Next Article in Journal
Feasibility and Acceptability of Photographic Food Record, Food Diary and Weighed Food Record in People with Serious Mental Illness
Previous Article in Journal
Lactobacillus plantarum GKM3 Promotes Longevity, Memory Retention, and Reduces Brain Oxidation Stress in SAMP8 Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Plant Adaptogens—History and Future Perspectives

by
Velislava Todorova
1,*,
Kalin Ivanov
1,
Cédric Delattre
2,3,
Vanya Nalbantova
1,
Diana Karcheva-Bahchevanska
1 and
Stanislava Ivanova
1
1
Department of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria
2
Université Clermont Auvergne, Clermont Auvergne INP, CNRS, Institut Pascal, 63000 Clermont-Ferrand, France
3
Institut Universitaire de France (IUF), 1 rue Descartes, 75005 Paris, France
*
Author to whom correspondence should be addressed.
Nutrients 2021, 13(8), 2861; https://doi.org/10.3390/nu13082861
Submission received: 11 July 2021 / Revised: 12 August 2021 / Accepted: 17 August 2021 / Published: 20 August 2021
(This article belongs to the Section Phytochemicals and Human Health)

Abstract

:
Adaptogens are synthetic compounds (bromantane, levamisole, aphobazole, bemethyl, etc.) or plant extracts that have the ability to enhance the body’s stability against physical loads without increasing oxygen consumption. Extracts from Panax ginseng, Eleutherococcus senticosus, Rhaponticum carthamoides, Rhodiola rosea, and Schisandra chinensis are considered to be naturally occurring adaptogens and, in particular, plant adaptogens. The aim of this study is to evaluate the use of plant adaptogens in the past and now, as well as to outline the prospects of their future applications. The use of natural adaptogens by humans has a rich history—they are used in recovery from illness, physical weakness, memory impairment, and other conditions. About 50 years ago, plant adaptogens were first used in professional sports due to their high potential to increase the body’s resistance to stress and to improve physical endurance. Although now many people take plant adaptogens, the clinical trials on human are limited. The data from the meta-analysis showed that plant adaptogens could provide a number of benefits in the treatment of chronic fatigue, cognitive impairment, and immune protection. In the future, there is great potential to register medicinal products that contain plant adaptogens for therapeutic purposes.

1. Introduction

Adaptogens are pharmacologically active compounds or plant extracts from different plant classes (for example: Araliaceae—Panax ginseng, Eleutherococcus senticosus, Asteraceae—Rhaponticum carthamoides, Crassulaceae—Rhodiola rosea, and Schisandraceae—Schisandra chinensis) [1,2,3]. They have the ability to enhance the body’s stability against physical loads without increasing oxygen consumption. The intake of adaptogens is associated not only with the body’s better ability to adapt to stress and maintain/normalize metabolic functions, but also with better mental and physical performance [1,2,3].
There are two main classes of adaptogens. The first class includes plant adaptogens, while the other includes synthetic adaptogens, which are also called actoprotectors.
Although plant adaptogens have been used by people since ancient times, the term “adaptogen” is relatively young—it was introduced in 1947 by the Soviet scientist Lazarev [1,4]. It defines adaptogens as substances that cause non-specific resistance of the living organisms [1,2,4]. Adaptogens have a positive effect on humans and animals. The use of plant adaptogens has a rich history. They have been used by man for hundreds of years in different parts of the world, while data on the use of the first synthetic adaptogen, bemethyl, date back to the 1970s. Bemethyl was introduced in the 1970s by Professor Vladimir Vinogradov. Since then, numerous synthetic adaptogens have been developed: bromantane, levamisole, aphobazole, chlodantane, trekrezan. Their intake is associated not only with increased physical and mental resistance, but also with vasodilation and decreased blood sugar and lactate [3]. They are widely used in sports medicine, but since 2009, WADA has included bromantane in the prohibited list and since 2018, bemethyl has also been included in the monitoring program of WADA [5,6].
In 1980, the scientists Breckham and Dardimov found that adaptogens increase the body’s resistance not only to physical but also to chemically and biologically harmful agents [1,2,7], which further expands the potential of their use.
Breckham and Dardimov systematized the plants with adaptogenic properties. These are: Panax ginseng, Eleutherococcus senticosus, Rhaponticum carthamoides, Rhodiola rosea, and Schisandra chinensis [7].
Although plant adaptogens have been used for centuries, their effects continue to be studied to this day. They also have promising potential for wider applications in the future.
The biological effects of plant adaptogens are related to the complex of biologically active compounds they contain. Plant adaptogens have very rich phytochemical composition. Some of the most important phytochemicals with adaptogenic properties are: triterpenoid saponins (in Panax ginseng—ginsenosides; in Eleutherococcus senticosus—eleutherosides); phytosterols and ecdysone (in Rhaponticum carthamoides); lignans (in Schisandra chinensis); alkaloids; flavonoids, vitamins, etc. [4,8,9].
The mechanism of action of the plant adaptogens is complex and is not fully understood. Recent studies report that the intake of plant adaptogens like extracts of Eleutherococcus senticosus root, Schisandra chinensis root, Rhodiola rosea root is associated with affecting the hypothalamic—pituitary—adrenal axis [10] and some stress mediators [11]. Moreover, the intake of such extracts affects nitric oxide levels, lactate levels, blood glucose levels, cortisol levels, plasma lipid profile, hepatic enzymes, etc. [8,11,12,13,14].
Current and potential uses of these medicinal plants are associated with mental diseases and behavioural disorders, cognitive function and stress-induced diseases (anxiety, cardiovascular diseases, diabetes) [4,9,15,16,17,18,19,20]. The intake of plant adaptogens is not associated with serious side effects [20,21].
The aim of our study is to evaluate the data about the use of adaptogens in the past and the future perspectives of their use.

2. Materials and Methods

The first step of the screening process included identifying eligible studies. An expanded search for articles without language restrictions was conducted on the following databases: Google Scholar, PubMed, and Web of Science. In the search process, the following search key words were used: “adaptogens”, “actoprotectors”, “plant adaptogens”, “Panax ginseng”, “Eleutherococcus senticosus”, “Schisandra chinensis”, “Rhodiola rosea”, “Rhaponticum carthamoides”, “Leuzea carthamoides”, “ginseng adaptogen properties study”, “Eleutherococcus senticosus adaptogen properties study”, “randomized controlled study Eleutherococcus senticosus”, “Leuzea carthamoides adaptogen properties study”, “Rhaponticum carthamoides adaptogen properties study”, "ecdysterone adaptogen properties study”, “20-hydroxyecdisone adaptogen study”, “ecdysterone from Leuzea adaptogen studies”, “Leuzea carthamoides adaptogen properties in rats”, “performance gain Rhaponticum carthamoides”, “Rhodiola rosea in sport”, “Rhodiola rosea adaptogen properties study”, “Schisandra chinensis in sport”, “Schisandra chinensis randomized study”, “Schisandra chinensis adaptogen properties study”, and “Schisandra chinensis mice study”.
Further records concerned the history of plant adaptogens, botanical and phytochemical characteristics, and application of adaptogens.
In the second step, we followed the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analysis) presented in Figure 1 [22], guide for a systematic review.
In the third step, the relevant studies were chosen based on exclusion and inclusion criteria. The exclusion criteria were: (i) articles written in a language other than English or Russian; (ii) webinars or blogs; (iii) articles with irrelevant topics, and (iv) lack of data information. Inclusion criteria were: (i) human studies; (ii) animal studies, and (iii) studies investigating the impacts of adaptogens.
During the fourth step, the selected full articles were read and identified.
In total, 56 studies were selected and included in the present review.

3. Results and Discussion

3.1. Panax Ginseng

The first evidence of the use of Panax ginseng (Panax ginseng C.A. Mey.) dates back more than 2000 years [23]. In the past, the stems, leaves, and mainly the roots of ginseng were used. Extracts were prepared and used to maintain homeostasis in the human body, treat fatigue and weakness, increase immune protection, and treat hypertension, diabetes type 2, and erectile dysfunction [2,23,24,25,26]. In Chinese traditional medicine, ginseng extracts have also been used as nootropic agents and as a tonic [24,25,26]. The exact mechanism of adaptogenic action of Panax ginseng is unknown, but it is supposed that it affects the hypothalamic-pituitary-adrenal axis in Figure 2 and the antioxidant effect [24,27,28,29].
Panax ginseng is an example of a medicinal plant, widely used in ancient times, but also with great application today. It is also one of the plants defined as a natural adaptogen.
Panax ginseng has a rich botanical phytochemical composition. Today, more than 200 substances isolated from Korean ginseng are known, ginsenosides predominantly: Ra1, Ra2, Ra3, malonyl-G-Rb1, malonyl-G-Rb2, malonyl-G-Rc, malonyl-GRd, Rs1, Rs2, Rs3, Rg3, Rg5, Rh2, K-R2, Rf, Rf2, 20(R)-G-Rg2, Rg6, 20(R)-G-Rh1, 20(E)-G-F4, Rh4, K-R1, and poly-acetyleneginsenoside-Ro [30].
It is considered that the adaptogenic properties of Panax ginseng extract are due to the ginsenosides [27].
Nowadays, numerous products containing ginseng extracts are available. Most of these products are sold as food supplements but there are also many over-the-counter medicines. Ginseng radix is included in European Pharmacopeia and in the US Pharmacopeia. The principal root has a cylindrical form, sometimes branched, up to about 20 cm long and 2.5 cm in diameter. Its surface is pale yellow/cream in white ginseng, while it is brownish-red in red ginseng. The rootlets, which are many in the lower part of white ginseng, are usually absent in red ginseng. Reduced to a powder, it is light yellow. Ginseng dry extract is produced from the root by a suitable procedure using a hydroalcoholic solvent equivalent in strength to ethanol. The main compounds that can be detected in the extract are: ginsenoside Rg1, ginsenoside Re, ginsenoside Rf, ginsenoside Rb1, ginsenoside Rc, ginsenoside Rd, ginsenoside Rb2 [31].
Although the extract has been used for more than two millenniums, there are a limited number of clinical studies that have investigated the benefits or the side effects of its use. More double-blind randomized studies are needed to be performed.
In Table 1, we have summarized the main benefits of the Panax ginseng extract intake.
The data suggest that an intake of Panax ginseng extracts is associated with ergogenic effects and enhanced muscle strength. The inclusion of Panax ginseng in the diet of athletes would help increase the body’s physical resilience and help the body recover between workouts.
Intake of Panax ginseng extract is associated also with improved plasma lipid profile and blood glucose level. Extracts of Panax ginseng might be included in the diet of patients with cardiovascular diseases, hyperlipidemia, and diabetes. Extracts from the plant not only increase cognitive function and memory functions, but also improve sleep and fatigue.
We did not find multicenter randomized double-blind studies including the intake of Panax ginseng. For better exploration of the benefits and future applications of the Panax ginseng extract, multicenter randomized double-blind studies should be performed. The plant has a great potential to be included in medicines for treatment of different conditions.
The intake of Panax ginseng extract is not associated with serious side effects [21,32,33,34,35,36,37,38,39,40,41]. The safety of the extract is another benefit to be considered.

3.2. Eleutherococcus Senticosus

Siberian ginseng (Eleutherococcus senticosus ((Rupr. and Maxim.) Maxim.) was first described by Porfiry Kirilov in the 19th century [42]. Its adaptogenic effects were widely studied in Russia between 1960 and 1970 [43]. The first data on the use of plant extracts in athletes were reported from Russia. Today, plant extracts are used not only by athletes around the world, but also by many other consumers who are not actively involved in sports.
The phytochemical composition consists of phenylpropanoid—syringin; lignans—sesamin; saponins—daucosterol; coumarins, terpenoids, flavonoids, organic acids, and vitamins [44,45]. Extracts of Eleutherococcus senticosus root are obtained, stimulating the immune system, influencing adaptation against external factors, improving mental and physical conditions and memory functions, and have a hypoglycaemic effect and are anti-inflammatory [13,45,46]. Eleutherococcus senticosus is thought to exert its adaptogenic effects by influencing the hypothalamic-pituitary-adrenal axis [44].
Most of the extracts of Eleutherococcus senticosus are prepared by the roots. The rhizome of Eleutherococcus senticosus is described in European pharmacopeia. The rhizome has a diameter of 1.5 to 4 cm and an irregular cylindrical shape. The surface is longitudinally wrinkled with grayish-brown to blackish-brown colour [47].
Although Eleutherococcus senticosus were described a few hundred years ago, research into its use continues. In Table 2, we summarized results of studies with Eleutherococcus senticosus.
The data suggest that the intake of Eleutherococcus senticosus supports the physical activity, weight reduction, mental health, and fatigue. Extracts of the plant can be used only for fatigue, but also in the case of disturbed sleep.
The intake of Eleutherococcus senticosus extract may help to increase the cognitive function. Siberian ginseng extract could be included in the diet of patients with hyperlipidemia because it has a beneficial effect on the lipid profile. There is a potential for development of medicinal products containing Eleutherococcus senticosus extract to be taken by patients with various pathologies such as: obesity, overweight, hyperlipidemia, etc. New nootropic drugs containing a standardized plant extract could also be registered.
We did not find multicenter randomized double-blind studies that included the intake of Siberian ginseng. For better exploration of the benefits and future applications of the Eleuterococcus senticosus extract, multicenter randomized double-blind studies should be performed. The plant has a great potential to be included in medicines for treatment of different conditions; moreover, the intake of Eleuterococcus senticosus extract is not associated with serious side effects [46,48,49,50,51,52,53,54,55].

3.3. Rhaponticum Carthmoides

Rhaponticum carthamoides (Rhaponticum carthamoides IIjin.) is a perennial herb which has been used for centuries in Russia, China, and Mongolia [14]. The plant is also known as Leuzea. Extracts of the plant have been used to treat weakness [14,56], lung diseases, kidney diseases, fever, and angina [57].
In 1969, Brekhman and Dardymov classified this plant as an adaptogen [14]. The use of products containing Rhaponticum carthamoides root extract has increased in recent decades. The extract has many beneficial effects on humans like: enhanced physical endurance and performance, anabolic effect, hypocholesterolemic effects, neuroprotective effect, antidiabetic properties, anti-oxidative, and increased immunity [14,57]. The mechanism by which ecdysteroids act is binding with signal transduction pathways, rather than with steroid and estrogen receptors [58].
In the 1970s, the extracts from Leuzea showed beneficial effects in athletes and their use became a routine practice in the training of many athletes. The intake of Leuzea extract increases the body’s adaptation to various factors, which can be defined as stress for the body [14,57] and at the same time has a good level of safety.
The phytochemical composition of Rhaponticum carthamoides is rich of ecdysteroids and phenols. The main ecdysteroid is 20-hydroxyecdysone in Figure 3 [14]. The adaptogenic properties of the extract are related mainly to its presence [14]. Other components of the phytochemical composition are phenols and essential oil [14].
20-hydroxyecdysone has a typical steroidal structure. A recent study, funded by WADA and conducted in 2019, reported 20-hydroxyecdisone as a non-conventional anabolic agent, which can significantly increase the muscle mass. A significant dose-responsive anabolic effect of 20-hydroxyecdysone was reported [59].
In 2020, 20-hydroxyecdysone was included in the WADA monitoring program and it is very likely that the substance will be included in the prohibited list in the next few years [60]. In the past few years, other adaptogens were included in WADA’s prohibited list and monitoring program, because these compounds were considered to improve the performance of the athletes. Bromantane is included in the prohibited list as a non-specified stimulant. Since 2018, bemethyl is monitored by WADA, but it is still not included on the prohibited list [5,6]. The main difference between 20-hydroxyecdysone and these two compounds is that 20-hydroxyecdysone is a natural compound while the other two are synthetic adaptogens. The main reason 20-hydroxyecdysone is monitored by WADA is that the intake of this compound improves the performance of athletes. Nowadays, food supplements containing Leuzea (which contain 20-hydroxyecdysone) are often included in the supplementation of professional athletes.
Table 3 presents studies related to the use and effects of Rhaponticum carthamoides.
We expanded our search with animal studies; found results are presented in Table 4.
The data suggest that intake of Rhaponticum carthamoides extracts is associated with anabolic effects—increased body mass weight and enhanced muscle strength. Other important benefits are improved mental endurance and improved plasma lipid profiles. Improvement of cardiac and cognitive functions has also been reported.
Because of improvement in cardiac functions, the Rhaponticum carthamoides extract might be especially useful for patients with cardiovascular diseases.
Despite the benefits of the extract use, the studies on humans are limited in number and not sufficient for a more complete and comprehensive assessment.
The anabolic effects were also reported in animal studies. The data from animal studies suggests that application of Rhaponticum carthamoides is also associated with a neuroprotective effect.
The intake of Rhaponticum carthamoides is not associated with serious side effects [19,59,60,61,62,63,64,65,66,67,68,69,70,71,72].
The data obtained from human and animal studies about synergetic anabolic effect of Rhaponticum carthamoides and Rhodiola rosea are contradictory. Further studies are needed to confirm the synergetic effect of these adaptogenic plants.
For better exploration of the benefits and future applications of the Rhaponticum carthamoides extract, more in vivo studies and multicenter randomized double-blind studies should be performed.

3.4. Rhodiola rosea

In traditional medicine, its (Rhodiola rosea L.) applications are described as an adaptive agent that increases physical endurance, affects fatigue, depression, and disorders of the nervous system. It has been used in the past in Asia to treat flu and colds, and there is reported use in tuberculosis. In the Scandinavian part of Europe, plant extracts have been used to increase physical endurance [73].
Six groups of compounds predominate in the phytochemical composition of the plant: phenylpropanoids, phenylethanol derivatives, flavonoids, phenolic acids, and mono- and triterpene [74].
The main phenylethanol derivatives are salidroside (rhodioloside), para-tyrosol, and phenylpropanoid-rosavin. These are also responsible for the adaptogenic and ergogenic effects of Rhodiola rosea [8,73,75].
The adaptogenic effect of Rhodiola rosea is associated with activation of the cerebral cortex by increasing norepinephrine and serotonin levels. In addition, it affects the hypothalamic-pituitary-adrenal axis, reducing the levels of corticotropin-releasing hormones, corticotropin, cortisol, and epinephrine [29,73,76].
Studies of Rhodiola rosea began with Dioscorides and continues nowadays. In Table 5, we summarized beneficial effects of Rhodiola rosea based on studies.
The data suggest that the intake of the extract of Rhodiola rosea is associated with antioxidant and adaptogen properties. Rhodiola rosea extract might be used not only for overcoming the fatigue but might be included in the diet of people with heart diseases, because of the beneficial effect on heart rate and muscle contractions. The use of plant extracts is recommended for sleep disorders and anxiety. The hepatoprotective effect of the plant determines the use of its extract in liver diseases. Improving physical strength during exercise and recovery after training are the reasons why Rhodiola rosea extracts are also taken by athletes as a supplement to their diet.
The intake of Rhodiola rosea is not associated with serious side effects [77,78,79,80,81,82,83,84,85,86].
For better exploration of the benefits and future applications of the Rhodiola rosea extract, more multicenter randomized double-blind studies should be performed. The plant has a great potential to be included in medicines for treatment of different conditions.

3.5. Schisandra chinensis

Schisandra chinensis (Schisandra chinensis (Turcz.) Bail) is first described in the book Shen Nong Ben Cao Jing, around 200 AD, as a remedy for cough and asthma [87]. In the past, Schisandra chinensis fruits and seeds were used to improve night vision, reduce hunger, thirst, and exhaustion. In 1960 in Russia, the adaptogenic properties of the plant were proven [88]. The fruits of Schisandra chinensis are used today [87,88,89,90].
According European Pharmacopoeia, Schisandra berry is more or less spherical, up to 8 mm in diameter. It could be red/reddish-brown/blackish, it could be covered in a whitish frost. It has a strongly shriveled pericarp. The seeds are only 1 or 2, yellowish-brown and lustrous. The seed-coat is thin [91].
For production of the extract, the fruit should be reduced to a powder. The colour of powder should be reddish-brown.
The Schisandra fruit has a complex phytochemical composition in which the lignans are the major characteristic constituents. Five classes of different lignans are found in the fruits of Schisandra: dibenzocyclooctadiene lignans (type A), spirobenzofuranoid dibenzocyclooctadiene lignans (type B), 4-aryltetralin lignans (type C), 2,3-dimethyl-1,4-diarylbutane lignans (type D), and 2,5-diaryltetrahydrofuran lignans (type E) [88].
The adaptogenic properties of Schisandra chinensis are due to the lignin complex, mainly dibenzocycloocstadiene lignans, the main schisandrin [92,93]. Schizandrin was isolated and identified for the first time by N.K. Kochetkov in 1961 [88,92,93].
One of methods for identification of the plant, described in the European Pharmacopeia, involves a TLC technique with identification of γ-Schisandrin in Figure 4.
Several studies reported that some lignans (gomisin A, gomisin G, schizandrin, and schisanhenol) possess antitumor bioactivities [94,95,96,97,98].
The Schisandra fruit, particularly the seeds, contains many volatile compounds as well: α-ylangene, α-cedrene, β-chamigrene, and β-himachalene [99,100].
From Schisandra chinensis fruit are also isolated polysaccharides, glycosides (dihydrophaseic acid-3-O--d-glucopyranoside, benzyl alcohol-O--d-glucopyranosyl (1→6)--d-glucopyranoside and benzyl alcohol-O--d-glucopyranosyl (1→2)--d-glucopyranoside), organic acids (vitamin C, malic acid, citric acid, and tartaric acid), and vitamin E. [12,87,101,102]. In small quantities, the fruit of Schisandra chinensis contained flavonoids such as rutine [103]. Preschsanartanin, schintrilactones A–B, schindilactones A–C, and wuweizidilactones A–F are the new isolated triterpenoids from Schisandra chinensis fruit [88].
All of phytochemicals determined beneficial effects such as: cytotoxic, antioxidant, neuroprotective, hepatoprotective, increased physical strength, stress-protective, anti-inflammatory [12,88,104]. The adaptogenic effect of Schisandra chinensis is associated with the antioxidant effect and the influence on the hypothalamic-pituitary-adrenal axis by lowering the level of corticotropin-releasing hormone [29,76].
Research on the effects of Schisandra chinensis continues.
In Table 6, we present summarized data about beneficial effects of Schisandra chinensis.
Discovered trials are insufficient to summarize the use of Schisandra chinensis. We expanded our search with animal studies, the results found are presented in Table 7.
The data from animal and human studies suggest that the application of extract of Schisandra chinensis is associated with adaptogenic, antioxidant properties, tonic, and stress-protective effect.
The intake of plant extract can improve memory and concentration. The plant extract has serious potential to be included in medicinal products for use in the treatment of patients with hypercholesterolemia or patients with cardiovascular disease. Of course, the application of the extract would have a number of benefits in healthy patients due to its pronounced antioxidant properties.
The positive effect of the plant on blood sugar and liver enzymes suggests the inclusion of Rhodiola rosea extracts in the treatment of patients with diabetes and liver disease. In recent decades, Schisandra chinensis extract has been taken by athletes to increase physical activity and the body’s adaptation to stress.
The studies on humans are limited in number, so a full assessment of the effects is difficult to make.
The intake of Schisandra chinensis is not associated with serious side effects [105,106,107,108,109,110,111,112,113,114,115,116].
For a better exploration of the benefits and future applications of the Schisandra chinensis extract, more in vivo studies and multicenter randomized double-blind studies should be performed.

4. Conclusions

The natural adaptogens have the ability to increase the body’s resistance to stress changes caused by different types of stressors. Unlike the synthetic adaptogens, the natural are extracts with an extremely rich phytochemical composition. Their adaptogenic properties are not due to one molecule, but to the combination of different substances. The use of natural adaptogens by humans has a rich history—they have been used in recovery from illness, physical weakness, impaired mental function, and other conditions. For about 50 years, plant adaptogens have been used by professional athletes due to their high potential to increase the body’s resilience and improve physical endurance. Nowadays, some of the most used plant adaptogens are Panax ginseng, Eleutherococcus senticosus, and Rhaponticum carthamoides. Since 2020, ecdysterone, which is rich in Leuzea extract, has been included in WADA’s monitoring program, with prospects for inclusion in the prohibited list. Studies examining the benefits of using extracts of Rhodiola rosea, Eleutherococcus senticosus, Panax ginseng, Schisandra chinensis and Rhaponticum carthamoides are a limited in number. However, there are potentials for the inclusion of the extracts of these plants in medicinal products aimed at treating chronic fatigue, cognitive impairment, as well as boosting immune defenses. Double-blind randomized multicenter studies would be extremely valuable in evaluating the use of the extracts in patients with cardiovascular disease, in patients with compromised immunity, and in patients with chronic fatigue.

Author Contributions

Conceptualization, V.T., K.I. and S.I.; data curation, V.T. and S.I.; writing—original draft preparation, V.T., K.I., C.D., V.N., D.K.-B. and S.I.; writing—review and editing, K.I., C.D., V.N., D.K.-B. and S.I.; visualization, V.T., K.I., V.N., D.K.-B. and S.I.; supervision, K.I. and C.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wagner, H.; Nörr, H.; Winterhoff, H. Plant adaptogens. Phytomedicine 1994, 1, 63–76. [Google Scholar] [CrossRef]
  2. Panossian, A.; Wikman, G.; Wagner, H. Plant adaptogens III. Earlier and more recent aspects and concepts on their mode of action. Phytomedicine 1999, 6, 287–300. [Google Scholar] [CrossRef]
  3. Oliynyk, S.; Oh, S.-K. The pharmacology of Actoprotectors: Practical application for improvement of mental and physical performance. Biomol. Ther. 2012, 20, 446–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Panossian, A.G.; Efferth, T.; Shikov, A.N.; Pozharitskaya, O.N.; Kuchta, K.; Mukherjee, P.K.; Banerjee, S.; Heinrich, M.; Wu, W.; Guo, D.; et al. Evolution of the adaptogenic concept from traditional use to medical systems: Pharmacology of stress and aging related diseases. Med. Res. Rev. 2020, 41, 630–703. [Google Scholar] [CrossRef]
  5. The World Anti-Doping Agency—WADA. Executive Committee Approved the List of Prohibited Substances and Methods for 2009. Available online: https://www.wada-ama.org/en/media/news/2008-09/wada-executive-committee-approves-2009-prohibited-list-new-delhi-laboratory-0 (accessed on 1 May 2021).
  6. The World Anti-Doping Agency—WADA. Prohibited List. 2018. Available online: https://www.wada-ama.org/sites/default/files/prohibited_list_2018_en.pdf (accessed on 1 May 2021).
  7. Brekhman, A.I.; Dardymov, I.V. New substances of plant origin which increase nonspecific resistance. Annu. Rev. Pharmacol. 1969, 9, 419–430. [Google Scholar] [CrossRef]
  8. Kelly, G.S. Rhodiola rosea: A possible plant adaptogen. Altern. Med. Rev. 2001, 6, 293–302. [Google Scholar]
  9. Kamal, M.; Arif, M.; Jawaid, T. Adaptogenic medicinal plants utilized for strengthening the power of resistance during chemotherapy—A review. Orient. Pharm. Exp. Med. 2017, 17, 1–18. [Google Scholar] [CrossRef]
  10. Panossian, A.; Wikman, G.; Kaur, P.; Asea, A. Adaptogens exert a stress-protective effect by modulation of expression of molecular chaperones. Phytomedicine 2009, 16, 617–622. [Google Scholar] [CrossRef]
  11. Pawar, V.S.; Shivakumar, H. A current status of adaptogens: Natural remedy to stress. Asian Pac. J. Trop. Dis. 2012, 2, S480–S490. [Google Scholar] [CrossRef]
  12. Li, Z.; He, X.; Liu, F.; Wang, J.; Feng, J. A review of polysaccharides from Schisandra chinensis and Schisandra sphenanthera: Properties, functions and applications. Carbohydr. Polym. 2018, 184, 178–190. [Google Scholar] [CrossRef]
  13. Hikino, H.; Takahashi, M.; Otake, K.; Konno, C. Isolation and Hypoglycemic Activity of Eleutherans A, B, C, D, E, F, and G: Glycans of Eleutherococcus senticosus Roots. J. Nat. Prod. 1986, 49, 293–297. [Google Scholar] [CrossRef]
  14. Kokoska, L.; Janovska, D. Chemistry and pharmacology of Rhaponticum carthamoides: A review. Phytochemistry 2009, 70, 842–855. [Google Scholar] [CrossRef]
  15. Panossian, A. Understanding adaptogenic activity: Specificity of the pharmacological action of adaptogens and other phytochemicals. Ann. N. Y. Acad. Sci. 2017, 1401, 49–64. [Google Scholar] [CrossRef]
  16. Mendes, F.R.; Carlini, E. Brazilian plants as possible adaptogens: An ethnopharmacological survey of books edited in Brazil. J. Ethnopharmacol. 2007, 109, 493–500. [Google Scholar] [CrossRef]
  17. Ajala, T.O. The effects of adaptogens on the physical and psychological symptoms of chronic stress. DISCOV. Ga. State Honor. Coll. Undergrad. Res. J. 2017, 4, 2. [Google Scholar] [CrossRef]
  18. Domene, A.M. Effects of adaptogen supplementation on sport performance. A recent review of published studies. J. Hum. Sport Exerc. 2013, 8, 1054–1066. [Google Scholar] [CrossRef] [Green Version]
  19. Krasutsky, A.G.; Cheremisinov, V.N. The use of Levzey’s extract to increase the efficiency of the training process in fitness clubs students. In Proceedings of the Actual Problems of Biochemistry and Bioenergy of Sport of the XXI Century, Moscow, Russia, 10–26 April 2017; pp. 382–388. [Google Scholar]
  20. Aslanyan, G.; Amroyan, E.; Gabrielyan, E.; Nylander, M.; Wikman, G.; Panossian, A. Double-blind, placebo-controlled, randomised study of single dose effects of ADAPT-232 on cognitive functions. Phytomedicine 2010, 17, 494–499. [Google Scholar] [CrossRef]
  21. Reay, J.L.; Scholey, A.; Kennedy, D. Panax ginseng (G115) improves aspects of working memory performance and subjective ratings of calmness in healthy young adults. Hum. Psychopharmacol. Clin. Exp. 2010, 25, 462–471. [Google Scholar] [CrossRef]
  22. Page, M.J.; McKenzie, J.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.; Brennan, S.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, 71. [Google Scholar] [CrossRef]
  23. Baeg, I.-H.; So, S.-H. The world ginseng market and the ginseng (Korea). J. Ginseng Res. 2013, 37, 1–7. [Google Scholar] [CrossRef] [Green Version]
  24. Kiefer, D.S.; Pantuso, T. Panax ginseng. Am. Fam. Physician 2003, 68, 1539–1542. [Google Scholar]
  25. Patel, S.; Rauf, A. Adaptogenic herb ginseng (Panax) as medical food: Status quo and future prospects. Biomed. Pharmacother. 2016, 85, 120–127. [Google Scholar] [CrossRef] [PubMed]
  26. Shergis, J.; Zhang, A.L.; Zhou, W.; Xue, C.C. Panax ginseng in randomised controlled trials: A systematic review. Phytother. Res. 2012, 27, 949–965. [Google Scholar] [CrossRef]
  27. Nocerino, E.; Amato, M.; Izzo, A. The aphrodisiac and adaptogenic properties of ginseng. Fitoterapia 2000, 71, S1–S5. [Google Scholar] [CrossRef]
  28. Mahady, G.B.; Gyllenhaal, C.; Fong, H.H.; Farnsworth, N.R. Ginsengs: A review of safety and efficacy. Nutr. Clin. Care 2000, 3, 90–101. [Google Scholar] [CrossRef]
  29. Wilson, L. Review of adaptogenic mechanisms: Eleuthrococcus senticosus, panax ginseng, rhodiola rosea, schisandra chinensis and withania somnifera. Aust. J. Med. Herbal. 2007, 19, 126–138. [Google Scholar] [CrossRef]
  30. Christensen, L.P. Ginsenosides: Chemistry, biosynthesis, analysis, and potential health effects. Adv. Food Nutr. Res. 2008, 55, 1–99. [Google Scholar] [CrossRef]
  31. European Directorate for the Quality of Medicines & Health Care. Ginseng radix. In European Pharmacopoeia, Monograph 07/2019:1523; European Directorate for the Quality of Medicines & Health Care: Strasburg, France, 2019. [Google Scholar]
  32. Kim, S.H.; Park, K.S. Effects of panax ginseng extract on lipid metabolism in humans. Pharmacol. Res. 2003, 48, 511–513. [Google Scholar] [CrossRef]
  33. Bhattacharjee, I.; Bandyopadhyay, A. Effects of acute supplementation of panax ginseng on endurance performance in healthy adult males of Kolkata, India. Int. J. Clin. Exp. Physiol. 2020, 7, 63–68. [Google Scholar] [CrossRef]
  34. Etemadifar, M.; Sayahi, F.; Abtahi, S.-H.; Shemshaki, H.; Dorooshi, G.-A.; Goodarzi, M.; Akbari, M.; Fereidan-Esfahani, M. Ginseng in the treatment of fatigue in multiple sclerosis: A randomized, placebo-controlled, double-blind pilot study. Int. J. Neurosci. 2013, 123, 480–486. [Google Scholar] [CrossRef]
  35. Engels, H.-J.; Said, J.M.; Wirth, J.C. Failure of chronic ginseng supplementation to affect work performance and energy metabolism in healthy adult females. Nutr. Res. 1996, 16, 1295–1305. [Google Scholar] [CrossRef]
  36. Perazzo, F.F.; Fonseca, F.L.; Souza, G.H.B.; Maistro, E.L.; Rodrigues, M.; Carvalho, J.C. Double-blind clinical study of a multivitamin and polymineral complex associated with panax ginseng extract (Gerovital®). Open Complement. Med. J. 2010, 2, 100–104. [Google Scholar]
  37. Ziemba, A.W. The effect of ginseng supplementation on psychomotor performance, indices of physical capacity and plasma concentration of some hormones in young well fit men. In Proceedings of the Ginseng Society Conference, Seoul, Korea, 1 October 2002; pp. 145–158. [Google Scholar]
  38. Zarabi, L.; Arazi, H.; Izadi, M. The effects of panax ginseng supplementation on growth hormone, cortisol and lactate response to high-intensity resistance exercise. Biomed. Hum. Kinet. 2018, 10, 8–14. [Google Scholar] [CrossRef] [Green Version]
  39. Lee, S.A.; Kang, S.G.; Lee, H.J.; Jung, K.Y.; Kim, L. Effect of Korean red ginseng on sleep: A randomized, placebo-controlled Trial. Sleep Med. Psychophysiol. 2010, 17, 85–90. [Google Scholar]
  40. Kim, H.-G.; Cho, J.-H.; Yoo, S.-R.; Lee, J.-S.; Han, J.-M.; Lee, N.-H.; Ahn, Y.-C.; Son, C.-G. Antifatigue Effects of Panax ginseng CA Meyer: A randomised, double-blind, placebo-controlled trial. PLoS ONE 2013, 8, e61271. [Google Scholar] [CrossRef] [Green Version]
  41. Ping, F.W.C.; Keong, C.C.; Bandyopadhyay, A. Effects of acute supplementation of Panax ginseng on endurance running in a hot & humid environment. Indian J. Med. Res. 2011, 133, 96–102. [Google Scholar]
  42. Davydov, M.; Krikorian, A. Eleutherococcus senticosus (Rupr. & Maxim.) maxim. (Araliaceae) as an adaptogen: A closer look. J. Ethnopharmacol. 2000, 72, 345–393. [Google Scholar] [CrossRef]
  43. World Health Organization. WHO Monographs on Selected Medicinal Plants; World Health Organization: Geneva, Switzerland, 2002; Volume 2. [Google Scholar]
  44. Bleakney, T.L. Deconstructing an adaptogen: Eleutherococcus Senticosus. Holist. Nurs. Pract. 2008, 22, 220–224. [Google Scholar] [CrossRef]
  45. Jia, A.; Zhang, Y.; Gao, H.; Zhang, Z.; Zhang, Y.; Wang, Z.; Zhang, J.; Deng, B.; Qiu, Z.; Fu, C. A review of Acanthopanax senticosus (Rupr and Maxim.) harms: From ethnopharmacological use to modern application. J. Ethnopharmacol. 2020, 268, 113586. [Google Scholar] [CrossRef]
  46. Asano, K.; Takahashi, T.; Miyashita, M.; Matsuzaka, A.; Muramatsu, S.; Kuboyama, M.; Kugo, H.; Imai, J. Effect of eleutheroccocus senticosus extract on human physical working capacity. Planta Med. 1986, 52, 175–177. [Google Scholar] [CrossRef]
  47. European Directorate for the Quality of Medicines & Health Care. Eleutherococci radix. In European Pharmacopoeia, Monograph 01/2008:1419; Corrected 7.0; European Directorate for the Quality of Medicines & Health Care: Strasburg, France, 2016. [Google Scholar]
  48. Dowling, E.A.; Redondo, D.R.; Branch, J.D.; Jones, S.; McNabb, G.; Williams, M.H. Effect of Eleutherococcus senticosus on submaximal and maximal exercise performance. Med. Sci. Sports Exerc. 1996, 28, 482–489. [Google Scholar] [CrossRef] [PubMed]
  49. Kuo, J.; Chen, K.W.; Cheng, I.S.; Tsai, P.H.; Lu, Y.J.; Lee, N.Y. The effect of eight weeks of supplementation with Eleutherococcus senticosus on endurance capacity and metabolism in human. Chin. J. Physiol. 2010, 53, 105–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Cicero, A.; DeRosa, G.; Brillante, R.; Bernardi, R.; Nascetti, S.; Gaddi, A. effects of siberian ginseng (eleutherococcus senticosus maxim.) on elderly quality of life: A randomized clinical trial. Arch. Gerontol. Geriatr. 2004, 38, 69–73. [Google Scholar] [CrossRef]
  51. Szołomicki, S.; Samochowiec, L.; Wójcicki, J.; Droździk, M. The influence of active components of eleutherococcus senticosus on cellular defence and physical fitness in man. Phytother. Res. 2000, 14, 30–35. [Google Scholar] [CrossRef]
  52. Schaffler, K.; Wolf, O.; Burkart, M. No Benefit Adding Eleutherococcus senticosus to Stress Management Training in Stress-Related Fatigue/Weakness, Impaired Work or Concentration, A Randomized Controlled Study. Pharmacopsychiatry 2013, 46, 181–190. [Google Scholar] [CrossRef] [PubMed]
  53. Eschbach, L.C.; Webster, M.J.; Boyd, J.C.; McArthur, P.D.; Evetovich, T.K. The Effect of Siberian Ginseng (Eleutherococcus Senticosus) on Substrate Utilization and Performance during Prolonged Cycling. Int. J. Sport Nutr. Exerc. Metab. 2000, 10, 444–451. [Google Scholar] [CrossRef]
  54. Krasutsky, A.G.; Cheremisinov, V.N. Research of the influence of adaptogens on increasing the efficacy of the training process in fitness clubs. In Proceedings of the Current Problems of Biochemistry and Bioenergy Sport of the XXI Centyry, Moscow, Russia, 10–12 April 2018; pp. 267–282. [Google Scholar]
  55. Jacquet, A.; Grolleau, A.; Jove, J.; Lassalle, R.; Moore, N. Burnout: Evaluation of the efficacy and tolerability of TARGET 1® for professional fatigue syndrome (burnout). J. Int. Med. Res. 2015, 43, 54–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Buděšínský, M.; Vokáč, K.; Harmatha, J.; Cvačka, J. Additional minor ecdysteroid components of Leuzea carthamoides. Steroids 2008, 73, 502–514. [Google Scholar] [CrossRef]
  57. Timofeev, N.P. Leuzea Carthamoides DC: Application prospects as pharmpreparations and biologically active components. In Functional Foods for Chronic Diseases; Martirosyan, D.M., Ed.; Richardson: Texas, TX, USA, 2006; pp. 105–120. [Google Scholar]
  58. Bathori, M.; Toth, N.; Hunyadi, A.; Marki, A.; Zador, E. Phytoecdysteroids and anabolic-androgenic steroids—Structure and effects on humans. Curr. Med. Chem. 2008, 15, 75–91. [Google Scholar] [CrossRef] [Green Version]
  59. Isenmann, E.; Ambrosio, G.; Joseph, J.F.; Mazzarino, M.; de la Torre, X.; Zimmer, P.; Kazlauskas, R.; Goebel, C.; Botrè, F.; Diel, P.; et al. Ecdysteroids as non-conventional anabolic agent: Performance enhancement by ecdysterone supplementation in humans. Arch. Toxicol. 2019, 93, 1807–1816. [Google Scholar] [CrossRef]
  60. The World Anti-Doping Agency—WADA. The 2020 Monitoring Program. Available online: https://www.wada-ama.org/sites/default/files/resources/files/wada_2020_english_monitoring_program_pdf (accessed on 6 May 2021).
  61. Vanyuk, A.I. Evaluation of the effectivnness of rehabilitation measures among female volleyball players 18-22 years old in the competitive period of the annual training cycle. Slobozhanskiy Sci. Sports Visnik. 2012, 5, 95–98. [Google Scholar]
  62. Timofeev, N.P.; Koksharov, A.V. Study of Leuzea from leaves: Results of 15 years of trials in athletics. New Unconv. Plants Prospect. Use 2016, 12, 502–505. [Google Scholar]
  63. Wilborn, C.D.; Taylor, L.W.; Campbell, B.I.; Kerksick, C.; Rasmussen, C.J.; Greenwood, M.; Kreider, R.B. Effects of Methoxyisoflavone, ecdysterone, and sulfo-polysaccharide supplementation on training adaptations in resistance-trained males. J. Int. Soc. Sports Nutr. 2006, 3, 19–27. [Google Scholar] [CrossRef] [Green Version]
  64. Ryan, E.D.; Gerstner, G.R.; Mota, J.A.; Trexler, E.T.; Giuliani, H.K.; Blue, M.N.M.; Hirsch, K.R.; Smith-Ryan, A.E. The acute effects of a multi-ingredient herbal supplement on performance fatigability: A double-blind, randomized, and placebo-controlled trial. J. Diet. Suppl. 2020, 1–10. [Google Scholar] [CrossRef]
  65. Selepcova, L.; Sommer, A.; Vargova, M. Effect of feeding on a diet containing varying amounts of rhaponticum car-thamoides hay meal on selected morphological parameters in rats. Eur. J. Entornol. 2013, 92, 391–397. [Google Scholar]
  66. Plotnikov, M.B.; Aliev, O.I.; Vasil’Ev, A.S.; Andreeva, V.Y.; Krasnov, E.A.; Kalinkina, G.I. Effect of Rhaponticum carthamoides extract on structural and metabolic parameters of erythrocytes in rats with cerebral ischemia. Bull. Exp. Biol. Med. 2008, 146, 45–48. [Google Scholar] [CrossRef]
  67. Wu, J.; Gao, L.; Shang, L.; Wang, G.; Wei, N.; Chu, T.; Chen, S.; Zhang, Y.; Huang, J.; Wang, J.; et al. Ecdysterones from Rhaponticum carthamoides (Willd.) Iljin reduce hippocampal excitotoxic cell loss and upregulate mTOR signaling in rats. Fitoter 2017, 119, 158–167. [Google Scholar] [CrossRef]
  68. Seidlova-Wuttke, D.; Ehrhardt, C.; Wuttke, W. Metabolic effects of 20-OH-Ecdysone in ovariectomized rats. J. Steroid Biochem. Mol. Biol. 2010, 119, 121–126. [Google Scholar] [CrossRef]
  69. Koudela, K.; Tenora, J.; Bajer, J.; Mathova, A.; Slama, K. Stimulation of growth and development in Japanase quails after oral administration of ecdysteroid-containing diet. Eur. J. Entomol. 1995, 92, 349. [Google Scholar]
  70. Sláma, K.; Koudela, K.; Tenora, J.; Maťhová, A. Insect hormones in vertebrates: Anabolic effects of 20-hydroxyecdysone in Japanese quail. Experientia 1996, 52, 702–706. [Google Scholar] [CrossRef]
  71. Xia, X.; Zhang, Q.; Liu, R.; Wang, Z.; Tang, N.; Liu, F.; Huang, G.; Jiang, X.; Gui, G.; Wang, L.; et al. Effects of 20-hydroxyecdysone on improving memory deficits in streptozotocin-induced type 1 diabetes mellitus in rat. Eur. J. Pharmacol. 2014, 740, 45–52. [Google Scholar] [CrossRef] [PubMed]
  72. Roumanille, R.; Vernus, B.; Brioche, T.; Descossy, V.; Van Ba, C.T.; Campredon, S.; Philippe, A.G.; Delobel, P.; Bertrand-Gaday, C.; Chopard, A.; et al. Acute and chronic effects of Rhaponticum carthamoides and Rhodiola rosea extracts supplementation coupled to resistance exercise on muscle protein synthesis and mechanical power in rats. J. Int. Soc. Sports Nutr. 2020, 17, 1–13. [Google Scholar] [CrossRef] [PubMed]
  73. Brown, R.P.; Gerbarg, P.L.; Ramazanov, Z. Rhodiola rosea: A phytomedicinal overview. Herbal. Gram. 2002, 56, 40–52. [Google Scholar]
  74. Pu, W.-L.; Zhang, M.-Y.; Bai, R.-Y.; Sun, L.-K.; Li, W.-H.; Yu, Y.-L.; Zhang, Y.; Song, L.; Wang, Z.-X.; Peng, Y.-F.; et al. Anti-inflammatory effects of Rhodiola rosea L.: A review. Biomed. Pharmacother. 2020, 121, 109552. [Google Scholar] [CrossRef]
  75. Khanum, F.; Bawa, A.S.; Singh, B. Rhodiola rosea: A versatile adaptogen. Compr. Rev. Food Sci. Food Saf. 2005, 4, 55–62. [Google Scholar] [CrossRef]
  76. Panossian, A.; Seo, E.-J.; Efferth, T. Novel molecular mechanisms for the adaptogenic effects of herbal extracts on isolated brain cells using systems biology. Phytomedicine 2018, 50, 257–284. [Google Scholar] [CrossRef]
  77. Ballmann, C.G.; Maze, S.B.; Wells, A.C.; Marshall, M.R.; Rogers, R.R. Effects of short-term Rhodiola Rosea (golden root extract) supplementation on anaerobic exercise performance. J. Sports Sci. 2019, 37, 998–1003. [Google Scholar] [CrossRef]
  78. 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. J. Sport Health Sci. 2018, 7, 473–480. [Google Scholar] [CrossRef] [Green Version]
  79. Abidov, M.; Grachev, S.; Seifulla, R.D.; Ziegenfuss, T.N. Extract of Rhodiola rosea radix reduces the level of c-reactive protein and creatinine kinase in the blood. Bull. Exp. Biol. Med. 2004, 138, 63–64. [Google Scholar] [CrossRef]
  80. Edwards, D.; Heufelder, A.; Zimmermann, A. Therapeutic effects and safety of Rhodiola rosea extract WS® 1375 in subjects with life-stress symptoms—Results of an open-label study. Phytother. Res. 2012, 26, 1220–1225. [Google Scholar] [CrossRef]
  81. Shevtsov, V.; Zholus, B.; Shervarly, V.; Vol’Skij, V.; Korovin, Y.; Khristich, M.; Roslyakova, N.; 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, 10, 95–105. [Google Scholar] [CrossRef] [Green Version]
  82. Darbinyan, V.; Aslanyan, G.; Amroyan, E.; Gabrielyan, E.; Malmström, C.; Panossian, A. Clinical trial of Rhodiola rosea L. extract SHR-5 in the treatment of mild to moderate depression. Nord. J. Psychiatry 2007, 61, 343–348. [Google Scholar] [CrossRef]
  83. Shanely, R.A.; Nieman, D.C.; Zwetsloot, K.A.; Knab, A.M.; Imagita, H.; Luo, B.; Davis, B.; Zubeldia, J.M. Evaluation of Rhodiola rosea supplementation on skeletal muscle damage and inflammation in runners following a competitive marathon. Brain Behav. Immun. 2013, 39, 204–210. [Google Scholar] [CrossRef]
  84. Stejnborn, A.S.; Pilaczyńska-Szcześniak, S.; Basta, P.; Deskur-Śmielecka, E. The influence of supplementation with Rhodiola rosea L. Extract on selected redox parameters in professional rowers. Int. J. Sport Nutr. Exerc. Metab. 2009, 19, 186–199. [Google Scholar] [CrossRef] [Green Version]
  85. Parisi, A.; Tranchita, E.; Duranti, G.; Ciminelli, E.; Quaranta, F.; Ceci, R.; Sabatini, S. Effects of chronic Rhodiola Rosea sup-plementation on sport performance and antioxidant capacity in trained male: Preliminary results. J. Sports Med. Phys. Fit. 2010, 50, 57. [Google Scholar]
  86. Bystritsky, A.; Kerwin, L.; Feusner, J.D. A pilot study of Rhodiola rosea (Rhodax®) for generalized anxiety disorder (GAD). J. Altern. Complement. Med. 2008, 14, 175–180. [Google Scholar] [CrossRef] [Green Version]
  87. Hancke, J.; Burgos, R.; Ahumada, F. Schisandra chinensis (Turcz.) Baill. Fitoterapia 1999, 70, 451–471. [Google Scholar] [CrossRef]
  88. Lu, Y.; Chen, D.-F. Analysis of Schisandra chinensis and Schisandra sphenanthera. J. Chromatogr. A 2009, 1216, 1980–1990. [Google Scholar] [CrossRef]
  89. Panossian, A.; Wikman, G. Pharmacology of Schisandra chinensis bail.: An overview of Russian research and uses in medicine. J. Ethnopharmacol. 2008, 118, 183–212. [Google Scholar] [CrossRef]
  90. Slanina, J.; Táborská, E.; Lojková, L. Lignans in the seeds and fruits of Schisandra chinensis cultured in Europe. Planta Med. 1997, 63, 277–280. [Google Scholar] [CrossRef]
  91. European Directorate for the Quality of Medicines & Health Care. Schisandrae chinensis fructus. In European Pharmacopoeia, Monograph 07/2016:2428; Corrected 9.1, Corrected 7.0; European Directorate for the Quality of Medicines & Health Care: Strasburg, France, 2016. [Google Scholar]
  92. Szopa, A.; Barnaś, M.; Ekiert, H. Phytochemical studies and biological activity of three Chinese Schisandra species (Schisandra sphenanthera, Schisandra henryi and Schisandra rubriflora): Current findings and future applications. Phytochem. Rev. 2019, 18, 109–128. [Google Scholar] [CrossRef] [Green Version]
  93. Kochetkov, N.; Khorlin, A.; Chizhov, O.; Sheichenko, V. Schizandrin—Lignan of unusual structure. Tetrahedron Lett. 1961, 2, 730–734. [Google Scholar] [CrossRef]
  94. Chen, D.-F.; Zhang, S.-X.; Kozuka, M.; Sun, Q.-Z.; Feng, J.; Wang, Q.; Mukainaka, T.; Nobukuni, Y.; Tokuda, H.; Nishino, H.; et al. Interiotherins C and D, two new lignans from Kadsurainteriorand antitumor-promoting effects of related neolignans on Epstein−Barr Virus Activation. J. Nat. Prod. 2002, 65, 1242–1245. [Google Scholar] [CrossRef]
  95. Yoo, H.H.; Lee, M.; Lee, M.W.; Lim, S.Y.; Shin, J.; Kim, D.-H. Effects of Schisandra lignans on P-Glycoprotein-mediated drug efflux in human intestinal Caco-2 Cells. Planta Med. 2007, 73, 444–450. [Google Scholar] [CrossRef]
  96. Fong, W.-F.; Wan, C.-K.; Zhu, G.-Y.; Chattopadhyay, A.; Dey, S.; Zhao, Z.; Shen, X.-L. Schisandrol A from Schisandra chinensis reverses P-Glycoprotein-mediated multidrug resistance by affecting Pgp-substrate complexes. Planta Med. 2007, 73, 212–220. [Google Scholar] [CrossRef] [Green Version]
  97. Chen, M.; Kilgore, N.; Lee, K.-H.; Chen, D.-F. Rubrisandrins A and B, lignans and related anti-HIV compounds from Schisandra rubriflora. J. Nat. Prod. 2006, 69, 1697–1701. [Google Scholar] [CrossRef]
  98. Chen, D.-F.; Zhang, S.-X.; Xie, L.; Xie, J.-X.; Chen, K.; Kashiwada, Y.; Zhou, B.-N.; Wang, P.; Cosentino, L.; Lee, K.-H. Anti-aids agents—XXVI. Structure-activity correlations of Gomisin-G-related anti-HIV lignans from Kadsura interior and of related synthetic analogues. Bioorganic Med. Chem. 1997, 5, 1715–1723. [Google Scholar] [CrossRef]
  99. Liu, C.; Zhang, S.; Zhang, J.; Liang, Q.; Li, D. Chemical composition and antioxidant activity of essential oil from berries of Schisandra chinensis(Turcz.) Baill. Nat. Prod. Res. 2012, 26, 2199–2203. [Google Scholar] [CrossRef]
  100. Chen, X.; Zhang, Y.; Zu, Y.; Yang, L. Chemical composition and antioxidant activity of the essential oil of Schisandra chinensisfruits. Nat. Prod. Res. 2012, 26, 842–849. [Google Scholar] [CrossRef]
  101. Xu, M.; Yan, T.; Gong, G.; Wu, B.; He, B.; Du, Y.; Xiao, F.; Jia, Y. Purification, structural characterization, and cognitive improvement activity of a polysaccharides from Schisandra chinensis. Int. J. Biol. Macromol. 2020, 163, 497–507. [Google Scholar] [CrossRef]
  102. Liu, Y.; Guo, J.-T.; Wang, Z.-B.; Li, Z.-Y.; Zheng, G.-X.; Xia, Y.-G.; Yang, B.-Y.; Kuang, H.-X. Aromatic monoterpenoid glycosides from rattan stems of Schisandra chinensis and their neuroprotective activities. Fitoterapia 2019, 134, 108–112. [Google Scholar] [CrossRef] [PubMed]
  103. Mocan, A.; Crișan, G.; Vlase, L.; Crișan, O.; Vodnar, D.C.; Raita, O.; Gheldiu, A.-M.; Toiu, A.; Oprean, R.; Tilea, I. Comparative studies on polyphenolic composition, antioxidant and antimicrobial activities of schisandra chinensis leaves and fruits. Molecules 2014, 19, 15162–15179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Yang, B.-Y.; Guo, J.-T.; Li, Z.-Y.; Wang, C.-F.; Wang, Z.-B.; Wang, Q.-H.; Kuang, H.-X. New Thymoquinol Glycosides and Neuroprotective Dibenzocyclooctane Lignans from the Rattan Stems ofSchisandra chinensis. Chem. Biodivers. 2016, 13, 1118–1125. [Google Scholar] [CrossRef] [PubMed]
  105. Park, J.; Han, S.; Park, H. Effect of Schisandra chinensis extract supplementation on quadriceps muscle strength and fatigue in adult women: A randomized, double-blind, placebo-controlled trial. Int. J. Environ. Res. Public Health 2020, 17, 2475. [Google Scholar] [CrossRef] [Green Version]
  106. Park, J.Y.; Kim, K.H. A randomized, double-blind, placebo-controlled trial of Schisandra chinensis for menopausal symptoms. Climacteric 2016, 19, 574–580. [Google Scholar] [CrossRef]
  107. Song, M.-Y.; Wang, J.; Eom, T.; Kim, H. Schisandra chinensis fruit modulates the gut microbiota composition in association with metabolic markers in obese women: A randomized, double-blind placebo-controlled study. Nutr. Res. 2015, 35, 655–663. [Google Scholar] [CrossRef]
  108. Cao, S.; Shang, H.; Wu, W.; Du, J.; Putheti, R. Evaluation of anti-athletic fatigue activity of Schizandra chinensis aqueous extracts in mice. Afr. J. Pharm. Pharmacol. 2009, 3, 593–597. [Google Scholar]
  109. Li, J.; Wang, J.; Shao, J.-Q.; Du, H.; Wang, Y.-T.; Peng, L. Effect of Schisandra chinensis on interleukins, glucose metabolism, and pituitary-adrenal and gonadal axis in rats under strenuous swimming exercise. Chin. J. Integr. Med. 2014, 21, 43–48. [Google Scholar] [CrossRef]
  110. Chen, X.; Cao, J.; Sun, Y.; Dai, Y.; Zhu, J.; Zhang, X.; Zhao, X.; Wang, L.; Zhao, T.; Li, Y.; et al. Ethanol extract of Schisandrae chinensis fructus ameliorates the extent of experimentally induced atherosclerosis in rats by increasing antioxidant capacity and improving endothelial dysfunction. Pharm. Biol. 2018, 56, 612–619. [Google Scholar] [CrossRef] [Green Version]
  111. Sun, J.-H.; Liu, X.; Cong, L.-X.; Li, H.; Zhang, C.-Y.; Chen, J.-G.; Wang, C.-M. Metabolomics study of the therapeutic mechanism of Schisandra chinensis lignans in diet-induced hyperlipidemia mice. Lipids Health Dis. 2017, 16, 227. [Google Scholar] [CrossRef] [Green Version]
  112. Ip, S.-P.; Poon, M.; Wu, S.; Che, C.; Ng, K.; Kong, Y.; Ko, K. Effect of Schisandrin B on hepatic glutathione antioxidant system in mice: Protection against carbon tetrachloride toxicity. Planta Med. 1995, 61, 398–401. [Google Scholar] [CrossRef]
  113. Giridharan, V.V.; Thandavarayan, R.A.; Sato, S.; Ko, K.M.; Konishi, T. Prevention of scopolamine-induced memory deficits by schisandrin B, an antioxidant lignan from Schisandra chinensis in mice. Free. Radic. Res. 2011, 45, 950–958. [Google Scholar] [CrossRef]
  114. Kim, K.-Y.; Ku, S.-K.; Lee, K.-W.; Song, C.-H.; An, W.G. Muscle-protective effects of Schisandrae fructus extracts in old mice after chronic forced exercise. J. Ethnopharmacol. 2018, 212, 175–187. [Google Scholar] [CrossRef]
  115. Chen, Y.; Tang, J.; Wang, X.; Sun, F.; Liang, S. An immunostimulatory polysaccharide (SCP-IIa) from the fruit of Schisandra chinensis (Turcz.) Baill. Int. J. Biol. Macromol. 2011, 50, 844–848. [Google Scholar] [CrossRef]
  116. Zhao, T.; Mao, G.-H.; Zhang, M.; Li, F.; Zou, Y.; Zhou, Y.; Zheng, W.; Zheng, D.-H.; Yang, L.-Q.; Wu, X.-Y. Anti-diabetic effects of polysaccharides from ethanol-insoluble residue of Schisandra chinensis (Turcz.) baill on alloxan-induced diabetic mice. Chem. Res. Chin. Univ. 2012, 29, 99–102. [Google Scholar] [CrossRef]
Figure 1. PRISMA 2020 flow diagram for new systematic reviews which included searches of databases and registers only.
Figure 1. PRISMA 2020 flow diagram for new systematic reviews which included searches of databases and registers only.
Nutrients 13 02861 g001
Figure 2. Mechanism of action of Panax ginseng.
Figure 2. Mechanism of action of Panax ginseng.
Nutrients 13 02861 g002
Figure 3. 20-hydroxyecdysone structure.
Figure 3. 20-hydroxyecdysone structure.
Nutrients 13 02861 g003
Figure 4. Structure of γ-Schisandrin.
Figure 4. Structure of γ-Schisandrin.
Nutrients 13 02861 g004
Table 1. Panax ginseng studies.
Table 1. Panax ginseng studies.
Study ObjectivesStudy DesignMain ResultsReferences
Evaluation of the effects on subjective mood and memory of a single and sub-chronic Panax ginseng dose.Thirty adults, aged 22.87 ± 4.01 years,
participated in the study. They received a placebo, 200 or 400 mg Panax ginseng
extract per day for 3 treatments—8 days with 6 days washout.
Period of the study—32 days.
Improved calmness, mood,
and mental health.
[21]
Examine the effects of Panax ginseng on the lipid profile.Eight men, aged 21.1 ± 2.1 years,
participated in the study. All participants received 2 g of ginseng extract 3 times per day.
Period of the study—8 weeks.
Decreased levels of serum total
cholesterol (TC), triglycerides (TG), low-density lipoprotein (LDL), plasma malondialdehyde (MDA), and an increase in high-density lipoprotein (HDL).
[32]
Examine the effects on
endurance performance of acute supplementation
of Panax ginseng.
Twelve men, aged 20–24 years,
participated in the study. All participants received 200 mg ginseng extract or a
placebo one hour before the exercise.
Increased endurance time, blood
glucose and insulin levels, catalase,
superoxide-dismutase, and total thiol.
[33]
Evaluation of benefits on
fatigue in multiple sclerosis with Panax ginseng treatment.
Fifty-two women, aged 18–50 years,
participated in the study. There were 26 participants who received 500 mg daily of Korean ginseng
tablets and 26 were in the placebo group.Period of the study—3 months.
Reduced fatigue.
Improved quality of life.
[34]
Examine the effects of Panax ginseng on work performance. Nineteen women, 21–35 years of age,
participated in the study. There were 10 participants who received 200 mg of Panax ginseng extract daily, and nine of them were in the placebo group.
Period of the study—2 months.
No change in maximal work
performance, oxygen consumption (VO2), respiratory exchange ratio, minute
ventilation, heart rate, and blood
lactic acid levels.
[35]
Evaluation of the efficacy of a combination of Panax ginseng and vitamins on physical and mental stress.One-hundred and fourteen women and men, aged 30–60 years, participated in the study. There were 59 participants who received 200 mg daily of ginseng dried extract and vitamins; 55 were in the placebo group.
Period of the study—8 weeks.
Increased quality of life,
without a difference in blood pressure and heart rate.
[36]
Evaluation of the efficacy of Panax ginseng extract on physical and mental
performance.
Fifteen men, aged 19.07 ± 0.62 years,
participated in the study. Seven of them received 200 mg daily of ginseng extract; eight of the participants were in the placebo group. Period of study—6 weeks.
Increased lactate levels. No change in VO2 and heart rate. Decreased cortisone levels and no change in testosterone levels.[37]
The influence of Panax ginseng on cortisol, growth hormone, and lactate. Ten women, aged 23.4 ± 0.69 years,
participated in the study. In the first
4 weeks, five received 100 mg per day and five were in the placebo group. In the second 4 weeks, they were switched.Period of study—8 weeks.
Increased cortisol. No change in
growth hormone and lactate.
[38]
Evaluation of the effects of Panax ginseng on sleep.Fifteen men, aged 19–25 years, participated in the study. There were eight participants who received
4.5 g of ginseng extract daily;
seven participants were in the placebo group. Period of the study—2 weeks.
Increased deep sleep.
Decreased shallow sleep.
[39]
Evaluation of anti-fatigue
effects of Panax ginseng.
Eighty-eight men and women 20–60 years of age participated in the study. There were
30 participants who received 1 g of ginseng
extract daily; 29 participants had an
intake of 2 g of ginseng extract daily. There were29 participants who were in the placebo group.
Period of study—2 months.
Reduced the severity of fatigue.
Increased glutathione reductase
and total glutathione.
[40]
Assessment of an ergogenic effect on Malaysian population of Panax ginseng in humid and hot conditions. Nine men, 25.4 ± 6.9 years of age, participated in the study. In the first trial, they had an intake of 200 mg of Panax ginseng one hour before an exercise test and in the second trial, they had an intake placebo.Decreased lactate, plasma glucose, plasma insulin.
Increased free fatty acids. No change
in heart rate, VO2, skin, and
body temperature.
[41]
Table 2. Eleutherococcus senticosus studies.
Table 2. Eleutherococcus senticosus studies.
Study ObjectivesStudy DesignMain ResultsReferences
Examine the effects of Eleutherococcus senticosus extract on
physical working capacity.
Six men, aged 21–22 years, participated in the study. They were in the control group, placebo group, and the group that received 2 mL
ethanol extract (125 mg dried extract)
Eleutherococcus senticosus twice daily.
Period of study—8 days.
Increased maximal oxygen uptake, oxygen pulse, total work, and
exhaustion time.
[46]
Examine the effects of
Eleutherococcus senticosus during maximal and submaximal
aerobic exercise.
Twenty men and women, aged 37 ± 8 years, participated in the study. There were 10 participants who received 3.4 mL Eleutherococcus senticosus
extract; 10 participants were in the placebo group. Period of study—8 weeks.
No changes in heart rate, VO2, lactate.[48]
Assessment of the effects of Eleutherococcus senticosus on
metabolism, endurance capacity, and cardiovascular functions.
Nine men, aged 19 ± 2.1 years, participated in the study. The participants received 800 mg daily of Eleutherococcus senticosus or placebo.
Period of study—8 weeks.
Decreased blood glucose levels.
Increased VO2, endurance time,
heart rate, and free fatty acids.
[49]
Assessment of the impact of Eleutherococcus senticosus
on quality of life.
Twenty volunteers, aged over 65 years,
participated in the study. There were 10 participants who
received 300 mg/day Siberian ginseng extract and 10 participants who were in placebo group.
Period of study—2 months.
Improved mental health and social functioning, but prolonged use
decreased these improvements. Blood pressure was not affected.
[50]
Assessment of the influence of Eleutherococcus senticosus
on physical fitness and
cellular defence.
Forty-six men and women, aged 23–73 years, participated in the study. There were 31 participants who
received 75 drops of Eleutherococcus senticosus extract daily and 15 participants who received
120 drops of Echinacea extract daily
for one month.
Decreased total cholesterol, LDL,
triglycerides, free fatty acids, and
glucose. Increased maximal oxygen consumption (VO2max).
[51]
Examine the effects of a dietary supplement containing
Eleutherococcus senticosus extract on burnout symptoms.
Eighty-seven volunteers, aged 27–63 years, participated in the study. There were 44 participants who
had an intake of 100 mg dry extract
from Eleutherococcus senticosus;
43 were in the placebo group.
Period of study—12 weeks.
Decreased fatigue score and
Beck depression.
[52]
Evaluate the effects of
Eleutherococcus senticosus
extract on stress.
One-hundred and thirty women and men, aged 30–50 years, participated in the study. There were 49 participants who received 120 mg/day of dry Eleutherococcus senticosus extract,
40 participants who worked out, and 41 who worked out and took 120 mg/day dry extract.
Period of study—2 months.
Improvement in fatigue, exhaustion, sleep, and restlessness.[52]
Examine physiological reactions from the intake of Eleutherococcus senticosus on cyclists.Nine men, aged 28 ± 2 years, participated in the study. The participants received 1200 mg per day of Eleutherococcus senticosus extract for
7 days and the placebo for 7 days.
No difference in respiratory
exchange ratio, oxygen consumption, heart rate, perceived exertion, plasma lactate, plasma glucose.
[53]
Evaluate the effects of
Eleutherococcus senticosus during the training process in fitness.
In the second series of study,
17 men and women participated; 10 of them received Eleutherococcus senticosus extract, and
7 participants were in the controlled group.
Decreased body weight. Increased physical endurance and
performance.
[54]
Table 3. Rhaponticum carthamoides studies.
Table 3. Rhaponticum carthamoides studies.
Study ObjectivesStudy DesignMain ResultsReferences
Evaluation of the effects of an increased dose of Rhaponticum carthamoides during the training
process.
Twenty women, aged 25–40 years,
participated in the study. There were 12 of them whoreceived 5–15 mg/kg/day ecdysterone;
8 were in controlled group.
Decreased body weight. Increased physical endurance and performance. Improvement of cardiac and cognitive function.[19]
Examine the effect of ecdysterone-containing products on sport physical exercises.Forty-six men, aged 25.6 ± 3.7 years,
participated in the study. There were 12 participants whohad an intake of 200 mg ecdysterone;
10 participants received 800 mg ecdysterone, 12 participants received the placebo, and
12 of the participants were in the control group—they had an intake of 200 mg
ecdysterone without training.
Period of study—10 weeks.
Ecdysterone increased body weight, muscle mass. Increased power and strength of performance. Without negative effects on creatinine, glutamate–oxaloacetate transaminase, gamma-glutamyl transferase, and glutamate–pyruvate transaminase. Did not affect steroid profile.[59]
Evaluation of the
effectiveness of
ecdysterone in athletes.
Twenty-six women aged 18–22 years
participated in the study. There were 12 participants who
received ecdysterone from 37.5 to 50 mg;
14 participants were in the controlled group.
Period of study—9 moths.
Increased VO2 lactate,
performance activity.
[61]
Evaluation of the
effectiveness of
ecdysterone from
Rhaponticum carthamoides leaves in athletes.
No information—number of participants. The age of the participants ranged between 27–58 years. Participants received 2–3 g Rhaponticum carthamoides tea, infusion, tincture,
fermented tea without bitterness.
Period of study—15 years.
Increased resistance to disease, physical, and mental endurance. [62]
Assessment of effects
of methoxyisoflavone, 20-hydroxyecdysone, and sulfopolysaccharides intake on training adaptation and markers of muscle
anabolism and catabolism.
Forty-five men, aged 20.5 ± 3 years,
participated in the study. The participants were divided randomly into four groups:
the placebo group, the group that received
methoxyisoflavone—800 mg daily, the
group that received 20-hydroxyecdysone—200 mg/day, and the group that received
sulfo-polysaccharides—1000 mg daily.
Period of study—8 weeks.
No change in training adaptation and in anabolic and catabolic effect
in training.
[63]
Evaluation of the effects of the combination of Rhaponticum carthamoides and Rhodiola rosea on
performance fatigability and reactions before and after training.
Twenty-seven men, aged 22.3 ± 4.1 years, participated in the study. The participants received a 350 mg tablet which contains 70:30 Rhaponticum carthamoides extract and
Rhodiola rosea extract, or a tablet containing
175 mg maltodextrin, and 175 mg
Rhaponticum carthamoides and Rhodiola rosea extract in ratio 70:30 or placebo.
No change in muscle strength
and total work.
[64]
Table 4. Rhaponticum carthamoides animal studies.
Table 4. Rhaponticum carthamoides animal studies.
Study ObjectivesStudy DesignMain ResultsReferences
Evaluating Rhaponticum carthamoides effects—growth,
increased body weight, and
behaviour on rats.
For study, 60 rats were used, divided into
10 groups with six rats in group. Females were
fed with 5% Raponticum hay meal, males were fed with 10–20% Raponticum hay meal. Period of study—21 days.
Increased growth and
body weight.
[65]
Evaluating Rhaponticum carthamoides effects on lipid profile on rats with cerebral ischemia.For the study, 18 rats were used and
received 150 mg/kg Rhaponticum carthamoides extract or placebo. Period of study—5 days.
Decreased lysophospholipids in erythrocyte membrane. Increased total lipids and phospholipids.[66]
Examine ecdysterone
neuroprotective mechanism
of action.
For the study, 35 rats were used, aged
6–8 weeks. They were divided into seven groups: non-operated, two controlled groups, and three experimental groups. In the experimental groups, the rats were administrated ecdysterone in
5 mg/kg, 10 mg/kg, 20 mg/kg.
Period of study—7 days.
Removed glutamatergic excitotoxicity. Neuroprotective effect.[67]
Examine the anti-obesity effect of 20-hydroxyecdysone.For the study, 60 rats were used, aged
3 months. They were divided into 12 rats in each group and three of the groups were treated with 18, 56, and 116 mg per day
20-hydroxyedysone.
Period of study—3 months.
Decreased LDL. Increased muscle mass. No changes in
thyroid-stimulating hormone (TSH), tetraiodothyronine (T4),
and triiodothyronine (T3).
[68]
Examine anabolic effects of
Leuzea carthamoides in quails.
For the study, 1000 quails were used. They were divided into six groups—the control group, four groups that had received a standard diet
combined with Leuzea seed 0.2—5%, and the other two groups. Period of study—50 days.
Increased body mass and growth.[69]
Examine anabolic effects of
20-hydroxyecdysone in quails.
For the study, 200 quails were used; 160 were in the control group. There were 10 that received 20 mg/kg,
10 that received 100 mg/kg, and 10 that received
500 mg/kg 20-hydroxyecdysone isolated from Leuzea carthamoides. Period of study—4 weeks.
Increased growth. Anabolic effect.[70]
Examine the improving memory effect of
20-hydroxyecdysone.
For the study, 80 rats were used, divided into two groups: 10 were in the control group, 70 in the experimental group.
They received 1, 10, and 100 mg/kg 20-hydroxyecdysone per day.
Period of study—12 weeks.
Induced superoxide dismutase (SOD), catalase, glutathione peroxidase (GSH-Px), and glutathione
reductase (GR). Decreased glucose levels, nuclear factor-kB (NF-kB).
[71]
Examine the effects of Rhapoticum carthamoides, Rhodiola rosea, and their combination on resistance exercise and mechanical power.For the study, 56 rats were used. Rats were divided into seven groups, eight rats in each group: control group, a group that received only Rhodiola rosea extract, a group that received only Rhaponticum carthamoides extract, and four groups that received a combination of Rhaponticum carthamoides and Rhodiola rosea extracts in different quantitative ratios.Rhaponticum carthamoides extract
increased muscle protein synthesis. The combination of Rhaponticum carthamoides and Rhodiola rosea
increased muscle protein synthesis and mean power performance.
[72]
Table 5. Rhodiola rosea studies.
Table 5. Rhodiola rosea studies.
Study ObjectivesStudy DesignMain ResultsReferences
Studying the effects of short-term supplementation with
Rhodiola rosea.
Eleven women, aged 19.4 ± 0.8 years,
participated in the study. They had an intake of 1.5 g/day Rhodiola rosea extract or placebo for 3 days. A 500 mg additional dose of Rhodiola rosea extract was taken before each trial.
Increased anaerobic capacity,
anaerobic power, and total work. No change in fatigue index.
[77]
Examine hormonal and oxidative stress of Rhodiola rosea supplementation and the effects on mental and physical performance. Twenty-six men participated in the study.
Thirteen of them had an intake of 600 mg/day extract of Rhodiola rosea and 13 were in placebo group.
Period of study—4 weeks.
Improved reaction and response time. Increased antioxidant capacity. Without changes in hormone profile and endurance exercise capacity.[78]
Examine the levels of inflammatory C-reactive protein and creatinine kinase in blood after intake of Rhodiola rosea.Thirty-six volunteers aged 21–24 years
participated in the study. Twelve of them had an intake of 340 mg Rhodiola rosea extract twice
a day, 12 participants were in the placebo group, and 12 participants were in the control group.
Period of study—36 days.
Increased levels of C-reactive
protein and creatinine kinase.
[79]
Examine the effects and safety of Rhodiola rosea extract for
4 weeks of treatment.
There were 101 women and men, aged 30–60 years, who
participated in study. All participants had an intake of Rhodiola rosea extract 400 mg/day.
Period of the study—1 month.
Improved mood, stress symptoms, and quality of life.[80]
Examine the effects of a single dose of standardized Rhodiola rosea extract.There were 121 men aged 19–21 years participated in the study; 41 participants received 370 mg dry
extract Rhodiola rosea, 20 participants received 555 mg dry extract of Rhodiola rosea before test, 40 of participants were in the placebo group,
20 participants were in the controlled group.
Improvement in the
anti-fatigue index.
[81]
Examine the effects of
standardized Rhodiola rosea
extract in patients suffering from depression.
Eighty-nine women and men, aged 18–70 years, participated in the study. Thirty-one participants received 340 mg/day extract of Rhodiola rosea,
29 participants received 680 mg/day extract of Rhodiola rosea, and 29 participants
were in the placebo group.
Period of study—42 days.
Improved in overall depression, insomnia, somatization, and
emotional instability. No
improvements in self-belief.
[82]
Evaluating the changes of
Rhodiola rosea supplementation on muscle damage and
inflammation.
There were 48 men and women, aged 25–60 years, who participated in the study. Twenty-four participants
received a 300 mg capsule per day containing Rhodiola rosea extract, and 24 participants
were in the placebo group.
Period of study—38 days.
Increased myoglobin, creatine phosphokinase, aspartate
aminotransferase, alanine
aminotransferase, and interleukin (IL-6, IL-8, IL-10) without a
difference in both groups.
[83]
Examine the effects of Rhodiola rosea supplementation on
selected redox parameters
in athletes.
Twenty-two men aged 20.4 ± 1.2 participated in the study. Eleven of them had an intake of
200 mg/day Rhodiola rosea extract,
and 11 were in the placebo group.Period of study—4 weeks. Decreased levels of superoxide dismutase. Increased total
antioxidant capacity.
Decreased levels of superoxide
dismutase. Increased total
antioxidant capacity.
[84]
Examine the effects of chronic intake of Rhodiola rosea on
physical performance and
antioxidant capacity during
exercise in athletes.
Fourteen men, aged 25 ± 5 years, participated in the study. All of the participants received a
placebo; after that, all of them received
170 mg R. rosea extract for 1 month.
Decreased free fatty acids levels, blood lactate, and creatinine kinase levels. No change in VO2max.[85]
The efficacy of Rhodiola rosea in generalized anxiety disorder.Ten men and women, aged 34–55 years,
participated in the study. All participants had intake 340 mg Rhodiola rosea extract per day
for 10 weeks.
Decreased scores in Hamilton
Anxiety Rating Scale and Hamilton Depression Rating Scale.
[86]
Table 6. Schisandra chinensis studies.
Table 6. Schisandra chinensis studies.
Study ObjectivesStudy DesignMain ResultsReferences
Examine the effects of
Schisandra chinensis extract on muscle strength and lactate.
Forty-five volunteers aged 61.9 ± 8.4 years
participated in the study. Twenty-four participants received 1000 mg/day extract of Schisandra chinensis and
21 participants were in the placebo group.
Period of study—3 months.
Decreased lactate levels.
Increased quadriceps and
muscle strength.
[105]
Examine the effects of
Schisandra chinensis extract for menopausal symptoms.
Thirty-six women aged 40–70 years participated in the study. Eighteen participants received Schisandra chinensis extract 748 mg twice a day for 6 weeks; 18 were in the placebo group.
Period of study—12 weeks.
Decreased hot flushes, sweating, and heart rate.[106]
Examine the effects of
Schisandra chinensis fruit
on gut microbiota.
Twenty-eight women participated in the study. Thirteen participants received 6.7 g/day dried
Schisandra chinensis fruits, and 15 participants were in placebo group. Period of study—12 weeks.
Decreased blood glucose,
triglycerides, alanine
aminotransferase, aspartate
aminotransferase, and fat mass.
Increased Bacteroides
and Bacteroidetes.
[107]
Table 7. Schisandra chinensis animal studies.
Table 7. Schisandra chinensis animal studies.
Study ObjectivesStudy DesignMain ResultsReferences
Examine the anti-athletics
fatigue effects of
Schisandra chinensis.
For the study, eight mice were used, divided into five groups—the control group, low-dose group—treated with 15 mg/kg Schisandra chinensis aqueous extract; the medium-dose group—treated with
30 mg/kg Schisandra chinensis aqueous extract;
intermediate-high group—treated with 50 mg/kg Schisandra chinensis aqueous extract; and
high-group—treated with 80 mg/kg Schisandra chinensis aqueous extract for 28 days.
Prevented an increase of lactate levels and blood urea nitrogen. Increased blood
hemoglobin levels.
[108]
Examine Schisandra chinensis
effects on pituitary-adrenal and gonadal axis, interleukins, and blood glucose levels.
For the study, 45 rats were used, aged 6 weeks old.
There were 15 mice in the control group, 15 in the stress group, and 15 in the group that received 5 g/kg/day
Schisandra chinensis and exercise.
Period of study—11 days.
Decreased blood glucose levels, cortisol, and interleukins
(IL-1 and IL-2).
[109]
Examine Schisandra chinensis
effects on atherosclerosis in rats.
For the study, there were 60 rats used, aged 4 weeks, and 20 mice, aged 6 weeks. They were divided into
five groups—normal, model, simvastatin (received 4 mg/kg/day), and low-dose group—which received an extract of Schisandra chinensis, 0.35 mg/kg day; medium-dose group—received extract of Schisandra chinensis 0.7 mg/kg/day; and high-dose group—received 1.4 mg/kg/day Schisandra chinensis extract for 3 weeks. Period of study—12 weeks.
Decreased TG and LDL levels. Increased HDL. [110]
Examine the effect of Schisandra chinensis on mice with hyperlipidemia. For the study, 48 mice were used. There were 24 mice in the control group and 24 received 100 mg/kg/day Schisandra chinensis lignans for 4 weeks.Decreased TC, TG, LDL.
Increased HDL, inhibited the mRNA expression of liver
X receptor alfa and the mRNA expression level of hepatic
lipogenesis.
[111]
Examine Schisandrine B effects on hepatic glutathione
antioxidant system.
For the study, mice were used, with no information for number of mice. They were divided into five groups. Control group and the rest had received Schisandrine B 1 to 4 mmol/kg for 3 days; after that, they were threatened with CCl4 0.1 mL/kg.Decreased glucose-6-phosphate dehydrogenase, gama-glutamilcysteine synthetase, and Se-glutathione peroxidase. Increased hepatic gluthatione S-transferase and glutathione reductase. [112]
Examine the preventive effect of Schisandrine B on scopolamine-induced dementia in mice.For the study, 30 mice were used, aged 12–14 weeks. They were divided into groups—the control group, three groups that had received
Schisandrine B, respectively, 10, 25, or 50 mg/kg/daily before treatment with scopolamine, and a group that had received 10 mg/kg/day tacrine. Period of study—7 days.
Prevented scopolamine-induced oxidative stress. Prevented the decrease of acetylcholine levels.[113]
Evaluated muscle-protective
effects of Schisandra chinensis
extract in mice, after exercise.
For the study, 48 mice were used, aged 10 months. They were divided into six groups with eight mice in each group. The first group received distilled water.
The second group received distilled water with exercise control. The third group
received 50 mg/kg oxymetholone. The rest of the three groups received, respectively, 125, 250, or 500 mg/kg per day of Schisandra chinensis extract and exercises. Period of study—28 days.
Decreased creatine, creatine
kinase, and lactate dehydrogenase. Increased myofibre diameter. Inhibited lipid peroxidation, reactive oxygen species.
[114]
Examine the immunostimulatory effect of polysaccharides from Schisandra chinensis.For the study, 50 mice were used, divided into five groups. Three of the groups were administered with 50, 100, or 200 mg/kg/day Schisandra chinensis polysaccharides IIa; the other two groups were the control and model groups. Period of study—10 days.Increased the phagocytic
activity of peritoneal macrophages and lymphocyte
transformation.
[115]
Examine the anti-diabetic effect of polysaccharides from
Schisandra chinensis.
For the study, 60 mice were used, divided into six groups: control group, control group with diabetes, alloxan-induced diabetic mice treated with sodium chloride solution, and three groups treated with 162, 324, or 648 mg/kg Schisandra chinensis polysaccharides, and a placebo group.
Period of study—21 days.
Decreased blood glucose levels. Improved lipid metabolism.[116]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Todorova, V.; Ivanov, K.; Delattre, C.; Nalbantova, V.; Karcheva-Bahchevanska, D.; Ivanova, S. Plant Adaptogens—History and Future Perspectives. Nutrients 2021, 13, 2861. https://doi.org/10.3390/nu13082861

AMA Style

Todorova V, Ivanov K, Delattre C, Nalbantova V, Karcheva-Bahchevanska D, Ivanova S. Plant Adaptogens—History and Future Perspectives. Nutrients. 2021; 13(8):2861. https://doi.org/10.3390/nu13082861

Chicago/Turabian Style

Todorova, Velislava, Kalin Ivanov, Cédric Delattre, Vanya Nalbantova, Diana Karcheva-Bahchevanska, and Stanislava Ivanova. 2021. "Plant Adaptogens—History and Future Perspectives" Nutrients 13, no. 8: 2861. https://doi.org/10.3390/nu13082861

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

Article Metrics

Back to TopTop