Submitted:

02 January 2024

Posted:

03 January 2024

You are already at the latest version

Abstract
Substances derived from herbals are much in demand in doping practices because of their interesting biological properties for physical exercise and sport. This review aimed to investigate anti-fatigue herbals, their properties, analysis methods and toxicity. Data indicate that phytomolecules have beneficial effects on the physiological and biochemical parameters of the body undergoing exercise. They reduce fatigue and promote recovery by improving antioxidant enzyme activities, immune responses, blood pressure, body composition and by reducing the accumulation of toxic metabolites and systemic inflammation. Thus, the use of herbal could provide relief to people who exert intense physical activities such as farmers and athletes; for this, these peoples must do careful, the use of doping herbal must be done in accordance with all the previsions which take into account the need for the use of herbal supplement, the doses. In addition, these benefits attributed to herbals should not conceal the drawbacks of using them in sport.Some phytomolecules are toxic when used in large quantities, and others, in this case alkaloids, should be avoided by athletes.
Keywords: 
;  ;  ;  ;  

1. Introduction

Today's daily life places human beings in an environment where they face many challenges to overcome if they want to demonstrate their existence. The major challenges are adapting quickly to new situations that are imposed on him and the requirement for results, proof of performance. Performance in competitive societies is the essential criterion in all spheres of activity. It is more remarkable in the fields of physical and sports activities[1]. Generally, regular physical and sports activity is a health factor both in terms of preventing the main chronic diseases, maintaining or even improving health capital and managing most chronic diseases[2,3]. The medical prescription of a physical and sports activity is an important element of public health in primary and secondary prevention of many diseases [4]. Thus it is recommended 2 hours and 30 minutes (150 minutes) of moderate intensity aerobic activity every week and muscle-strengthening activities on 2 or more days a week that work all major muscle groups [5].
However, physical activity practiced chronically or in competitive sport can develop deficiencies in physiological functions and myocardial damage [6]. It is considered as a powerful stimulus influencing the overall physiology of the human body and lead to biochemical changes in various tissues and impact gene expression [7](Światowy et al., 2021). Exercise itself can be a form of stress and cause fatigue or various types of damage to the body [8]. Also, psychological stress, mental tension and lack of sleep in their daily life could lead to fatigue (J. Chen et al., 2023).
Fatigue is defined as physical and/or mental weariness resulting from exertion, and an inability to continue exercise at the same intensity. Accumulated fatigue can affect an individual’s performance [9](Teng & Wu, 2017). Fatigue also is a common symptom both in sickness and in health and is also a common symptom in the community, with up to half of the general population complaining of fatigue [10,11](L. Hu et al., 2015; Jinchun & Jie, 2011).
The factors which induce fatigue are the consumption and depletion of energy sources, production and accumulation of metabolic products in the body, production of imbalance between the body’s oxidation and anti-oxidation system which will put the body in a state of oxidative stress and bring injury to the body by attacking large molecules and cell organs[7,12]. The exercise leads to muscular fatigue development and can increase the risk of athlete's injuries[13] (Morana & Perrey, 2009). The competition in sports is getting tougher, and the drives to achieve better result has led the athletes to indiscriminately use dietary supplements, including herbal ones [14]. Thus, it is common to observe doping behavior in the context of these physical and sporting activities. Indeed, for a very long time, man has sought to improve his sporting and intellectual performance by resorting to doping behaviors. Doping practices are defined as the use of artificial means to improve physical or intellectual performance [1]. Doping practices are based on the tools and methods of modern medicine and above all alternative medicine, which one is based on phytotherapy. Various synthetic or natural substances called performance enhancing drugs (of animal or vegetable origin) are used and these drugs are grouped into several broad categories: improving concentration and alertness; enhancing strength, power and explosiveness; improving the oxygen-carrying capacity of blood, augmenting recovery, recuperation, and reconstitution of the athlete and ameliorating inflammation and pain [5]. Herbals can improve physiological capacities by decreasing exercise-induced physical fatigue and improve [15,16,17]. That can explain the increase of use of herbal medicinal products and supplements during last decades[18]. The exercise performance can be improved by various compounds found in tropical fruits such as banana, cherry, grape, pomegranate, and watermelon. Overall, tropical fruits can aid sports performance by improving physical strength, increasing the recovery in injury, attenuate muscle soreness, and reducing fatigue [14].
The objective of this review aimed to investigate anti-fatigue plants in the world through a description of the plants, their compounds or molecules, the analysis of the methods used to study their anti-fatigue properties, the techniques and methods of evaluation of sport performance and the toxicity of anti-fatigue plants.

2. Methods

To collect scientific papers, databases as “ScienceDirect”, “PubMed” and “Google Scholar” were queried. Key words “Plants used in doping”, “Anti-fatigue activity”, “Antioxidant plants”, “immunity system and sports activity”, “sports performance and sport”, “sport activity and intellectual capacity”, “sport and neuropsychiatric disorders, “antifatigue compounds from plants”, “plants and bioenergy”, “herbal toxicity”. This review, covered the period from 1985 to 2023 and analyzed 199 documents.

3. Anti-fatigue herbals

The various social pressures encountered in work and non-work situations give rise to physical fatigue, which can impair work efficiency and quality of life [19]. Fatigue is also a common symptom of many serious illnesses [20,21]. Exercise-induced fatigue is a multi-origin physical and mental phenomenon [22]. According to Luo et al., the mechanisms involved in fatigue could be due to by reduction or polymorphism of neurotransmitters in the central nervous system, by alteration of the autonomic system in the peripheral nervous system, by accumulation of metabolites, reduction in the free energy of ATP degradation, limited availability of oxygen or substrate, by mitochondrial dysfunction, reduced glycogen storage and blood sugar levels at the metabolic level or by increased levels of pro-inflammatory cytokines in the immune system[20]. It is therefore necessary and important to develop effective and safe drugs or treatment regimens to combat fatigue and facing failure of main pharmacological therapies for fatigue with their serious side-effects, natural drugs with anti-fatigue effects offer new alternatives[23]
An increasing number of studies have shown that active components of natural foods can safely and effectively prevent and relieve fatigue, indicative of the potential of these components as anti-fatigue agents. Meanwhile, antioxidants from some natural herbals were found to possess a remarkable anti-fatigue effect [20].
Herbs have been used as medicine throughout history [24] and can be used for medicine, cooking, cosmetics and as a perfume or dye [25]. In the traditional sports, herbs are used to increase physical resistance and performance[26,27]. In their ethnobotanical surveys carried out in Burkina Faso, researchers have identified a multitude of plants species used in the optimization of exercise and these plants are known to prevent muscle and skeletal disorders, aches and pains, fatigue and mental disorders[28,29]. Athletes are among those who use herbs for their own benefit, to help them improve performance, speed recovery, maintain health and fitness during intense training periods, increase muscle mass and reduce their body fat[25]. More and more researchers devoted themselves to studying natural active ingredients in organism as the anti-fatigue drugs and scientific scrutiny with controlled clinical trials has only recently been used to study such effects [10,26].

3.1. Rhodiola rosea L. (Crassulaceae)

This herb is in traditional medicine to stimulate the nervous system, treat stress-induced fatigue and depression, improve physical performance and work productivity [30] and is considered as an adaptogen plant [31]. There are a large number of scientific publications on the clinical efficacy in terms of cognitive function and mental performance, including various symptoms of life stress, fatigue and physical performance in athletes of various preparations of this plant [30,32]. It has been demonstrated that chronic supplementation of extract provide anti-fatigue potential, improve body resistance to physical strenuous efforts by decreasing the fatigue biochemical marker levels and increasing the time to fatigue through the increase in tissue oxygenation level, liver glycogen content [31,33]. However Jówko et al. indicated that chronic ingestion does not affect physical performance, but may improve the results of certain psychomotor tests in young, healthy, physically active males[34]. A study examined the protective effects of fermented extract against fatigue and exercise stress and has demonstrated that fermented extract effectively protects against fatigue caused by strenuous exercise by increase swimming time, hepatic superoxide dismutase content, and serum lactate dehydrogenase in mice [32]. The effects of short-term has been examined by Williams et al. who indicated that supplementation may enhance explosive resistance training performance but may also impair upper body strength-endurance on blood lactate, catecholamines, and performance during repeated bench press exercise[35]. The effects of an acute dose on endurance exercise performance in eighteen female students engaged in recreational activities showed a decrease in submaximal exercise heart rate and an improvement in exercise performance, endurance exercise by reducing perceived exertion [36]. By investigating the effect of acute doses and 4 weeks, De Bock et al. showed that both doses can improve endurance capacity in young volunteer athletes[37]. A randomized, double-blind, placebo-controlled, parallel-group clinical study with an additional no-treatment group was performed by Shevtsov et al. on the one hand to measure the effect of a single dose of standardized extract on the capacity for mental work in a context of fatigue and stress and on the other hand to find the difference between two doses [38](a standard average dose in accordance with well-established medical use as a psychostimulant, the other dose being higher). This study showed a pronounced anti-fatigue effect with a dose dependent relationship in favor of the lower dose in psychometric tests but without any dose relationship in physiological tests . Another study on supplementation (Rhodiola rosea and caffeine) provided practical evidence that this combination could serve as a potential supplement for individuals, especially resistance exercise trained subjects, to enhance physical performance effectively and safely security[39].

3.2. Camellia sinensis (L.) Kuntze (Theaceae)

The tea plant Camellia sinensis is a member of the theaceae family. Green tea is provided from fresh leaves at high temperatures, thereby inactivating the oxidizing enzymes and leaving the polyphenol content intact [40]. This herbal has showed an anti-fatigue effect on forced swimming on mice and these effects were dose-dependent; when dosage was high, it normally would improve the effects much better [10,40]. Machado et al. has demonstrated that tea extract preserves neuromuscular activation and muscle damage markers in athletes subjected to cumulative fatigue and suggested that a supplementation may have potential to serve as a strategy to improve performance and recovery in conditions of cumulative exercise[41]. Jówko et al. has demonstrated that supplementation prevents oxidative stress induced by repeated cycle sprint tests in sprinters, but this does not seem to hinder training adaptation in antioxidant enzyme system. On the other hand, neither prevention of exercise-induced muscle damage, nor an improvement in sprint performance is noted after green tea extract administration[34].

3.3. Panax ginseng C.A. MEYER (Araliaceae)

Panax ginseng C. A. Meyer (ginseng), a Chinese medicinal plant of the genus Panax, boasts a rich historical record of usage that dates back to the paleolithic period, is becoming increasingly popular as a remedy for fatigue and asthenia[42,43,44,45]. This plant, a worldwide renowned adaptogen, is believed to reduce stress and augment endurance performance [46]. Panax ginseng treatment (100 mg/kg) protected muscles from eccentric exercise injuries and was effective in preserving mitochondrial membrane integrity and reduced carbonyl contents in the muscles [47]. An investigation concluded that acute supplementation of 200 mg, one hour prior to the exercise exerted significant effect on the physiological parameters measured during the endurance running by ameliorating exercise induced oxidative stress and there by augmenting exercise performance[46]. An high-dose ginsenoside complex supplementation on exercise performance in healthy adults during a 12-week supervised exercise program showed that high doses of ginseng supplementation could improve aerobic capacity during physical training [42]. The evaluation of the potential beneficial effects of the mix formulation (Antrodia camphorata and Panax ginseng) on fatigue and ergogenic functions following physiological challenge demonstrated a reduction of serum lactate, ammonia, BUN and CK activity after the swimming test, as well as increase in glucose [48]. Oh et al. has investigated the anti-fatigue activities of its compound in mice, using the weight loaded swimming and the rota-rod tests[49]. Their authors found that certain ginseng compound significantly increased the weight loaded swimming time, inhibited the weight loaded swimming-induced increase in corticosterone, lactate, lactate dehydrogenase, and creatinine levels as well as the reduction in glucose level [12,49]. Therefore, ginseng supplementation did not exert an ergogenic property on aerobic fitness enhancement in well-fit individuals [50]. Many other medicinal plants have been studied about their anti-fatigue properties. Luo et al. indicated that combined oyster peptide and ginseng extracts can be used as an effective anti-exercise-fatigue and sexual improvement supplement by decreasing the accumulation of metabolites like LA and BUN, declining the oxidative damage and the consumption of liver glycogen[12]. Also, the combination showed a synergistic effect on the improvement of mating ability after exercise fatigue in male ICR mice. The Table 1 gives an overview of some herbals whose effects following physical exercise have been studied.

4. Secondary metabolites with anti-fatigue properties

The antifatigue properties of herbals are associated with secondary metabolites that they contain. Authors which have evaluated anti-fatigue potential of herbal, has sought to know and identified the different groups of phytochemicals metabolites (polyphenols, terpenoids, saponins and alkaloids, saponins glycosides, polyacetylenes, phytosterols) which could lead to improvement of athletic performance and reducing fatigue [9,84].

4.1. Phenolics compounds

Phenolics compounds are characterized structurally by two or more hydroxyl groups attached to one or more benzene rings (Figure 1) and provide the taste and color characteristics of fruits and vegetables. They are radical scavengers and metal chelators. Polyphenols represent a natural and benign component of the human diet, possess anti-inflammatory properties and have been shown to enhance brain health and cardiovascular function via nitric oxide-mediated mechanisms [85,86]. Several studies have shown that the anti-fatigue potential of plants is attributable to their phenolic compounds, as taking polyphenol supplements reduces inflammation and oxidative damage induced by intensive exercise. [85,87]. It would appear that acute supplementation with 300 mg of polyphenols 1 to 2 hours before exercise improves exercise capacity and that 1000 mg of polyphenols per day for 3 days or more before and after exercise improves recovery from muscle damage via antioxidant, vascular and anti-inflammatory mechanisms [85]. Liudong et al. has indicated that green tea polyphenol has prolonged the swimming time of mice with less fatigue[40]. The same effect was observed with phenylethanoid-rich extract of Cistanche deserticola Y.C. Ma, by decreasing muscle damage, delaying the accumulation of lactic acid and by improving the energy storage [59]. Flavonoids compounds had also showed anti-fatigue activity [58,88,89]. Anti-fatigue activity of flavonoids of Castanea mollissima, Fagopyrum esculentum and Puerariae radix has been studied and authors has showed incrising energy reserves, oxidative stress inhibition and improvement of the metabolic control of exercise [58,88,89]. Assessment of biochemical parameters of blood related to fatigue, has demonstrated that acute or chronic resveratrol (Figure 1), a natural flavonoid polyphenol, may support the attenuation of soreness and inflammation, improve mitochondrial function in skeletal muscle and the antioxidant defense after extensive exercise and is beneficial for suppressing the aging-related decline in physical performance and for performance recovery [32,87,90,91,92]. Indicating that (-)-epigallocatechin-3-gallate (EGCG) (Figure 1) is the most abundant polyphenol in green tea that exhibits a variety of bioactivities, Teng & Wu demonstrated that EGCG significantly prolonged exhaustive swimming time and decreased the levels of BLA, SUN, SCK and MDA, which were accompanied by corresponding increases in liver and muscle glycogen contents, and SOD, CAT, and GPx activities[9]. Polyphenols supplementation may enhance exercise performance and recovery from intensive exercise with the mechanisms most likely to be related to antioxidant and vascular effects. However, this research is at an early stage and more work is required to optimize dosing strategies and to determine the specific modes, intensities and durations of exercise for which ergogenic effects may be achieved [85].

4.2. Terpenoids

Terpenoids are a class of natural products derived from mevalonic acid (MVA) which are composed of a plurality of isoprene (C5) structural units. They are a set of important in plants with diverse structures. Terpenoids have many potential applications, such as insect resistance, immunoregulation, antioxidation, antiaging, and neuroprotection [93]. Terpenoids have all been shown to improve relevant aspects of cognitive function and alertness [86]. It was suggested that the terpenoids are the active components contributing to the antioxidant effect such as inhibition of lipid peroxidation and prevention of the formation of proximities through scavenging nitric oxide and superoxide anion, and help to regulate the body testosterone level especially when the level was abnormally high by inhibiting the production of 5𝛼-reductase which was an important enzyme to activate testosterone[94]. Evaluation of the anti-fatigue activity of a pentacyclic triterpenoid extract from the bark of Bambusa tuldoides Munro in mice showed a great potential for their anti-fatigue activity [56]. Huang et al. identified ergostane-type triterpenoids, dehydrosulphurenic acid (Figure 2), 15α-acetyldehydrosulphurenic acid, lanostane-type triterpenoids in extracts of A. camphorata which has showed antifatigue activity by decreasing plasma lactate and ammonia levels and increasing blood glucose concentration and liver and muscle glycogen deposition, thereby elevating exercise performance in mice[54]. Major anthocyanins such as cyanidin-3-O-glucoside, cyanidin-3-O-rutinoside and peonidin-3-O-glucoside identified in purified anthocyanin of purple passion fruit epicarp has prolonged the weight-loaded forced swimming time, decreased lactate dehydrogenase, blood lactate and blood urea nitrogen levels, and increased the liver glycogen content[67]. Ginsenoside from panax ginseng supplementation has increased improvement of aerobic capacity [42]. Findings has suggested that 20(S)-ginsenoside Rg3, protopanaxadiol-type and protopanaxatriol-type ginsenosides had antifatigue effects, by activation of energy metabolism, elimination of metabolites accumulated, and protection from exercise induced oxidative stress[49,95] . The ginsenosides are considered as the main constituents of ginseng having different pharmacological activities such as anti-fatigue, anti-cancer, anti-aging, anti-oxidant, anti-hyperglycemic, anti-obesity [84].

4.3. Alkaloid

Alkaloids are a huge class of natural occurring organic molecules, which contain nitrogen atom or atoms in their structures. They are present not only in human daily life, in food and drinks but also as stimulant drugs, medicines, narcotics, insecticides and in many physiological activities. They showed strong biological effects on animal and human organisms even in very small doses [96]. Hsu et al. found that capsaicin (Figure 3) supplementation could improve grip strength and endurance performance[97]. Caffeine (Figure 3) has enough scientific evidence indicating an ergogenic effect [98]. Low doses of caffeine are also ergogenic in some exercise and sport situations, do not alter the peripheral wholebody responses to exercise; improve vigilance, alertness, and mood and cognitive processes during and after exercise[99]. In trained men, caffeine at 3–5 mg/kg may improve high-intensity sprint performance when ingested 30 minutes before exercise [100]. Nicotine seems to have ergogenic potential to Athletes and appear to benefit from activation of the sympathoadrenal system with increased catecholamine release and subsequent increases in muscle blood flow and lipolysis [98].

5. Primary metabolites with sports endurance property

Energy sources are essentially based on the three categories of primary metabolites that constitute the nutrients that meet the needs of basic physiological functions. Nevertheless, the anti-fatigue effects of primary metabolites have been evaluated among protein and polysaccharides extracted from plants and amino acids.

5.1. Protein

The effect on physical and athletic performance of proteins has been studied. Wang et al. have reported that natural bioactive peptides alleviate exercise-induced fatigue via a variety of complex biological reactions and molecular mechanisms that include involvement and regulation of energy metabolism, inhibition of inflammatory responses, reduction of reactive oxygen species and regulation of neurotransmitters[101]. The proteins used include aspartate, asparagine, cystine and glutamine mixture, L-Citrulline, taurine, NO3−rich supplements, dietary nucleotides [5,15,22,102,103,104]. American ginseng proteins showed anti-fatigue activity in mice, measured by physiological indices of fatigue according Qi et al.[66]. The results of aspartate and asparagine supplementation on the determinants of fatigue in Wistar rats exercised to exhaustion showed that their supplementation can increase the contribution of oxidative metabolism to energy production and delay fatigue during exercise performed above the anaerobic threshold[102]. Findings also suggested that NO3-rich supplements could be a suitable strategy to improve performance in sports modalities in which power and acceleration largely determine performance [15]. Data suggested that taurine supplementation modulates various cellular remodeling parameters after overuse-induced muscle damage, and that these positive effects may be related to its antioxidant capacity [103]. Known as a nitric oxide precursor and to enhance high-intensity exercise performance, beetroot juice supplementation has an ergogenic effect, NO3−rich supplements could be a suitable strategy to improve performance in sports modalities [15]. A study on the anti-fatigue effects of dietary nucleotides (NTs) on mice has suggested that NTs exert anti-fatigue effects, which may be attributed to the inhibition of oxidative stress and the improvement of mitochondrial function in skeletal muscles. NTs could be used as a novel natural agent for relieving exercise fatigue [104]. Ma et al. revealed that cystine and glutamine mixture supplementation attenuate fatigue during endurance exercise in healthy young men by enhancing fatty acid utilization[22]. High protein intake and exercise are two factors that preserve and enhance muscle and protein intake in the range of at least 1.2 to1.5 g/kg/day is recommended [5].

5.2. Polysaccharides

The polysaccharide extracts have been credited with multiple benefits such as improved muscle glycogen and athletic performance, increased endurance times, resistance to fatigue, decreased oxidative stress after intense exercise, detoxification of the body, immunomodulatory, hepatoprotective, antitumor, anti-aging, antioxidant and hypoglycemic activities [105,106]. Polysaccharides extracted from Portulaca oleracea L. prolonged the riding times and exhaustive swimming times of mice, decreasing blood lactic acid and serum urea nitrogen levels, as well as increasing the liver and muscle glycogen contents [17]. Results of Liu et al. provided theoretical support for the application of polysaccharides extracted from Hericium erinaceus (HEP ) in the field of sports nutrition with significant anti-fatigue activity[106]. Jo et al. indicatited that consumption of carbohydrate-rich meals before exercise resulted in less disruption of circulating numbers of leukocytes, neutrophils, and T-cell subsets, and less elevation of plasma IL-6 concentrations immediately after exercise and during the 2 h recovery period. Moon et al. revealed that large (48-g) and small (24-g) of daily isonitrogenous doses of rice and whey protein in combination with an eight-week resistance training program led to similar changes in body composition and performance outcomes such as improvements in strength and fat-free mass [107] The Table 2 and Table 3 summarizes products derived from plants whom enhance physical performance potential was studied.
Table 2. Results of studies of some Phyto molecules identified.
Table 2. Results of studies of some Phyto molecules identified.
Class Molecule Subjects Exercises Tests Doses et duration of administration Results References
steroidal glycoside
Ginsenoside sedentary Individuals Aerobic exercises and muscle strengthening. 100mg/j and 500mg/j
12 weeks
ꜛVO2max
ꜛmuscle strength
[42]
Ginsenoside-Rg1 Rats Exhaustive swimming 0.1 mg/kg bodyweight per day for 10-week. ꜛantioxidant defense system in skeletal muscle ꜜmembrane lipid peroxidation [108]
20(S)-Protopanaxadiol 20(S)-Protopanaxatriol Mice Weight-loaded wimming and rota-rod tests 5 and 10 mg/kg ꜛriding time in the rota-rod test, inhibiting corticosterone, lactate, and creatinine levels. [49]
20(S)-Ginsenoside
Rg3
Mice Forced swimming tests 10, 20 and 40 mg/kg once a day lasting for 28 days ꜛexhaustive exercise time, ꜜdecreased blood lactic acid, serum urea nitrogen levels, ꜛGLU, SOD, glutathione peroxidase and catalase in liver and muscle, ꜜMDA levels [95]
Alkaloid
Capsaicin Male ICR mice Forelimb grip strength, exhaustive swimming 4 weeks at 0, 205, 410, and 1025 mg/kg/day ꜜserum lactate, ammonia, BUN and CK levels
ꜛglucose concentration
[109]
Caffeine C + ephedrine E Athletes Cycling exercise C (5 mg / kg),
E (1 mg / kg), C+E
ꜛexercise time to exhaustion and central nervous system stimulation. [110]
Flavonoid
Eleutheroside Mice load-weighted swimming test, sleep deprivation test 500 mg/kg and 200 mg/kg Relief of physical and mental fatigue [51]
Resvératrol Mice forelimb grip strength, exhaustive swimming 25 mg/kg mice/day for 28 consecutive days Improved muscle strength and endurance
[111]
Resveratrol Male ICR mice forelimb grip strength,
exhaustive swimming time
0, 25, 50, and 125 mg/kg/day 21 days ꜛexhaustive swimming time and grip strength, CK activity and glucose levels
ꜜserum lactate, ammonia levels
[92]
(-)-Epigallocatechin-3-gallate mice forced swimming exercise 50, 100, and 200 mg/ kg by oral gavage for 28 days. ꜛexhaustive swimming time, glycogen contents, antioxydant activities.
ꜜlevels of blood lactic acid, serum urea nitrogen, serum CK and MDA
[9]
Glabridin Male mice swimming exhaustive exercise 5, 10, 20 mg/kg) for 28 consecutive days inhibited fatigue, delayed the elevation of blood lactic acid and increase storage of liver and muscle glycogen [57]
Vitamin vitamin C supplementation healthy endurance trained males single bout of endurance exercise (2.5 h cycling exercise in man) 2 weeks of supplementation with vitamin C, 1,000 mg /day ꜛantioxidant defence, modulating the leukocytosis and neutrophilia responses [56]
Amino acid Aspartate and Asparagine Wistar rats exercised to exhaustion by swimming 350 mM ASP + 400 mM ASG day–1 for 7 days ꜛexercise time
ꜜblood lactate concentration
[102]
Amino acid γ-aminobutyric acid Mice loaded-swimming test 0.15, 0.3, 0.9 g/kg GABA/animal weight, ꜛ swimming time
[112]
Amino acid L-Citrulline Athletes maximum effort test in a cycloergometer reduce the recovery heart rate and muscle soreness after 24 h. [83]
Table 2. Results of studies of some Phyto constituents identified.
Table 2. Results of studies of some Phyto constituents identified.
Substance Subjects Exercises Tests Doses Results ref
Panax quinquefolium proteins mice Forced swimming test 125, 250 and 500 mg/kg of body weight for 28 days ꜛswimming ability, ꜛGLU levels
ꜜaccumulation of BLA and SUN,
[66]
Ginseng polysaccharides mice Forced swim test 15 days ꜛanti-fatigue activity [45]
Polysaccharides from Portulaca oleracea L. mice, Rotarod test, Forced swimming test 75 /150and 300 mg/kg bw
duringg 30 days
ꜛGLU contents, ꜜblood lactic acid and serum urea nitrogen level [17]
Taraxacum officinale extract male Forced swimming test 10, 30 and 100mg/kg b.w for a period of 42 days. ꜛswimming capacity, ꜜlactate and triglyceride concentrations [11]
Extract of Rubus coreanus fruit Mice Forced swimming test (500 mg/kg/day for 4 weeks ꜛforced swimming capacity of mice ,ꜜplasma ammonia accumulation [76]
Extract of Toona sinensis Roemor Mice Swimming test 40, 80 and 160 mg/kg) for 21 days ꜛswimming time of the mice, ꜜlactic acid,ꜛliver and muscle GLU, ꜜoxidative stress. [79]
Extract of saponins of Radix notoginseng Mice Swimming test 20, 40 and 80 mg/kg body weight/day for 28 days ꜛexhaustive swimming time , ꜜlactate in the blood, ꜛtissue glycogen contents [72]
Extract of Siraitia grosvenorii Fruits Mice Swimming test 100, 200 and 400 mg/Kg bw/day for 28 days. ꜛswimming time,liver muscle glycogen , ꜜBLA, SUN [78]
Extract of Acanthopanax senticosus. Mice forced swimming test 100, 200 and 400 mg/kg ꜛtissue glycogen contents
ꜜ BLA, SUN contents
[52]
Triterpenoid-Rich Extract from Antrodia camphorata Mice Swimming test. 0, 50, and 200 mg/kg/day ꜛexhaustive swimming time, tissue GLU contents and activity of enzymes, ꜜBLA, BUN [54]
Pentacyclic triterpenoid from Bambusa tuldoides Munro Mice weight-loaded swimming test and climbing test 0.04; 0.08 and 0.25 g/kg body weight ꜛswim time, blood glucose, muscular and hepatic GLU levels, ꜜplasma lactate and ammonia levels and CK activity [56]
Antrodia camphorata and Panax ginseng extract Mice swimming test forelimb grip orally administered for 4 weeks at 0.984, 2.952 and 5.904 g/kg/day. ꜜlactate, ammonia, BUN and CK activity, ꜛexhaustive swimming time and forelimb grip strength [48]
Beetroot Juice young men Wingate and jump tests 70 mL of BJ (containing 6.4 mmol of NO3−) Ergogenic effect in a 30-s all-out Wingate test increasing Wpeak, [15]
watermelon juice seven athletes maximum effort test in a cycloergometer 500 mL of natural watermelon juice (1.17 g of L-citrulline) helped to reduce the recovery heart rate and muscle soreness after 24 h. [83]
Tartary buckwheat (Fagopyrum tataricum) Mice Exhaustive Swimming Time of Mice 60, 120 and 240 mg/kg body weight every day for 28 days ꜛexhaustive swimming time, tissue GLU contents and the activities of antioxidant enzymes, ꜜBLA, BUN [62]
Astragali Radix and Angelicae Sinensis Radix Mice weight-loaded and forced swimming 21.64 ;10.82 and 5.41 g/kg bw daily for 15 days ꜜlevels of BLA and BUN
ꜛ SOD.
[113]
Tea seed oil (Camellia oleifera Abel) Mice weight-Loaded Forced Swimming 0.5 ,1.0, 2.0 g/kg/day At 0 week, 2 weeks and 4 weeks anti-fatigue effects; improve the effects much better on BUN and hepatic GLU [10]
Eriobotrya japonica extract Mice Exhaustive Swimming 1 g kg-1 per day for 14 days Enhancing utilization of fatty acid, facilitating lipid catabolism, and
ꜛ antioxidant capability.
[61]
Irpex lacteus extract
Mice. swim test, rotating rod and running test. 0.04, 0.2 and 1.0g/kg Seven-day increased the level of super oxide dismutase reduced the level of malondialdehyde in the liver [114]
Extract of Trigonella foenum-graecum L. rat weight loaded
forced swim test
10 mg/kg of bw/per day) for a period of two weeks. Ameliorating various impairments associated with physical fatigue. [80]
Extract of Anisomeles indica (L.) Kuntze mice swimming test 125, 250; 500 mg/kg/day
for 28 days
ꜛswimming time, hepatic GLU muscle glycogen levels, ꜜtriglyceride, ammonia levels [53]
Extract of Cistanche deserticola Mice forced swimming test 0.25, 0.50, 1.00 g/kg administered orally to mice for 3 weeks. ꜛswimming capacity of mice, ꜜmuscle damage, ꜜlactic acid, ꜛenergy storage [59]
Extract of Allium sativum L. rats Endurance exercise 2.86 g/kg 30 min before every exercise for 4 weeks ꜜ levels of NO metabolites, ꜛSDH and SOD activities, [8]
γ-aminobutyric acid from Morus alba L. leaves Mice -swimming test 0.15, 0.3, 0.9 g/kg GABA/animal weight, Increase swimming time
[112]
vitamin and mineral supplement + guarana young adults ( Cognitive Demand Battery 200 mg/day, and 400 mg/day Improved cognitive performance and mental fatigue [115]
creatine + guarana athletes test of six maximal sprints creatine (1000 mg) + guarana (1500 mg) Beneficial effect on muscle power and decisional cognitive performance [116]

6. Methodology used for anti-fatigue phyto molecules identification

Bioactive compounds present in herbal material exist as multicomponent mixtures. Characterization, analysis, separation and identification of bioactive phytoconstituents that may be at the origin of the anti-fatigue potential of herbal involves approaches which the application of common phytochemical screening assays, chromatographic and spectrometrique techniques [23,117,118]. The development of applied untargeted metabolomic assays presents a new direction for the discovery of metabolite indicators for sport and exercise science[119]. The analytical techniques used in metabolomics are nuclear magnetic resonance spectroscopy (NMR) and mass spectrometry (MS) combined with chromatography [120]. Sportomics, application of metabolomics in sport to study the metabolic effects of physical exercise is carried out through the analysis of metabolites present in a biological fluid, such as saliva, blood, feces and urine. sportomics analyzes could be used to know muscle damage or fatigue, prevent and manage injuries, define the best diet and training programs to obtain the best performance from athletes, promote the choice of suitable physical [121]. The Table 4 presents some of the technique used according to the families of groups of compounds.

7. Antioxidant, immune system and sports activities

7.1. Exercise and the antioxidant system

Studies provide data on the effects of physical exercise on antioxidant systems, sometimes with contradictory results [138]. The antioxidant system groups together all the body's antioxidant defense tools, which are made up of antioxidant enzymes (superoxide dismutase, catalase and glutathione peroxidase, etc.) and non-enzymatic antioxidants (coenzyme Q10, glutathione, uric acid, lipoic acid, bilirubin, etc.). These antioxidants are molecules that reduce the rate of oxidation of a substrate and the oxidative damage induced by exercise [139]. Physical exercise is a factor that enhances the antioxidant response and promotes the activation of anabolic pathways and mitochondrial biogenesis in skeletal muscle[140]. It leads to an increase of glutathione content in skeletal muscle and in the activity of enzymes in the glutathione redox cycle [141] and to a specific adaptation of antioxidant systems in muscle tissue and fibers [142] . Although exercise leads to increased production of free radicals, Viña et al. report that there is growing evidence that reactive oxygen species (ROS) are not just toxic [143]. ROS generated during muscular activity may play a central role in the adaptive responses of muscles to exercise-induced oxidative stress [144]. They lead to an increase in antioxidant enzymes, their activity and thus increased resistance to oxidative challenges to such an extent that exercise itself could be considered an antioxidant [140,145]. These exercise-induced free radicals are involved in cell signalling and in the regulation of gene expression and activate redox-sensitive signal transduction [140,145]. Important signalling pathways that can be activated in response to ROS stimulation include NFκB, Nrf2 and MAPK [140,145]. Exercise induces activation of MAP kinases that activate the NF-κB pathway and, consequently, the expression of important enzymes associated with defence against ROS such as superoxide dismutase[146]. The exercise-induced production of ROS may also be an important signal to activate proliferator-activated receptor-γ coactivator-1α, a key player in the adaption of muscle cells to exercise peroxisome [147]. Moderate-intensity exercise increases the expression of SOD enzymes [148], the activity of xanthine oxidase and NADPH oxidase which are critical modulators of redox homeostasis [143,149,150]. Powers et al. indicates that ROS production could be a double-edged sword in those moderate levels of ROS production during exercise promote positive physiological adaptation in active skeletal muscle and high levels lead to damage to macromolecular structures[151].

7.2. Exercise and immune system

Immunity is a host defense function that aims to prevent abnormal cells such as tumor cells and foreign microorganisms from invading the body, and involves actions from several types of immune cells (T cells, macrophages, natural killer (NK) cells, neutrophils and bioactive substances (cytokines) that control these immune cells [152]. The effects of exercise have an impact on multiple aspects of the immune response, modulatory effects on immunocyte dynamics and possibly on immune function including T-cell phenotype and proliferation, antibody response to vaccination and cytokine production. These effects are mediated by diverse factors including exercise-induced release of proinflammatory cytokines, classical stress hormones, and hemodynamic effects leading to cell redistribution [153,154]. Physical training has controversial effects on the immune system, depending on the protocols adopted [148]. The impact of exercise on tissue homeostasis elicits an adaptive response that depends on the type, duration and intensity of the stimulus outcome [155,156]. The intensity, more than the duration of exercise, is responsible for the degree of increment in the number of NK cells. intense exercise of long duration declines the concentration of NK cells and NK cytolytic activity [153]. Prolonged exercise can alter T-cell, NK-cell and neutrophil function, alter lymphocyte proliferation, alter the balance of cytokines and blunt immune functions[153,156]. On the other hand, regular moderate exercise boosts neutrophil functions, including chemotaxis, phagocytosis, and oxidative burst activity, improves and strengthens immune function by reducing the production of ROS and thus attenuate the cytotoxic activity of immune cells [5,148,152,153,155,157]. Physical exercise decreases pro-inflammatory signals and promotes the activation of anabolic pathways and mitochondrial biogenesis in skeletal muscle [140]. Regular PE increases the percentage of regulatory macrophages in muscle and that IL-10 is an essential mediator in the analgesia produced by regular physical activity [158]. PE leads to the release of myokines. These molecules exert auto-, para- and/or endocrine effects and include cytokines, interleukins such as IL-6 and other peptides that are produced, expressed and released by muscle fibers and play a role in protecting against inflammation-associated diseases [145]. Skeletal muscle contraction produces IL-6, which is able to direct natural killer cells to the tumour site during exercise and therefore appears to be involved in mediating exercise-induced anti-cancer effects [159]. ). Physical exercise exerts beneficial impacts on the gut microbiota and a protective role in neuroimmunomodulator effects in part via changes in the gut microbiome [160].

8. Method and technique for evaluating sport performance

The evaluation of sports performance takes into account several contexts associated with the purposes of this evaluation. These contexts are variable and evaluated the performances either following a training program or following a supplementation to see the effects of a product or the effects of an exercise program associated with a supplementation and a better information on fatigue, physical capacity and performance characteristics takes into account measurements of body composition, metabolic and muscular capacity and exercise endurance [20,32,33,39,40,64,119]. From a practical point of view, the studies measured variables at two levels. On the one hand, the measurement of variables related to physical capacities and, on the other, the evaluation of physiological parameters. These measurements are taken before, during or after the administration of physical stress tests. The different tests above and the parameters to be assessed depend on the animal model used for the experiment (human model and animal model). In view of the anatomical and physiological similarities between humans and animals, new therapies are generally evaluated in animal models before being tested in humans [101].

8.1. Measurement of variables related to physical capacities

8.1.1. Non-human Animal model

-
Forced swim
Forced swim test or exhaustive swimming test involves using mice or rats allow to know the swimming endurance capacity, the most widely used comprehensive swim test to show an animal's ability to withstand exercise-induced fatigue. It was commonly accepted that swimming was an experimental exercise model [70]. Swimming time or time to exhaustion is measured at the time of exhaustion when the nose of the mouse was sinking below the water’s [101,102,118]. The forced swim test can also be discontinued after some time (normally 20-30 min), which could reflect changes in biochemical indices in mice/rats after exercise under similar conditions. intensity and same duration[161].
-
Rotarod test
The rotarod test is designed to screen the neurologic effects of drugs, to analyze motor behavior, to measure fatigue levels and physical performance and also becomes a useful method to evaluate motor function and was carried out to test drugs that interfere with motor coordination. The general procedures of rotarod test as follows: (i) Before the test, all mice are trained for a few days to adapt to the rotarod instrument (ii) On the test day, the mice are placed on the rotating lane of the rotarod instrument and the test starts. The time of the mice staying on the stick is recorded to evaluate the fatigue state. The difference in fall time observed between the negative control group and the group treated with the extract is an indication of muscle relaxant activity [101,118].
-
Treadmill test
This test assesses cardiac capacity. Generally, the animal undergoing running training is placed on the track and the exercise begins. When the mice are unable to keep up with the speed of the treadmill, this indicates the onset of fatigue. The running mileage is recorded [101,118].
-
Tail suspension test
The tail suspension test is another widely used method for assessing anti-fatigue activity in mice/rats. It is a rapid behavioral test that assesses the psychotropic effects of drug substances such as antidepressants and sedatives. The test involves suspending the mouse by the tail for a short period of time, and the animal's behavior during the session (which typically lasts 6 minutes) is recorded using a video recording device; the time of immobility during the session is determined by analyzing the video files [101,107].
-
Forelimb Grip Strength Test
This test is used to assess maximum strength and neuromuscular function in mice and rats [39,162].
-
Open field test
The open field is an unconditioned anxiety test to record general motor activity, locomotion and locomotion speed in mice[162,163].
-
Hole-board test
This test has been described for assessing specific components of mouse behaviour, such as curiosity or exploration and sedative effects (Garige et al., 2016)

8.1.2. Human Animal model test

The multiple and are a function of the parameters or physical qualities to be assessed (strength, flexibility, agility, balance, muscular strength and endurance, coordination, speed). Muscular strength can be assessed by the Grip Strength Test, the Sargent Test, the Trunk Strength Test (sit-ups) and the Wingate Test. To assess endurance and cardiorespiratory capacity, the 6-minute walk test, the Cooper test or the bicycle ergometer test can be used [39,107].

8.2. Evaluation of biochemical parameters

The most integrative approach of oxidative stress multi-marker panels in response to physical activity instead of selecting one preferential biomarker to quantify physical activity-induced oxidative stress [164]. Theses mesearements are made from preparation taken from saliva, blood, urine samples, organ tissue samples or the breath [162,163,165]. Thus, in several studies have determinate biochimistries parameters of physical performance or antifatigue effects by measuring of muscles damages by determination creatine kinase, oxidative stress biomarkers (lipid hydroperoxides, total antioxidant capacity and superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), malondialdehyde (MDA), serum blood urea nitrogen (BUN), lactate dehydrogenase (LDH), and lactic acid (LA), thiobarbituric acid–reacting substances (TBARS), Lactic acid (LD), blood urea nitrogen (BUN), liver glycogen (LG), malonaldehyde (MDA), glutathione peroxidase (GSH-Px), superoxide dismutase (SOD) and catalase (CAT) [32,34,163,165], by estimated total protein[1], skeletal muscle glycogen content, hepatic glycogen in the liver, malonylCoA content, oxygen consumption and respiratory exchange [32,90].

9. Toxicity of doping molecules and herbal

Doping molecules originate from synthetic products or plants. If natural doping products derived from plants are considered safe alternatives to synthetic doping products and seem to offer advantages with fewer inconveniences for physical activity and sport, it should be noted that whatever the source of the products can present toxic effects [86,166]. There is no guarantee that herbal supplements are safe for anyone and this has not helped clear up the confusion surrounding the use of herbs in sports in particular [18]. The results have shown that the extract rich in polysaccharides of corn silk a LD50 value in mice much more than 20g/ kg [167]. Lynch et al. indicated that there is a rare case of death associated with ingredients based on plants or food supplements and the cases of death encountered are linked to inappropriate use[168]. The presence of the toxicants shows that these leaves should be properly processed before consuming them (Smith, 2009). Thus, studies have evaluated the toxicity of certain antifatigue plant showed that leaves and stems of Rubus coreanus induced no toxicological changes in rats at a single dose of 2500 mg/kg/day and no adverse effects at or at repeated oral doses of 625, 1250 and 2500 mg/kg/day [169]. Moringa oleifera could have severe toxic and abortifacient effects when taken in large amounts; although signs of acute toxicity were observed with lethal dose of more than 3,873 mg kg−1 with no adverse effect observed at concentrations lower than 3,000 mg kg−1. The authors indicate that Moringa oleifera leaf extract has no toxic effects when administered in concentrated doses over a short period[170,171,172]. Hydro-alcoholic extract of Portulaca oleracea at the dose level of 500 mg/kg can be used for further pharmacological activity and at the dose level of 400mg/kg have significant hepato protection [173]. The toxicity of compounds (phenolics, terpenoids and alkaloids) of certain anti-fatigue plants was also evaluated.The flavonoid fraction of the leaves of the Anisomelles Indica Kuntze seems to be safe for oral administration and has been shown that it is non-toxic following acute exhibitions with mice under experience conditions where the maximum tolerated dose is assessed at more than 5000 mg / kg B.W [63,174]. Sergi, considered that EGCG is toxic in childhood because of the potential adverse effect on immature or fast-developing liver cells [175]. It was indicated that higher bolus doses of EGCG are hepatotoxic to mice associated to oxidative stress including increased hepatic lipid peroxidation, plasma 8-isoprostane, hepatic metallothionein and γ-histone 2AX protein expression [176]. Dietary EGCG supplementation exhibits antihypertensive and nutrigenomics effects through activation of intrarenal Ace and Agtr2 and suppression of Ren mediators, while a high dose of EGCG induced liver damage in spontaneously hypertensive rats. the no-observed-adverse-effect level of EGCG was established at 250 mg/kg b.w.[177]. Isbrucker et al. concluded that EGCG is non-genotoxic, even when administered to animals at doses which are significantly higher than those intended for humans[178]. The mean oral lethal dose (LD50) of 20(S)-ginsenoside Rg3 is above 1600 mg/kg and 800 mg/kg in animal in acute toxicity and the no-observed-adverse-effect level was 180 mg/kg (C. Li et al., 2020), more than 1,600 mg/kg/day subchronic toxicity (Shin et al., 2014). [179] draw attention to both the harmful and protective effects of certain toxic terpenoids identified plants (cicutoxin, atractyloside, daphnetoxin, digoxin and gibberellic acid). Authors suggest that molecule branching, the presence of heteroatoms and electronegativity play a dominant role in terpenoid toxicity, citing potentially toxic terpenoids such as β-citronellol, (E,E)-farnesol, (S)-citronellal, (Z))-nerolidol geranic acid, linalool, citral, geranic acid and (±)-α-terpinyl acetate found in plants [180]. Most alkaloids are inappropriate in a sports context due to their toxicity and legal status [86]. Numerous reports acid nephrotoxic alkaloids derived from plants are often implicated in kidney damage[166], cardiovascular dysfunction[181], abdominal pain, genotoxicity, neurotoxicity, tumorigenicity, acute toxicity, nausea, chronic toxicity [96]. For Meredith et al., a nontrivial proportion of caffeine users develops clinically meaningful features of caffeine dependence, including a persistent desire or unsuccessful efforts to cut down or control caffeine use, continued use despite harm, and a characteristic withdrawal syndrome[182].

10. Conclusions

The aim of this review was to study the plants used in physical activity and sport, their anti-fatigue potential, an analysis of the methods used to study their anti-fatigue and phytochemical properties, their toxicity and the techniques and methods used to assess physical performance. It is clear that the value of plants in physical exercise and sport has been the subject of numerous scientific studies for a very long time, although the approaches used have evolved considerably. Medicinal plants represent a potential reservoir of subjects for applied research into physical activity and sport, due to the impact of the consumption of these plants by the sporting world on health and on the physiological and biochemical parameters of physical and mental performance. The data indicate that herbals can improve glycogen storage, antioxidant enzyme activities and the regulation of immune responses, and reduce the accumulation of toxic metabolites. Phyto molecules improve plasma lipid concentrations, blood pressure, body composition, antioxidant capacity, systemic inflammation and glycogen storage, and control blood sugar levels. The data indicate that moderate exercise is considered as a booster of the antioxidant and immune systems like antioxidant herbal; Therefore, an alternative therapy strategy based on a combination of adapted physical exercise ant herbal could be promising for treating diseases. The data also provides evidence that plants help to promote recovery after physical effort and reduce fatigue. Thus, the use of herbal could provide relief to people who exert intense physical activities such as farmers and athletes; for this, these peoples must do careful, the use of doping herbal must be done in accordance with all the previsions which take into account the need for the use of herbal supplement, the doses. In addition, these benefits attributed to herbals should not conceal the drawbacks of using them in sport. From a toxicological point of view, certain phyto molecules derived from these plants are toxic when used in large quantities. The use of certain plants with high levels of alkaloids should be avoided by sportsmen and women, as the majority of alkaloids are prohibited substances. The plants whose anti-fatigue properties have been studied are mainly derived from traditional medicine in Asian countries where this alternative medicine is highly developed. Few similar studies have been carried out on plants from regions of sub-Saharan Africa, where the majority of the population uses medicinal plants. This increases the vulnerability of these populations to the dangers of the use of doping plants for their health, and in the practice of sport, to doping. These observations demonstrate the need for research into medicinal plants used for doping purposes south of the Sahara.

References

  1. Yesalis CE, Bahrke MS. Histoire du dopage dans le sport. 2002;24.
  2. Bouaziz W, Vogel T, Schmitt E, Kaltenbach G, Geny B, Lang PO. Bénéfices de l’activité physique en endurance chez les seniors âgés de 70 ans ou plus : une revue systématique. La Presse Médicale. 2017 Sep;46(9):794–807.
  3. Law J, Pennington CG. Physical Activity for Individuals with Cerebral Palsy. Int J Phys Educ Fit Sports. 2021 Jun 21;73–9. [CrossRef]
  4. Cazassus N, Cugy E. Impact de l’activité physique sur la fatigue. 2019 [cited 2023 Jul 27]; Available from: https://doi.org/10.13140/RG.2.2.33549.28642.
  5. Bagchi D, Nair S, Sen CK, editors. Nutrition and enhanced sports performance: muscle building, endurance, and strength. Amsterdam ; Boston: Elsevier/AP, Academic Press is an imprint of Elsevier; 2013. 540 p.
  6. Barnard N, Goldman D, Loomis J, Kahleova H, Levin S, Neabore S, Batts T. Plant-Based Diets for Cardiovascular Safety and Performance in Endurance Sports. Nutrients. 2019 Jan 10;11(1):130. [CrossRef]
  7. Światowy WJ, Drzewiecka H, Kliber M, Sąsiadek M, Karpiński P, Pławski A, Jagodziński PP. Physical Activity and DNA Methylation in Humans. IJMS. 2021 Nov 30;22(23):12989. [CrossRef]
  8. Morihara N, Ushijima M, Kashimoto N, Sumioka I, Nishihama T, Hayama M, Takeda H. Aged Garlic Extract Ameliorates Physical Fatigue. Biological & Pharmaceutical Bulletin. 2006;29(5):962–6. [CrossRef]
  9. Teng Y song, Wu D. Anti-fatigue effect of green tea polyphenols (-)-Epigallocatechin-3-Gallate (EGCG). Phcog Mag. 2017;13(50):326.
  10. Hu L, Fang X, Du M, Zhang J. Anti-Fatigue Effect of Blended <i>Camellia oleifera Abel</i> Tea Oil and Ge-132 in Mice. FNS. 2015;06(15):1479–87.
  11. Jinchun Z, Jie C. The effects of taraxacum officinale extracts (toe) supplementation on physical fatigue in mice. Afr J Trad Compl Alt Med [Internet]. 2011 Jan 17 [cited 2023 Jun 29];8(2). Available from: http://www.ajol.info/index.php/ajtcam/article/view/63198.
  12. Luo X, Liu W, Zhong H, Yan Y, Feng F, Zhao M. Synergistic effect of combined oyster peptide and ginseng extracts on anti-exercise-fatigue and promotion of sexual interest activity in male ICR mice. Journal of Functional Foods. 2021 Nov;86:104700. [CrossRef]
  13. Morana C, Perrey S. ÉVALUATION DE LA FATIGUE MUSCULAIRE. 2009.
  14. Chabib L, Trianloka AMB, Hidayat AUMJ, Awaluddin R, Firmansyah F. Potential Tropical Fruits to Aid Sports Performance and its Prospect to be Developed into Nano supplement. IOP Conf Ser: Earth Environ Sci. 2020 Mar 1;448(1):012019. [CrossRef]
  15. Cuenca E, Jodra P, Pérez-López A, González-Rodríguez L, Fernandes Da Silva S, Veiga-Herreros P, Domínguez R. Effects of Beetroot Juice Supplementation on Performance and Fatigue in a 30-s All-Out Sprint Exercise: A Randomized, Double-Blind Cross-Over Study. Nutrients. 2018 Sep 4;10(9):1222. [CrossRef]
  16. Megna M, Pamico A, Cristella G, Saggini R, Jirillo E, Ranieri M. Effects of Herbal Supplements on the Immune System in Relation to Exercise. Int J Immunopathol Pharmacol. 2012 Jan;25(1_suppl):43–9. [CrossRef]
  17. Xu Z, Shan Y. Anti-fatigue effects of polysaccharides extracted from Portulaca oleracea L. in mice. INDIAN J BIOCHEM BIOPHYS. 2014;51.
  18. Sellami M, Slimeni O, Pokrywka A, Kuvačić G, D Hayes L, Milic M, Padulo J. Herbal medicine for sports: a review. Journal of the International Society of Sports Nutrition. 2018 Jan 5;15(1):14.
  19. Zhang YD, Liu LY, Wang D, Yuan XL, Zheng Y, Wang Y. Isolation and identification of bioactive compounds from Antrodia camphorata against ESKAPE pathogens. Al-Judaibi AA, editor. PLoS ONE. 2023 Oct 27;18(10):e0293361. [CrossRef]
  20. Luo C, Xu X, Wei X, Feng W, Huang H, Liu H, Xu R, Lin J, Han L, Zhang D. Natural medicines for the treatment of fatigue: Bioactive components, pharmacology, and mechanisms. Pharmacological Research. 2019 Oct;148:104409. [CrossRef]
  21. Lu X, Chen J, Huang L, Ou Y, Wu J, Guo Z, Zheng B. The Anti-Fatigue Effect of Glycoprotein from Hairtail Fish (Trichiurus lepturus) on BALB/c Mice. Foods. 2023 Mar 14;12(6):1245. [CrossRef]
  22. Ma S, Ono M, Mizugaki A, Kato H, Miyashita M, Suzuki K. Cystine/Glutamine Mixture Supplementation Attenuated Fatigue during Endurance Exercise in Healthy Young Men by Enhancing Fatty Acid Utilization. Sports. 2022 Sep 27;10(10):147. [CrossRef]
  23. Sasidharan S, Chen Y, Saravanan D, Sundram KM, Yoga Latha L. Extraction, Isolation and Characterization of Bioactive Compounds from Plants’ Extracts. Afr J Tradit Complement Altern Med. 2010 Oct 2;8(1):1–10.
  24. Williams M. Dietary Supplements and Sports Performance: Herbals. Journal of the International Society of Sports Nutrition. 2006 Jun 1;3(1):1.
  25. Chen CK, Muhamad AS, Ooi FK. Herbs in exercise and sports. J Physiol Anthropol. 2012 Dec;31(1):4. [CrossRef]
  26. Bucci LR. Selected herbals and human exercise performance. The American Journal of Clinical Nutrition. 2000 Aug;72(2):624S-636S. [CrossRef]
  27. Kpatcha T, Dosseh K, Idoh K, Agbonon A, Gbéassor M. EFFECT OF ADANSONIA DIGITATA L. ON PHYSICAL PERFORMANCE AND HAEMATOLOGICAL PARAMETERS IN RATS. 2016.
  28. Sama H, Traoré M, Guenné S, Séré I, Hilou A, Dicko MH. Ethnobotanical and Phytochemical Profiling of Medicinal Plants from Burkina Faso Used to Increase Physical Performance. Medicines. 2022 Jan 28;9(2):10. [CrossRef]
  29. Ouédraogo C, Guenné S, Bienvenu Somda M, Blanche M’Po SM, Sidibé I, Séré I, Roland Méda NT. Perceptions sur les Conduites Dopantes dans le Sport dans la Ville de Bobo-Dioulasso (Burkina Faso). ESJ. 2022 Jan 31;18(3):262.
  30. Ivanova Stojcheva E, Quintela JC. The Effectiveness of Rhodiola rosea L. Preparations in Alleviating Various Aspects of Life-Stress Symptoms and Stress-Induced Conditions—Encouraging Clinical Evidence. Molecules. 2022 Jun 17;27(12):3902.
  31. Cerulli C, Borrione P, Sabatini S. Effects of chronic rhodiola rosea supplementation on sport performance and antioxidant capacpitryeilnimItrinaCainreydrAemsuallets: A. PARISI 1, E. TRANCHITA 1, G. DURANTI 2, E. CIMINELLI 1, F. QUARANTA 1, R. CECI 2,. THE JOURNAL OF SPORTS MEDICINE AND PHYSICAL FITNESS. 2010;50(1).
  32. Kang DZ, Hong HD, Kim KI, Choi SY. Anti-Fatigue Effects of Fermented Rhodiola rosea Extract in Mice. JFN. 2015 Mar 31;20(1):38–42. [CrossRef]
  33. Lee FT, Kuo TY, Liou SY, Chien CT. Chronic Rhodiola rosea Extract Supplementation Enforces Exhaustive Swimming Tolerance. Am J Chin Med. 2009 Jan;37(03):557–72. [CrossRef]
  34. Jówko E, Sadowski J, Długołęcka B, Gierczuk D, Opaszowski B, Cieśliński I. Effects of Rhodiola rosea supplementation on mental performance, physical capacity, and oxidative stress biomarkers in healthy men. Journal of Sport and Health Science. 2018 Oct;7(4):473–80. [CrossRef]
  35. Williams TD, Langley HN, Roberson CC, Rogers RR, Ballmann CG. Effects of Short-Term Golden Root Extract (Rhodiola rosea) Supplementation on Resistance Exercise Performance. IJERPH. 2021 Jun 29;18(13):6953. [CrossRef]
  36. Noreen EE, Buckley JG, Lewis SL, Brandauer J, Stuempfle KJ. The Effects of an Acute Dose of Rhodiola rosea on Endurance Exercise Performance. Journal of Strength and Conditioning Research. 2013 Mar;27(3):839–47. [CrossRef]
  37. De Bock K, Eijnde BO, Ramaekers M, Hespel P. Acute Rhodiola Rosea Intake Can Improve Endurance Exercise Performance. International Journal of Sport Nutrition and Exercise Metabolism. 2004 Jun;14(3):298–307. [CrossRef]
  38. Shevtsov VA, Zholus BI, Shervarly VI, Vol’skij VB, Korovin YP, Khristich MP, Roslyakova NA, Wikman G. A randomized trial of two different doses of a SHR-5 Rhodiola rosea extract versus placebo and control of capacity for mental work. Phytomedicine. 2003 Jan;10(2–3):95–105.
  39. Liu C, Zhao H, Yan Y, Yang W, Chen S, Song G, Li X, Gu Y, Yun H, Li Y. Synergistic Effect of Rhodiola rosea and Caffeine Supplementation on the Improvement of Muscle Strength and Muscular Endurance: A Pilot Study for Rats, Resistance Exercise-Untrained and -Trained Volunteers. Nutrients. 2023 Jan 22;15(3):582.
  40. Liudong F, Feng Z, Daoxing S, Xiufang Q, Xiaolong F, Haipeng L. Evaluation of antioxidant properties and anti- fatigue effect of green tea polyphenols. Scientific Research and Essays. 2011;6(13):2624–9.
  41. Machado ÁS, Da Silva W, Souza MA, Carpes FP. Green Tea Extract Preserves Neuromuscular Activation and Muscle Damage Markers in Athletes Under Cumulative Fatigue. Front Physiol. 2018 Aug 17;9:1137. [CrossRef]
  42. Lee ES, Yang YJ, Lee JH, Yoon YS. Effect of high-dose ginsenoside complex (UG0712) supplementation on physical performance of healthy adults during a 12-week supervised exercise program: A randomized placebo-controlled clinical trial. Journal of Ginseng Research. 2018 Apr;42(2):192–8. [CrossRef]
  43. Mishra JN, Verma NK. An Overview on Panax ginseng. 2017;3(3).
  44. Valdés-González JA, Sánchez M, Moratilla-Rivera I, Iglesias I, Gómez-Serranillos MP. Immunomodulatory, Anti-Inflammatory, and Anti-Cancer Properties of Ginseng: A Pharmacological Update. Molecules. 2023 May 3;28(9):3863. [CrossRef]
  45. Wang J, Li S, Fan Y, Chen Y, Liu D, Cheng H, Gao X, Zhou Y. Anti-fatigue activity of the water-soluble polysaccharides isolated from Panax ginseng C. A. Meyer. Journal of Ethnopharmacology. 2010 Jul;130(2):421–3. [CrossRef]
  46. Bhattacharjee I, Bandyopadhyay A. Effects of Acute Supplementation of Panax ginseng on Endurance Performance in Healthy Adult Males of Kolkata, India. IJCEP. 2020 Jul 7;7(2):63–8. [CrossRef]
  47. Oliveira ACCD, Perez AC, Prieto JG, Duarte IDG, Alvarez AI. Protection of Panax ginseng in injured muscles after eccentric exercise. Journal of Ethnopharmacology. 2005 Feb;97(2):211–4. [CrossRef]
  48. Hsiao CY, Hsu YJ, Tung YT, Lee MC, Huang CC, Hsieh CC. Effects of Antrodia camphorata and Panax ginseng supplementation on anti-fatigue properties in mice. The Journal of Veterinary Medical Science. 2018;80(2):284–91. [CrossRef]
  49. Oh HA, Kim DE, Choi HJ, Kim NJ, Kim DH. Anti-fatigue Effects of 20(S)-Protopanaxadiol and 20(S)-Protopanaxatriol in Mice. Biological & Pharmaceutical Bulletin. 2015;38(9):1415–9. [CrossRef]
  50. Kulaputana O, Thanakomsirichot S, Anomasiri W. Ginseng Supplementation Does Not Change Lactate Threshold and Physical Performances in Physically Active Thai Men. 2007;90(6).
  51. Huang LZ, Huang BK, Ye Q, Qin LP. Bioactivity-guided fractionation for anti-fatigue property of Acanthopanax senticosus. Journal of Ethnopharmacology. 2011 Jan;133(1):213–9. [CrossRef]
  52. Zhang XL, Ren F, Huang W, Ding RT, Zhou QS, Liu XW. Anti-Fatigue Activity of Extracts of Stem Bark from Acanthopanax senticosus. Molecules. 2010 Dec 24;16(1):28–37. [CrossRef]
  53. Chen CS, Wang ML, Liu RH, Chen SP, Lu TM, Tsai WY, Huang CF, Yang CC, Tzeng YM. Anti-Fatigue Effect of Aqueous Extract of Anisomeles indica (L) Kuntze in Mice. Trop J Pharm Res. 2016 Apr 8;15(3):489. [CrossRef]
  54. Huang CC, Hsu MC, Huang WC, Yang HR, Hou CC. Triterpenoid-Rich Extract from Antrodia camphorata Improves Physical Fatigue and Exercise Performance in Mice. Evidence-Based Complementary and Alternative Medicine. 2012;2012:1–8. [CrossRef]
  55. Miao X, Xiao B, Shui S, Yang J, Huang R, Dong J. Metabolomics analysis of serum reveals the effect of Danggui Buxue Tang on fatigued mice induced by exhausting physical exercise. Journal of Pharmaceutical and Biomedical Analysis. 2018 Mar;151:301–9. [CrossRef]
  56. Zhang Y, Yao X, Bao B, Zhang Y. Anti-fatigue activity of a triterpenoid-rich extract from Chinese bamboo shavings ( Caulis bamfusae in taeniam): Anti-fatigue activity of bamboo shavings. Phytother Res. 2006 Oct;20(10):872–6.
  57. Zhu J, Yi J, Kang Q, Huang J, Cui Y, Zhang G, Wang Z, Zhang L, Zheng Z, Lu J, Hao L. Anti-fatigue activity of hemp leaves water extract and the related biochemical changes in mice. Food and Chemical Toxicology. 2021 Apr; 150:112054. [CrossRef]
  58. Peng F, Yin H, Du B, Niu K, Ren X, Yang Y. Anti-fatigue activity of purified flavonoids prepared from chestnut (Castanea mollissima) flower. Journal of Functional Foods. 2021 Apr; 79:104365. [CrossRef]
  59. Cai RL, Yang MH, Shi Y, Chen J, Li YC, Qi Y. Antifatigue activity of phenylethanoid-rich extract from Cistanche deserticola: Antifatigue activity of phenylethanoid-rich extract from c. deserticola. Phytother Res. 2010 Feb;24(2):313–5.
  60. Zhong L, Zhao L, Yang F, Yang W, Sun Y, Hu Q. Evaluation of anti-fatigue property of the extruded product of cereal grains mixed with Cordyceps militaris on mice. Journal of the International Society of Sports Nutrition. 2017 Jan 3;14(1):15. [CrossRef]
  61. Jeong H, Kim OK, Park J, Kim K, Chung JW, Shim S, You Y, Jun W. Ethanol Extract of Eriobotrya japonica Leaves Enhanced Swimming Capacity in Mice. Journal of Food and Nutrition Research. 2017;5(6):413–7.
  62. Jin HM, Wei P. Anti-Fatigue Properties of Tartary Buckwheat Extracts in Mice. IJMS. 2011 Jul 25;12(8):4770–80. [CrossRef]
  63. Chen YJ, Baskaran R, Shibu MA, Lin WT. Anti-Fatigue and Exercise Performance Improvement Effect of Glossogyne tenuifolia Extract in Mice. Nutrients. 2022 Feb 27;14(5):1011. [CrossRef]
  64. Lamou B, Taiwe GS, Hamadou A, Abene, Houlray J, Atour MM, Tan PV. Antioxidant and Antifatigue Properties of the Aqueous Extract of Moringa oleifera in Rats Subjected to Forced Swimming Endurance Test. Oxidative Medicine and Cellular Longevity. 2016; 2016:1–9.
  65. Chen H, He X, Liu Y, Li J, He Q, Zhang C, Wei B, Zhang Y, Wang J. Extraction, purification and anti-fatigue activity of γ-aminobutyric acid from mulberry (Morus alba L.) leaves. Sci Rep. 2016 Jan 8;6(1):18933.
  66. Qi B, Liu L, Zhang H, Zhou G xin, Wang S, Duan X zheng, Bai X yuan, Wang S ming, Zhao D qing. Anti-fatigue effects of proteins isolated from Panax quinquefolium. Journal of Ethnopharmacology. 2014 Apr;153(2):430–4.
  67. Hu M, Du J, Du L, Luo Q, Xiong J. Anti-fatigue activity of purified anthocyanins prepared from purple passion fruit (P. edulis Sim) epicarp in mice. Journal of Functional Foods. 2020 Feb; 65:103725. [CrossRef]
  68. Dorneles IMP, Fucks MB, Fontela PC, Frizzo MN, Winkelmann ER. Guarana (Paullinia cupana) presents a safe and effective anti-fatigue profile in patients with chronic kidney disease: A randomized, double-blind, three-arm, controlled clinical trial. Journal of Functional Foods. 2018 Dec; 51:1–7. [CrossRef]
  69. Chu H, Sun H, Yan GL, Zhang&Chang Liu AH, Dong H, Meng XC, Wang XJ, National TCM Key Lab of Serum Pharmacochemistry, Key Laboratory of Metabolomics and Chinmedomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China. Metabolomics Analysis of Health Functions of Physalis Pubescens L. using by Ultra-performance Liquid Chromatography/Electrospray Ionization Quadruple Time-of-Flight Mass Spectrometry. World Journal of Traditional Chinese Medicine. 2015;1(3):9–20. [CrossRef]
  70. Ding X, Tang K, Lu P, Putheti R. Antifatigue effects of polydatin from Chinese herb Polygonum Cuspidatum in swimming mice. African Journal of Microbiology Research. 2009;3(7):358–61.
  71. D’Angelo S, Roberta R. L’impact de la supplémentation en grenade (punica granatum l.) sur la performance sportive. Sciences du sport. 2020;13.
  72. Yong-xin X, Jian-jun Z. Evaluation of anti-fatigue activity of total saponins of Radix notoginseng. INDIAN J MED RES. 2013.
  73. Shanely RA, Nieman DC, Zwetsloot KA, Knab AM, Imagita H, Luo B, Davis B, Zubeldia JM. Evaluation of Rhodiola rosea supplementation on skeletal muscle damage and inflammation in runners following a competitive marathon. Brain, Behavior, and Immunity. 2014 Jul;39:204–10. [CrossRef]
  74. Spasov AA, Wikman GK, Mandrikov VB, Mironova IA, Neumoin VV. A double-blind, placebo-controlled pilot study of the stimulating and adaptogenic effect of Rhodiola rosea SHR-5 extract on the fatigue of students caused by stress during an examination period with a repeated low-dose regimen. Phytomedicine. 2000 Apr;7(2):85–9. [CrossRef]
  75. Olennikov DN, Nikolaev VM, Chirikova NK. Sagan Dalya Tea, a New “Old” Probable Adaptogenic Drug: Metabolic Characterization and Bioactivity Potentials of Rhododendron adamsii Leaves. Antioxidants. 2021 May 27;10(6):863. [CrossRef]
  76. Jung KA, Han D, Kwon EK, Lee CH, Kim YE. Antifatigue Effect of Rubus coreanus Miquel Extract in Mice. Journal of Medicinal Food. 2007 Dec;10(4):689–93. [CrossRef]
  77. Agbodjogbe WKDD, Tito ASI, Adjakpahoun G, Messan F, Dansou PH. Antioxidant properties of Senna siamea and effects on sports performance in Wistar rats. Int J Bio Chem Sci. 2022 Aug 28;16(3):1193–203. [CrossRef]
  78. Liu DD, Ji XW, Li RW. Effects of Siraitia grosvenorii Fruits Extracts on Physical Fatigue in Mice. 2013.
  79. Feng H, Ma H, Lin H, Putheti R. Antifatigue activity of water extracts of Toona sinensis Roemor leaf and exercise-related changes in lipid peroxidation in endurance exercise. 2009;3(11):949–54.
  80. Kumar GP, Anand T, Singsit D, Khanum F, Anilakumar KR. Evaluation of antioxidant and anti-fatigue properties of Trigonella foenum-graecum L. in rats subjected to weight loaded forced swim test. Pharmacognosy Journal. 2013 Mar;5(2):66–71. [CrossRef]
  81. Didio FP, Duarte AR, Stefani GP. Effects of the Withania somnifera supplementation on sports performance: a systematic review and meta-analysis. Nor Afr J Food Nutr Res. 2022 Jan 2;6(13):1–8. [CrossRef]
  82. Zhao H peng, Zhang Y, Liu Z, Chen J yue, Zhang S yan, Yang X dong, Zhou H li. Acute toxicity and anti-fatigue activity of polysaccharide-rich extract from corn silk. Biomedicine & Pharmacotherapy. 2017 Jun;90:686–93.
  83. Tarazona-Díaz MP, Alacid F, Carrasco M, Martínez I, Aguayo E. Watermelon Juice: Potential Functional Drink for Sore Muscle Relief in Athletes. J Agric Food Chem. 2013 Aug 7;61(31):7522–8.
  84. Dave Mehta S, Rathore P, Rai G. Ginseng: Pharmacological Action and Phytochemistry Prospective. In: Hano C, Chen JT, editors. Ginseng - Modern Aspects of the Famed Traditional Medicine [Internet]. IntechOpen; 2022 [cited 2023 Jul 27]. Available from: https://www.intechopen.com/chapters/78426.
  85. Bowtell J, Kelly V. Fruit-Derived Polyphenol Supplementation for Athlete Recovery and Performance. Sports Med. 2019 Feb;49(S1):3–23. [CrossRef]
  86. Kennedy DO. Phytochemicals for Improving Aspects of Cognitive Function and Psychological State Potentially Relevant to Sports Performance. Sports Med. 2019 Feb;49(S1):39–58. [CrossRef]
  87. Jo E, Bartosh R, Auslander AT, Directo D, Osmond A, Wong MW. Post-Exercise Recovery Following 30-Day Supplementation of Trans-Resveratrol and Polyphenol-Enriched Extracts. Sports. 2019 Oct 20;7(10):226. [CrossRef]
  88. Miao F, Wu D, Ni G. Evaluation of Anti-Fatigue Activity of Flavonoids from Tartary Buckwheat in Mice. Afr J Trad Compl Alt Med. 2016 Feb 19;13(2):52.
  89. Xiaoming W, Ling L, Jinghang Z. Antioxidant and Anti-Fatigue Activities of Flavonoids from Puerariae radix. Afr J Trad Compl Alt Med. 2012 Jan 18;9(2):221–7. [CrossRef]
  90. Murase T, Haramizu S, Ota N, Hase T. Suppression of the aging-associated decline in physical performance by a combination of resveratrol intake and habitual exercise in senescence-accelerated mice. Biogerontology. 2009 Aug;10(4):423–34. [CrossRef]
  91. Qin L, Lu T, Qin Y, He Y, Cui N, Du A, Sun J. In Vivo Effect of Resveratrol-Loaded Solid Lipid Nanoparticles to Relieve Physical Fatigue for Sports Nutrition Supplements. Molecules. 2020 Nov 13;25(22):5302. [CrossRef]
  92. Wu RE, Huang WC, Liao CC, Chang YK, Kan NW, Huang CC. Resveratrol Protects against Physical Fatigue and Improves Exercise Performance in Mice. Molecules. 2013 Apr 19;18(4):4689–702. [CrossRef]
  93. Yang W, Chen X, Li Y, Guo S, Wang Z, Yu X. Advances in Pharmacological Activities of Terpenoids. Natural Product Communications. 2020 Mar;15(3):1934578X2090355. [CrossRef]
  94. Geng P, Siu KC, Wang Z, Wu JY. Antifatigue Functions and Mechanisms of Edible and Medicinal Mushrooms. BioMed Research International. 2017;2017:1–16. [CrossRef]
  95. Li S, Chan Z. Evaluation of Antifatigue Effects of 20(S)-Ginsenoside Rg3 in Forced Swimming Mice. pharmaceutical-sciences [Internet]. 2018 [cited 2023 Jun 30];80(3). Available from: http://www.ijpsonline.com/articles/evaluation-of-antifatigue-effects-of-20s-ginsenosiderg3-in-forced-swimming-mice-3493.html.
  96. Rajput A, Sharma R, Bharti R. Pharmacological activities and toxicities of alkaloids on human health. Materials Today: Proceedings. 2022;48:1407–15. [CrossRef]
  97. Hsu YJ, Huang WC, Chiu CC, Liu YL, Chiu WC, Chiu CH, Chiu YS, Huang CC. Capsaicin Supplementation Reduces Physical Fatigue and Improves Exercise Performance in Mice. Nutrients. 2016 Oct 20;8(10):648. [CrossRef]
  98. Pesta DH, Angadi SS, Burtscher M, Roberts CK. The effects of caffeine, nicotine, ethanol, and tetrahydrocannabinol on exercise performance. Nutr Metab (Lond). 2013 Dec;10(1):71. [CrossRef]
  99. Spriet LL. Exercise and Sport Performance with Low Doses of Caffeine. Sports Med. 2014 Nov;44(S2):175–84. [CrossRef]
  100. Trexler ET, Smith-Ryan AE, Roelofs EJ, Hirsch KR, Mock MG. Effects of coffee and caffeine anhydrous on strength and sprint performance. European Journal of Sport Science. 2016 Aug 17;16(6):702–10.
  101. Wang P, Wang D, Hu J, Tan BK, Zhang Y, Lin S. Natural bioactive peptides to beat exercise-induced fatigue: A review. Food Bioscience. 2021 Oct;43:101298. [CrossRef]
  102. Marquezi ML, Roschel HA, Costa ADS, Sawada LA, Lancha Jr. AH. Effect of Aspartate and Asparagine Supplementation on Fatigue Determinants in Intense Exercise. International Journal of Sport Nutrition and Exercise Metabolism. 2003 Mar;13(1):65–75.
  103. Thirupathi A, Freitas S, Sorato HR, Pedroso GS, Effting PS, Damiani AP, Andrade VM, Nesi RT, Gupta RC, Muller AP, Pinho RA. Modulatory effects of taurine on metabolic and oxidative stress parameters in a mice model of muscle overuse. Nutrition. 2018 Oct;54:158–64. [CrossRef]
  104. Xu M, Liang R, Li Y, Wang J. Anti-fatigue effects of dietary nucleotides in mice. Food & Nutrition Research. 2017 Jan;61(1):1334485. [CrossRef]
  105. Yen CH, Tsao TH, Huang CU, Yang CB, Kuo CS. Effects of sweet cassava polysaccharide extracts on endurance exercise in rats. Journal of the International Society of Sports Nutrition. 2013 Jan 3;10(1):18. [CrossRef]
  106. Liu J, Du C, Wang Y, Yu Z. Anti-fatigue activities of polysaccharides extracted from Hericium erinaceus. Experimental and Therapeutic Medicine. 2015 Feb;9(2):483–7. [CrossRef]
  107. Moon JM, Ratliff KM, Blumkaitis JC, Harty PS, Zabriskie HA, Stecker RA, Currier BS, Jagim AR, Jäger R, Purpura M, Kerksick CM. Effects of daily 24-gram doses of rice or whey protein on resistance training adaptations in trained males. Journal of the International Society of Sports Nutrition. 2020 Jan 3;17(1):60. [CrossRef]
  108. Yu SH, Huang HY, Korivi M, Hsu MF, Huang CY, Hou CW, Chen CY, Kao CL, Lee RP, Lee SD, Kuo CH. Oral Rg1 supplementation strengthens antioxidant defense system against exercise-induced oxidative stress in rat skeletal muscles. Journal of the International Society of Sports Nutrition. 2012 Feb 6;9(1):23. [CrossRef]
  109. Hsu YJ, Huang WC, Chiu CC, Liu YL, Chiu WC, Chiu CH, Chiu YS, Huang CC. Capsaicin Supplementation Reduces Physical Fatigue and Improves Exercise Performance in Mice. Nutrients. 2016 Oct 20;8(10):648. [CrossRef]
  110. Bell DG, Ira J, Jiri Z. Effects of caffeine, ephedrine and their combination on time to exhaustion during high-intensity exercise. Eur J Appl Physiol. 1998;(77):427–33.
  111. Kan NW, Ho CS, Chiu YS, Huang WC, Chen PY, Tung YT, Huang CC. Effects of Resveratrol Supplementation and Exercise Training on Exercise Performance in Middle-Aged Mice. Molecules. 2016 May 18;21(5):661. [CrossRef]
  112. Chen H, He X, Liu Y, Li J, He Q, Zhang C, Wei B, Zhang Y, Wang J. Extraction, purification and anti-fatigue activity of γ-aminobutyric acid from mulberry (Morus alba L.) leaves. Sci Rep. 2016 Jan 8;6(1):18933.
  113. Miao X, Xiao B, Shui S, Yang J, Huang R, Dong J. Metabolomics analysis of serum reveals the effect of Danggui Buxue Tang on fatigued mice induced by exhausting physical exercise. Journal of Pharmaceutical and Biomedical Analysis. 2018 Mar;151:301–9. [CrossRef]
  114. Wang J, Li C, Hu W, Li L, Cai G, Liu Y, Wang D. Studies on the anti-fatigue activities of Irpex lacteus polysaccharide- enriched extract in mouse model. Pak J Pharm Sci. 2019.
  115. Kennedy DO, Haskell CF, Robertson B, Reay J, Brewster-Maund C, Luedemann J, Maggini S, Ruf M, Zangara A, Scholey AB. Improved cognitive performance and mental fatigue following a multi-vitamin and mineral supplement with added guaraná (Paullinia cupana). Appetite. 2008 Mar;50(2–3):506–13. [CrossRef]
  116. Pomportes L, Davranche K, Hays A, Brisswalter J. Effet d’un complexe créatine–guarana sur la puissance musculaire et la performance cognitive chez des sportifs de haut niveau de performance. Science & Sports. 2015 Sep;30(4):188–95. [CrossRef]
  117. Hu M, Du J, Du L, Luo Q, Xiong J. Anti-fatigue activity of purified anthocyanins prepared from purple passion fruit (P. edulis Sim) epicarp in mice. Journal of Functional Foods. 2020 Feb 1;65:103725. [CrossRef]
  118. Kwon DA, Kim YS, Kim SK, Baek SH, Kim HK, Lee HS. Antioxidant and antifatigue effect of a standardized fraction (HemoHIM) from Angelica gigas, Cnidium officinale, and Paeonia lactiflora. Pharmaceutical Biology. 2021 Jan 1;59(1):389–98.
  119. Heaney LM, Deighton K, Suzuki T. Non-targeted metabolomics in sport and exercise science. Journal of Sports Sciences. 2019 May 3;37(9):959–67. [CrossRef]
  120. Duft RG, Castro A, Chacon-Mikahil MPT, Cavaglieri CR. Metabolomics and Exercise: possibilities and perspectives. Motriz: rev educ fis [Internet]. 2017 [cited 2023 Jul 27];23(2). Available from: http://www.scielo.br/scielo.php?script=sci_arttext&pid=S1980-65742017000200201&lng=en&tlng=en.
  121. Bongiovanni T, Pintus R, Dessì A, Noto A, Sardo S, Finco G, Corsello G, Fanos V. Sportomics: metabolomics applied to sports. The new revolution? European Review for Medical and Pharmacological Sciences. 2019;23:11011–9.
  122. Krambeck K, Oliveira A, Santos D, Pintado MM, Baptista Silva J, Sousa Lobo JM, Amaral MH. Identification and Quantification of Stilbenes (Piceatannol and Resveratrol) in Passiflora edulis By-Products. Pharmaceuticals. 2020 Apr 20;13(4):73. [CrossRef]
  123. Zakharenko AM, Razgonova MP, Pikula KS, Golokhvast KS. Simultaneous Determination of 78 Compounds of Rhodiola rosea Extract by Supercritical CO2-Extraction and HPLC-ESI-MS/MS Spectrometry. Tayyab S, editor. Biochemistry Research International. 2021 Jul 6;2021:1–16. [CrossRef]
  124. Wu F, Yao X, Xu J, Wu Y, Yang Y, Jin Y, Xie H, Liu Y, Yang Y, Zheng X. One New Phenolic Compound from Castanea mollissima Shells and its Suppression of HepatomaCell Proliferation and Inflammation by Inhibiting NF-κB Pathway. IJMS. 2019 Jan 22;20(3):466.
  125. Malathy R, Prabakaran M, Kalaiselvi K, Chung IM, Kim SH. Comparative Polyphenol Composition, Antioxidant and Anticorrosion Properties in Various Parts of Panax ginseng Extracted in Different Solvents. Applied Sciences. 2020 Dec 24;11(1):93.
  126. Zhang L, Gao H yuan, Baba M, Okada Y, Okuyama T, Wu L jun, Zhan L bin. Extracts and compounds with anti-diabetic complications and anti-cancer activity from Castanea mollissina Blume (Chinese chestnut). BMC Complement Altern Med. 2014 Dec;14(1):422.
  127. Zhang YN, Zhu SJ, Li N, Jing YN, Yue XF. Screening and identification of the active components from Puerariae Radix by HUVEC/CMC-LC-MS2. Journal of Chromatography B. 2019 Nov;1132:121825. [CrossRef]
  128. Rana A, Singh HP, Gulati A. Investigation of major phenolic antioxidants from Camellia sinensis fruits. Weaver G, editor. Cogent Chemistry. 2015 Dec 31;1(1):1080652. [CrossRef]
  129. Hu Y, Ding J, Sun Z, Zong Z, Song H, Sun X, Xu B, Qi Z, Liu B, Li W. Identification and analysis of chemical constituents of total glycosides of Cistanche deserticola YC Ma in rat hepatic metabolism by ultra performance liquid chromatography-quadrupole/time-of-flight mass spectrometry (UPLC-Q-TOF-MS). mat express. 2022 Aug 1;12(8):1059–71. [CrossRef]
  130. Fuzzati N, Gabetta B, Jayakar K, Pace R, Peterlongo F. Liquid chromatography–electrospray mass spectrometric identification of ginsenosides in Panax ginseng roots. Journal of Chromatography A. 1999 Aug;854(1–2):69–79. [CrossRef]
  131. Gao HY, Wang XB, Xi RG, Sun BH, Huang J, Wu LJ. Structure and Absolute Configuration of a Diterpenoid from Castanea mollissima. Natural Product Communications. 2010 Jan;5(1):1934578X1000500. [CrossRef]
  132. Lee DG, Lee JS, Kim KT, Jacinto SD, Lee S. Analysis of terpenoids and steroids in the seeds of Panax ginseng. Hortic Environ Biotechnol. 2020 Jun;61(3):609–13. [CrossRef]
  133. Basappa G, Kumar V, Sarojini BK, Poornima DV, Gajula H, Sannabommaji TK, Rajashekar J. Chemical composition, biological properties of Anisomeles indica Kuntze essential oil. Industrial Crops and Products. 2015;77:89–96. [CrossRef]
  134. Belliardo F, Martelli A, Valle MG. HPLC determination of caffeine and theophylline inPaullinia cupana Kunth (Guarana) and Cola spp. samples. Z Lebensm Unters Forch. 1985 May;180(5):398–401. [CrossRef]
  135. Ciulu-Costinescu F, Calina D, Chirigiu L, Averis LME, Rosulescu E, Bubulica MV. Identification and Quantification of Capsaicin from Capsicum Annuum L. by RP-HPLC. 2015.
  136. Benjamin MAZ, Ng SY, Saikim FH, Rusdi NA. The Effects of Drying Techniques on Phytochemical Contents and Biological Activities on Selected Bamboo Leaves. Molecules. 2022 Sep 30;27(19):6458. [CrossRef]
  137. Barreira S, Moutinho C, Silva AMN, Neves J, Seo EJ, Hegazy MEF, Efferth T, Gomes LR. Phytochemical characterization and biological activities of green tea (Camellia sinensis) produced in the Azores, Portugal. Phytomedicine Plus. 2021 Feb;1(1):100001. [CrossRef]
  138. Tessier F, Marconnet P. Radicaux libres, systèmes antioxydants et exercice. Science & Sports. 1995 Jan;10(1):1–13.
  139. Powers SK, Goldstein E, Schrager M, Ji LL. Exercise Training and Skeletal Muscle Antioxidant Enzymes: An Update. Antioxidants. 2022 Dec 25;12(1):39. [CrossRef]
  140. El Assar M, Álvarez-Bustos A, Sosa P, Angulo J, Rodríguez-Mañas L. Effect of Physical Activity/Exercise on Oxidative Stress and Inflammation in Muscle and Vascular Aging. IJMS. 2022 Aug 5;23(15):8713. [CrossRef]
  141. Atalay M, Seene T, Hänninen O, Sen CK. Skeletal muscle and heart antioxidant defences in response to sprint training. Acta Physiologica Scandinavica. 1996 Oct;158(2):129–34. [CrossRef]
  142. Leeuwenburgh C, Hollander J, Leichtweis S, Griffiths M, Gore M, Ji LL. Adaptations of glutathione antioxidant system to endurance training are tissue and muscle fiber specific. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology. 1997 Jan 1;272(1):R363–9. [CrossRef]
  143. Viña J, Salvador-Pascual A, Tarazona-Santabalbina FJ, Rodriguez-Mañas L, Gomez-Cabrera MC. Exercise training as a drug to treat age associated frailty. Free Radical Biology and Medicine. 2016 Sep;98:159–64. [CrossRef]
  144. Meo SD, Napolitano G, Venditti P. Mediators of Physical Activity Protection against ROS-Linked Skeletal Muscle Damage. Int J Mol Sci. 2019. [CrossRef]
  145. Simioni C, Zauli G, Martelli AM, Vitale M, Sacchetti G, Gonelli A, Neri LM. Oxidative stress: role of physical exercise and antioxidant nutraceuticals in adulthood and aging. Oncotarget. 2018 Mar 30;9(24):17181–98. [CrossRef]
  146. Hollander J, Fiebig R, Gore M, Bejma J, Ookawara T, Ohno H, Ji LL. Superoxide dismutase gene expression in skeletal muscle: fiber-specific adaptation to endurance training. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology. 1999 Sep 1;277(3):R856–62. [CrossRef]
  147. Steinbacher P, Eckl P. Impact of Oxidative Stress on Exercising Skeletal Muscle. Biomolecules. 2015 Apr 10;5(2):356–77. [CrossRef]
  148. Improta-Caria AC, Soci ÚPR, Pinho CS, Aras Júnior R, De Sousa RAL, Bessa TCB. Physical Exercise and Immune System: Perspectives on the COVID-19 pandemic. Rev Assoc Med Bras. 2021;67(suppl 1):102–7. [CrossRef]
  149. Ferreira LF, Laitano O. Regulation of NADPH oxidases in skeletal muscle. Free Radical Biology and Medicine. 2016 Sep; 98:18–28. [CrossRef]
  150. Jackson MJ, Vasilaki A, McArdle A. Cellular mechanisms underlying oxidative stress in human exercise. Free Radical Biology and Medicine. 2016 Sep; 98:13–7. [CrossRef]
  151. Powers SK, Deminice R, Ozdemir M, Yoshihara T, Bomkamp MP, Hyatt H. Exercise-induced oxidative stress: Friend or foe? Journal of Sport and Health Science. 2020 Sep;9(5):415–25.
  152. Suzuki K, Hayashida H. Effect of Exercise Intensity on Cell-Mediated Immunity. Sports. 2021 Jan 11;9(1):8. [CrossRef]
  153. Pedersen BK, Hoffman-Goetz L. Exercise and the Immune System: Regulation, Integration, and Adaptation. Physiological Reviews. 2000 Jul 1;80(3):1055–81. [CrossRef]
  154. Senchina DS, Kohut ML. Immunological outcomes of exercise in older adults. Clinical Interventions in Aging. 2007 Mar;2(1):3–16. [CrossRef]
  155. Scheffer DDL, Latini A. Exercise-induced immune system response: Anti-inflammatory status on peripheral and central organs. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 2020 Oct;1866(10):165823. [CrossRef]
  156. Simpson RJ, Campbell JP, Gleeson M, Krüger K, Nieman DC, Pyne DB, Turner E, Walsh NP. Can exercise affect immune function to increase susceptibility to infection? 2020.
  157. Valle L, Hernandez R. Physical activity as antioxidant and palliative beneficial option in human immunodeficiency virus infection. Oxid Antioxid Med Sci. 2013;2(4):231. [CrossRef]
  158. Leung A, Gregory NS, Allen LAH, Sluka KA. Regular physical activity prevents chronic pain by altering resident muscle macrophage phenotype and increasing IL-10 in mice. 2017.
  159. Ellingsgaard H, Hojman P, Pedersen BK. Exercise and health — emerging roles of IL-6. Current Opinion in Physiology. 2019 Aug;10:49–54.
  160. Chen H, Shen L, Liu Y, Ma X, Long L, Ma X, Ma L, Chen Z, Lin X, Si L, Chen X. Strength Exercise Confers Protection in Central Nervous System Autoimmunity by Altering the Gut Microbiota. Front Immunol. 2021 Mar 16;12:628629. [CrossRef]
  161. Moon PD, Kim KY, Rew KH, Kim HM, Jeong HJ. Anti-fatigue effects of porcine placenta and its amino acids in a behavioral test on mice. Can J Physiol Pharmacol. 2014 Nov;92(11):937–44. [CrossRef]
  162. Garige BSR, Keshetti S, Vattikuti UMR. CNS Depressant effects and muscle relaxant activity of Galphimia glauca leaf methanol extract. International Journal of PharmTech Research. 2016;9(6):230–40.
  163. Chen J, Lu X, Chen P, Shen Y, Zheng B, Guo Z. Anti-fatigue effect of glycoprotein from hairtail (Trichiurus lepturus) by-products in a behavioral mouse model. Food Chemistry: X. 2023 Jun;18:100645. [CrossRef]
  164. Squillacioti G, Guglieri F, Colombi N, Ghelli F, Berchialla P, Gardois P, Bono R. Non-Invasive Measurement of Exercise-Induced Oxidative Stress in Response to Physical Activity. A Systematic Review and Meta-Analysis. Antioxidants. 2021 Dec 17;10(12):2008. [CrossRef]
  165. Clarkson PM, Thompson HS. Antioxidants: what role do they play in physical activity and health? The American Journal of Clinical Nutrition. 2000 Aug;72(2):637S-646S.
  166. Enioutina EY, Job KM, Sherwin CMT. Spotlight Commentary: Why we need to pay attention to toxicity associated with herbal medicines. Brit J Clinical Pharma. 2020 Sep;86(9):1793–4. [CrossRef]
  167. Zhao H peng, Zhang Y, Liu Z, Chen J yue, Zhang S yan, Yang X dong, Zhou H li. Acute toxicity and anti-fatigue activity of polysaccharide-rich extract from corn silk. Biomedicine & Pharmacotherapy. 2017 Jun;90:686–93.
  168. Lynch H, Johnston C, Wharton C. Plant-Based Diets: Considerations for Environmental Impact, Protein Quality, and Exercise Performance. Nutrients. 2018 Dec 1;10(12):1841. [CrossRef]
  169. Om AS, Song YN, Noh G, Kim H, Choe J. Nutrition Composition and Single, 14-Day and 13-Week Repeated Oral Dose Toxicity Studies of the Leaves and Stems of Rubus coreanus Miquel. Molecules. 2016 Jan 8;21(1):65.
  170. Ajibade TO, Arowolo R, Olayemi FO. Phytochemical screening and toxicity studies on the methanol extract of the seeds of moringa oleifera. Journal of Complementary and Integrative Medicine [Internet]. 2013 Jan 12 [cited 2023 Aug 7];10(1). Available from: https://www.degruyter.com/document/doi/10.1515/jcim-2012-0015/html.
  171. Osman H, Me S, Em B, Ao F, Ahmed MM, Osman B, Elhassan M, Kk T. Assessment of acute toxicity and LD50 of Moringa oleifera ethanolic leave extract in albino rats and rabbits. 2015.
  172. Pareek A, Pant M, Gupta MM, Kashania P, Ratan Y, Jain V, Pareek A, Chuturgoon AA. Moringa oleifera: An Updated Comprehensive Review of Its Pharmacological Activities, Ethnomedicinal, Phytopharmaceutical Formulation, Clinical, Phytochemical, and Toxicological Aspects. IJMS. 2023 Jan 20;24(3):2098.
  173. Shafi S, Tabassum N. Toxicity evaluation of hydro-alcoholic extract of portulaca oleracea (whole plant) in swiss albino mice. International Journal of Pharmacy and Pharmaceutical Sciences. 2014;7(2).
  174. Basappa G, Kumar V. Toxicity Evaluation of Anisomeles indica Kuntze Leaf Flavonoid Fraction. J Pharmacogn Nat Prod [Internet]. 2016 [cited 2023 Jul 21];2(3). Available from: https://www.omicsonline.org/open-access/toxicity-evaluation-of-anisomeles-indica-kuntze-leaf-flavonoid-fraction-2472-0992-1000122.php?aid=78821.
  175. Sergi CM. Epigallocatechin-3-Gallate Toxicity in Children: A Potential and Current Toxicological Event in the Differential Diagnosis With Virus-Triggered Fulminant Hepatic Failure. Front Pharmacol. 2020 Jan 29;10:1563. [CrossRef]
  176. Lambert JD, Kennett MJ, Sang S, Reuhl KR, Ju J, Yang CS. Hepatotoxicity of high oral dose (−)-epigallocatechin-3-gallate in mice. Food and Chemical Toxicology. 2010 Jan;48(1):409–16. [CrossRef]
  177. Parn KW, Ling WC, Chin JH, Lee SK. Safety and Efficacy of Dietary Epigallocatechin Gallate Supplementation in Attenuating Hypertension via Its Modulatory Activities on the Intrarenal Renin–Angiotensin System in Spontaneously Hypertensive Rats. Nutrients. 2022 Nov 1;14(21):4605. [CrossRef]
  178. Isbrucker RA, Bausch J, Edwards JA, Wolz E. Safety studies on epigallocatechin gallate (EGCG) preparations. Part 1: Genotoxicity. Food and Chemical Toxicology. 2006 May;44(5):626–35.
  179. Mbaveng AT, Hamm R, Kuete V. Harmful and Protective Effects of Terpenoids from African Medicinal Plants. In: Toxicological Survey of African Medicinal Plants [Internet]. Elsevier; 2014 [cited 2023 Dec 24]. p. 557–76. Available from: https://linkinghub.elsevier.com/retrieve/pii/B9780128000182000194.
  180. Perestrelo R, Silva C, Fernandes MX, Câmara JS. Prediction of Terpenoid Toxicity Based on a Quantitative Structure–Activity Relationship Model. Foods. 2019 Dec 1;8(12):628. [CrossRef]
  181. Hudson A, Lopez E, Almalki AJ, Roe AL, Calderón AI. A Review of the Toxicity of Compounds Found in Herbal Dietary Supplements. Planta Med. 2018 Jul;84(09/10):613–26. [CrossRef]
  182. Meredith SE, Juliano LM, Hughes JR, Griffiths RR. Caffeine Use Disorder: A Comprehensive Review and Research Agenda. Journal of Caffeine Research. 2013 Sep;3(3):114–30. [CrossRef]
Figure 1. Strucutures of phenolic compounds.
Figure 1. Strucutures of phenolic compounds.
Preprints 95275 g001
Figure 2. Structure of Dehydrosulphurenic acid.
Figure 2. Structure of Dehydrosulphurenic acid.
Preprints 95275 g002
Figure 3. Structure of some alkaloids.
Figure 3. Structure of some alkaloids.
Preprints 95275 g003
Table 1. Anti-fatigue herbals studied.
Table 1. Anti-fatigue herbals studied.
Species names Famillies Properties Parts used References
Acanthopanax senticosus Araliaceae anti-fatigue Root [51]
Acanthopanax senticosus Araliaceae anti-fatigue stem bark [52]
Anisomeles indica Lamiaceae Anti-fatigue Whole plants [53]
Antrodia camphorata Fomitopsidaceae Physical fatigue Fruiting body [48,54]
Astragali Radix and Angelicae Sinensis Radix Antifatigue root [55]
Bambusa tuldoides Munro Gramineae Anti-fatigue [56]
Cannabis sativa L Cannabaceae Anti-fatigue leaves [57]
Castanea mollissima Blume Fagaceae anti-fatigue flowers [58]
Cistanche deserticola Y.C. Ma Orobanchaceae Antifatigue stems [59]
Camellia sinensis (L.) O. Kuntze var. Theacaea Antifatigue [34,40,41]
Cordyceps militaris Cordycipitaceae anti-fatigue [60]
Eriobotrya japonica (Thunb.) Lindl. Rosaceae Physical capacity leave [61]
Fagopyrum tataricum Polygonaceae Antifatigue grain [62]
Fagopyrum esculentum Moench Polygonaceae Antifatigue grain [55]
Glossogyne tenuifolia Anti-Fatigue [63]
Moringa oleifera Lam. Moringaceae antifatigue leave [64]
Morus alba L. (Mulberry ) Moraceae anti-fatigue leave [65]
Panax ginseng CA Meyer Araliaceae anti-fatigue [42,45,46,48]
Panax quinquefolium Araliaceae anti-fatigue [66]
Passiflora edulis Sim Passifloraceae Anti-fatigue fruit epicarp [67]
Paullinia cupana linn Sapindaceae Anti-fatigue [68]
Physalis pubescens L Solanaceae anti-fatigue Fruit [69]
Polygonum cuspidatum Polygonaceae Physical capacity root , rhizome [70]
Portulaca oleracea L. Portulacaceae Anti-fatigue leave [17]
Punica granatum L. Punicaceae physical performance fruit [71]
Radix notoginseng Araliaceae anti-fatigue Root [72]
Rhodiola rosea Crassulaceae Antifatigue root [30,32,34,35,39,73,74]
Rhododendron adamsii Rehder Ericaceae physical performance Leave [75]
Rubus coreanus Miquel Rosaceae Antifatigue Fruit [76]
Senna siamea Polygonaceae Antifatigue leaves [77]
Siraitia grosvenorii Cucurbitaceae anti-fatigue. Fruits [78]
Taraxacum officinale Asteraceae physical fatigue [11]
Toona sinensis Roemor Meliaceae antifatigue leavef [79]
Trigonella foenum-graecum L. Fabaceae anti-fatigue Dry seeds [80]
Withania somnifera Solanaceae physical performance root [81]
Zea mays L. Poaceae anti-fatigue Corn silk [82]
Citrullus lanatus cv Cucurbitaceae Sore Muscle Relief in Athletes watermelon juice [83]
Table 4. Methodology used for anti-fatigue phyto molecules identification.
Table 4. Methodology used for anti-fatigue phyto molecules identification.
flavonoid Identification technical Compounds identified Plant species Ref.
Phenolic HPLC -RP Resveratrol Passiflora edulis [122]
Phenolic HPLC - MS/MS salidroside, rhodiolosides, luteolin, catechin, quercetin,quercitrin, sacranoside Rhodiola rosea [123]
Phenolic HPLC - MS castanolB Castanea mollissima [124]
Phenols UHPLC Chlorogenic acid, gentisic acid, rutin, p-coumaric acid; m-coumaric acid, protocatechuic acid; p-hydroxybenzoic P. ginseng [125]
Flavonoid H-NMR, 13C-NMR, HMBC, HMQC and ESI-Q-TOF MS Kaempferol Castanea mollissina Blume [126]
Flavonoids HUVEC/CMC-LC-MS puerarin, daidzin, pueroside D and 3'-hydroxypuerarin Puerariae Radix [127]
Flavonoids HPLC, ESI-MS and NMR epigallocatechin, epicatechin, EGCG and epicatechin gallate C. sinensis fruits [128]
Flavonoids PLC-MS/MS analysis myricetin-3-O-glucoside, quercetin-3-O-rutinoside, kaempferol Castanea mollissima flower
anthocyanins HPLC/UPLC-MS/MS analysis cyanidin-3-O-glucoside ; cyanidin-3-O-rutinoside, peonidin 3-O-glucoside Passiflora edulis Sims fruits [67]
Terpenoids UPLC-Q-TOF-MS Kankanoside E , Daucosterol, Diosgenin Cistanche deserticola YC Ma [129]
Terpenoids LC electrospray mass spectrometric Ginsenosides Panax ginseng [130]
TERPENOID HR-FAB-MS, 1D, 2D-NMR and CD MS kauranoid diterpene glycoside mollioside nuts of Castanea mollissima Blume [131]
terpenoids HPLC analysis. lupeol, stigmasterol, β-sitosterol, and squalene seeds of Panax ginseng [132]
terpenoids NMR and MS analyses antcins B, C, H, I, K (ergostane-type triterpenoids) and dehydrosulphurenic acid, 15α-acetyldehydrosulphurenic acid (lanostane-type triterpenoids). fruiting of Antrodia camphorata [54]
terpenoids GC–MS analysis. Farnesyl acetone, nootkatone and jasmatone leaf Anisomeles indica Kuntze essential oil [133]
allkaloid RP-HPLC caffeine and theophylline seeds of Paulinia cupana Kunth [134]
allkaloid DART -TOF-MS capsaicin Capsicum annuum
allkaloid RP-HPLC capsaicin Capsicum annuum [135]
Alkaloid LC-MS analysis Sparteine, papaverine, Caffeine, naloxone Bambusa. tuldoides [136]
alkaloid HPLC-ESI-MS- NMR caffeine Camellia sinensis fruits [128]
alkaloid HPLC-DAD Theophiline; Caffeine Camellia sinensis
leaves
[137]
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.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.

Downloads

517

Views

112

Comments

0

Subscription

Notify me about updates to this article or when a peer-reviewed version is published.

Email

Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

© 2025 MDPI (Basel, Switzerland) unless otherwise stated