunity by activating natural killer cells. Mar Drugs 18, 626
    Pubmed KoreaMed CrossRef
  116. Monmai C, Rod-In W, Jang AY et al (2020) Immune-enhancing effects of anionic macromolecules extracted from Codium fragile coupled with arachidonic acid in RAW264.7 cells. PLoS One 15, e0239422
    Pubmed KoreaMed CrossRef
  117. Kolsi RBA, Jardak N, Hajkacem F et al (2017) Anti-obesity effect and protection of liver-kidney functions by Codium fragile sulphated polysaccharide on high fat diet induced obese rats. Int J Biol Macromol 102, 119-129
    Pubmed CrossRef
  118. Etman SM, Mehanna RA, Bary AA, Elnaggar YSR and Abdallah OY (2021) Undaria pinnatifida fucoidan nanoparticles loaded with quinacrine attenuate growth and metastasis of pancreatic cancer. Int J Biol Macromol 170, 284-297
    Pubmed CrossRef
  119. Li L, Wang Y, Yuan J, Liu Z, Ye C and Qin S (2020) Undaria pinnatifida improves obesity-related outcomes in association with gut microbiota and metabolomics modulation in high-fat diet-fed mice. Appl Microbiol Biotechnol 104, 10217-10231
    Pubmed CrossRef
  120. Maqueshudul Haque Bhuiyan M, Mohibbullah M, Hannan MA et al (2015) Undaria pinnatifida promotes spinogenesis and synaptogenesis and potentiates functional presynaptic plasticity in hippocampal neurons. Am J Chin Med 43, 529-542
    Pubmed CrossRef
  121. Lee HH, Cho Y, Kim GH and Cho H (2020) Undaria pinnatifida fucoidan-rich extract recovers immunity of immunosuppressed mice. J Microbiol Biotechnol 30, 439-447
    Pubmed CrossRef
  122. Oh JH, Kim J and Lee Y (2016) Anti-inflammatory and anti-diabetic effects of brown seaweeds in high-fat diet-induced obese mice. Nutr Res Pract 10, 42-48
    Pubmed KoreaMed CrossRef
  123. Rungratanawanich W, Cenini G, Mastinu A et al (2019) Gamma-oryzanol improves cognitive function and modulates hippocampal proteome in mice. Nutrients 11, 753
    Pubmed KoreaMed CrossRef
  124. Araujo SM, Bortolotto VC, Poetini MR et al (2021) Gamma-oryzanol produces an antidepressant-like effect in a chronic unpredictable mild stress model of depression in Drosophila melanogaster. Stress 24, 282-293
    Pubmed CrossRef
  125. De Mattei L, Francisqueti-Ferron FV, Garcia JL et al (2021) Antioxidant and anti-inflammatory properties of gamma-oryzanol attenuates insulin resistance by increasing GLUT-4 expression in skeletal muscle of obese animals. Mol Cell Endocrinol 537, 111423
    Pubmed CrossRef
  126. Wang L, Lin Q, Yang T et al (2017) Oryzanol modifies high fat diet-induced obesity, liver gene expression profile, and inflammation response in mice. J Agric Food Chem 65, 8374-8385
    Pubmed CrossRef
  127. Shin SY, Kim HW, Jang HH et al (2017) Gamma-oryzanol-rich black rice bran extract enhances the innate immune response. J Med Food 20, 855-863
    Pubmed CrossRef
  128. Eslami S, Esa NM, Marandi SM, Ghasemi G and Eslami S (2014) Effects of gamma oryzanol supplementation on anthropometric measurements & muscular strength in healthy males following chronic resistance training. Indian J Med Res 139, 857-863
    Pubmed KoreaMed
  129. Han HS, Lee HH, Gil HS et al (2021) Standardized hot water extract from the leaves of Hydrangea serrata (Thunb.) Ser. alleviates obesity via the AMPK pathway and modulation of the gut microbiota composition in high fat diet-induced obese mice. Food Funct 12, 2672-2685
    Pubmed CrossRef
  130. Myung DB, Han HS, Shin JS et al (2019) Hydrangenol isolated from the leaves of hydrangea serrata attenuates wrinkle formation and repairs skin moisture in uvb-irradiated hairless mice. Nutrients 11, 2354
    Pubmed KoreaMed CrossRef


This Article


Cited By Articles

Author ORCID Information

Funding Information

Collections

Services
Social Network Service

e-submission

Archives