Next Article in Journal
A Novel Heterozygous Mutation c.1627G>T (p.Gly543Cys) in the SLC34A1 Gene in a Male Patient with Recurrent Nephrolithiasis and Early Onset Osteopenia: A Case Report
Next Article in Special Issue
Berberine Protects against High-Energy and Low-Protein Diet-Induced Hepatic Steatosis: Modulation of Gut Microbiota and Bile Acid Metabolism in Laying Hens
Previous Article in Journal
Interaction between Selected Adipokines and Musculoskeletal and Cardiovascular Systems: A Review of Current Knowledge
Previous Article in Special Issue
Relevance of Phytochemical Taste for Anti-Cancer Activity: A Statistical Inquiry
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Applications of Ginseng Natural Compounds for Health Management

by
Syed Sayeed Ahmad
1,2,
Khurshid Ahmad
1,2,
Ye Chan Hwang
1,
Eun Ju Lee
1,2 and
Inho Choi
1,2,*
1
Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
2
Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(24), 17290; https://doi.org/10.3390/ijms242417290
Submission received: 15 November 2023 / Revised: 6 December 2023 / Accepted: 7 December 2023 / Published: 9 December 2023

Abstract

:
Ginseng is usually consumed as a daily food supplement to improve health and has been shown to benefit skeletal muscle, improve glucose metabolism, and ameliorate muscle-wasting conditions, cardiovascular diseases, stroke, and the effects of aging and cancers. Ginseng has also been reported to help maintain bone strength and liver (digestion, metabolism, detoxification, and protein synthesis) and kidney functions. In addition, ginseng is often used to treat age-associated neurodegenerative disorders, and ginseng and ginseng-derived natural products are popular natural remedies for diseases such as diabetes, obesity, oxidative stress, and inflammation, as well as fungal, bacterial, and viral infections. Ginseng is a well-known herbal medication, known to alleviate the actions of several cytokines. The article concludes with future directions and significant application of ginseng compounds for researchers in understanding the promising role of ginseng in the treatment of several diseases. Overall, this study was undertaken to highlight the broad-spectrum therapeutic applications of ginseng compounds for health management.

1. Introduction

Natural products are viewed as a primary source of therapeutic agents and have been identified in plants, microorganisms, animals, insects, and marine organisms [1]. Natural products have diverse pharmacological properties and play important roles in drug discovery and development by serving as novel lead templates. Aspirin (from willow tree bark), digoxin (from the flower; Digitalis lanata), morphine (from opium), artemisinin, camptothecin, lovastatin, maytansine, reserpine, and silibinin are just a few examples of drugs directly or indirectly derived from natural products [2]. Some semi-synthetic therapeutic agents (hybrids of natural and synthetic sources), such as penicillin [3] and paclitaxel (an anti-cancer drug derived from the Pacific yew, Taxus brevifolia) [4], are typically produced by chemically transforming natural products [5]. The chemical, functional, and structural diversities of small molecule natural products have been explored [2]. The interactions between biological macromolecules (mainly proteins) and natural products explain the therapeutic efficacies of natural products. Furthermore, natural products do not cause as many adverse effects as synthetic compounds and combinatorial libraries [2].
The use of Chinese medicine has risen in popularity after the 2015 Nobel Prize was awarded for the discovery that artemisinin is an effective treatment for malaria [6]. Ginseng (a medicinal herb) and its derived natural products are amongst the most popular natural remedies and are used to treat various diseases and conditions such as diabetes [7], anti-oxidative [8], inflammation [9], cancers [10], fungal, bacterial, viral, stress [11], and neurodegenerative diseases (ND) [12], as well as brain ischemia [13], hypertension [14], obesity [15], cardiovascular diseases and stroke [16], sarcopenia [17], muscle-wasting conditions [18,19,20], muscle aging, and cancer cachexia [21,22,23]. Known side effects of ginseng include headaches, diarrhea, blood pressure changes, skin irritations, and vaginal bleeding [24]. Overall, ginseng has been reported to be a useful management option for many diseases, as is suggested by its name—Panax is derived from the Greek pan akheia, meaning “cures all diseases” [25]. A summary of known ginseng compounds is provided in Table 1.
Seventeen Panax species are now recognized, but commercial P. ginseng cultivars are largely found in South Korea and China [26]. The two most well-known are Panax ginseng and Panax quinquefolius [27]. The world’s major ginseng producers are China, South Korea, Canada, and the United States [27], and South Korea is the largest ginseng distributor [27]. P. ginseng and its ginsenoside components are non-toxic and used to treat chemotherapy-induced side effects such as nephrotoxicity, hepatotoxicity, cardiotoxicity, immunotoxicity, and hematopoietic suppression [28]. Here, we report the broad-spectrum therapeutic applications of known ginseng compounds. The strategy used to identify the articles include performing thorough searches in reputable academic databases such as PubMed, Scopus, SciFinder, Science Direct, Google Scholar, and the Scientific Information Database. The study focused on English language papers with particular keywords related to ginseng plants, natural compounds, biological investigations, and activities. The aim of this review was to explore ginseng-related natural compounds with potential use for the management of human health.

2. Protopanaxadiol (PPD) and Protopanaxatriol (PPT)

Protopanaxadiol (PPD) and protopanaxatriol (PPT) are active compounds found in members of the Panax genus, mainly in the roots, stems, leaves, and flowers. PPD is used to treat endometriosis and has been shown to significantly upregulate endometrial receptivity-related molecules, such as interleukin 6 family cytokine, insulin-like growth factor-binding protein 1, and collagens, to restrict the pelvic macrophage inflammatory response and to recover fertility in mice with endometriosis. Thus, the literature shows that PPD prevents and is a promising treatment for endometriosis [29]. On the other hand, PPT ginsenosides have pharmacological effects on the central nervous and cardiovascular systems [30], and PPT reportedly acts as a PPARγ antagonist [31]; thus, targeting PPARγ is considered a promising treatment option for obesity.

3. Ginsenoside F1 (GF1)

GF1 is a ginseng saponin isolated from a traditional Chinese medicine used to treat ischemic stroke. GF1 activates the IGF-1/IGF1R pathway [32] to promote angiogenesis, which reduces cerebral ischemia. In addition, GF1 may improve cerebrovascular function and accelerate recovery from ischemic stroke. In zebrafish, GF1 repaired vascular defects caused by axitinib [33], and in vivo and in vitro studies revealed that GF1 protects against Aβ accumulation. At 2.5 μM, GF1 reduced Aβ-induced cytotoxicity by reducing Aβ accumulation in mouse neuroblastoma neuro-2a (N2a) and human neuroblastoma SH-SY5Y neuronal cell lines. Additionally, GF1 reduced Aβ plaques in the hippocampus of (APP/PS1) double-transgenic AD mice [34,35]. Collectively, studies have shown GF1 is a highly active component in P. ginseng that can cross the BBB and has therapeutic potential for treating ND. Furthermore, GF1 reduces eosinophilic inflammation in chronic rhinosinusitis by enhancing NK cell activity [36].

4. Ginsenoside F2 (GF2)

GF2 is a minor component in P. ginseng with therapeutic applications in inflammatory diseases [37]. GF2 treatment attenuated liver damage in C57BL/6J WT mice (a model of alcoholic liver injury) [37] and suppressed the expression of TGF-β2 (a pro-apoptotic factor) to reduce hair loss in a dihydrotestosterone-induced mouse model [38]. It has been well established that excessive alcohol consumption can result in vitamin/mineral shortages (possibly due to liver damage) and subsequent hair loss because appropriate nourishment is required to maintain hair quality. In addition, alcohol suppresses nutrient breakdown and the body’s capacity to absorb nutrients. Currently, hair loss is a major issue among young men. We suggest that the effects of GF2 on known targets of hair loss should be investigated to manage this condition.

5. Ginsenoside F3 (GF3)

GF3 was isolated from the leaves of P. ginseng [39]. At concentrations ranging from 0.1 to 100 μmol/L, GF3 not only stimulated murine spleen cell proliferation but also raised the production of IL-2 and IFN-γ. It improved immunity by modulating the synthesis and gene expression of type 1 and 2 cytokines in murine spleen cells [40].

6. Ginsenoside F4 (GF4)

GF4 considerably enhanced the hyperglycemic state of db/db mice, alleviated dyslipidemia, and helped in SM glucose uptake. Protein tyrosine phosphatase 1B (PTP1B) is a major negative regulator of the insulin signaling pathway. The inhibition of this enzyme by GF4 resulted in increased insulin receptor and insulin receptor substrate 1 tyrosine phosphorylation and enhanced insulin sensitivity. Overall, GF4 activates the insulin signaling pathway by inhibiting PTP1B [41]. Further, GF4 has an inhibitory effect on human lymphocytoma Jurkat (JK) cell by inducing apoptosis [42].

7. Ginsenoside Ra1 (GRa1)

GRa1 is a key active ingredient in ginseng with immune regulatory, anti-inflammatory, and anti-oxidant properties [43]. Little is known about the effects of GRa1 on human health compared to other known ginseng compounds. GRa1 can affect the cardiovascular system, immune regulation, and nervous system [44]. It is present in both red ginseng powder and red ginseng concentrate samples [45]. GRa1 has been reported as a significant inhibitor of protein tyrosine kinase activation induced by in vitro hypoxia/reoxygenation in cultured human umbilical vein endothelial cells [46]. However, GRa1 has been shown to have anti-inflammatory and anti-oxidant properties, and thus it might be effective for the management of cancers and other inflammation-related diseases such as autoimmune diseases (rheumatoid arthritis), cardiovascular diseases (high blood pressure and heart disease), and gastrointestinal disorders (inflammatory bowel disease).

8. Ginsenoside Rb1 (GRb1)

In vitro and in vivo studies have demonstrated GRb1 to have diverse pharmacological applications in metabolic disorders due to its anti-apoptotic effects and ability to regulate oxidative stress, inflammatory responses, and autophagy. In addition, GRb1 suppresses obesity, hyperglycemia, and diabetes by regulating glycolipid metabolism and improving insulin and leptin sensitivities [47]. GRb1 may increase insulin sensitivity by downregulating 11β-hydroxysteroid dehydrogenase type I in T2D [48]. The administration of GRb1 (60 mg/kg of body mass intraperitoneally (i.p.)) daily for 12 days decreased adipose tissue and leptin levels in KK-Ay DM mice [49]. Furthermore, GRb1 (20 mg/kg of body mass, daily) reduced hepatic fat formation and enhanced insulin sensitivity in obese diabetic db/db mice; these effects were confirmed by reductions in liver weight and hepatic triglyceride contents [50]. GRb1 at 10 mg/kg of body mass (i.p.) daily considerably reduced body weight gain, improved glucose tolerance, and increased fasting plasma insulin levels in high-fat diet-induced obese mice and rats [51,52]. In addition, GRb1 increased GLUT4 translocation in C2C12 myotubes and 3T3-L1 cells by activating the adiponectin signaling pathway [53]. Notably, GRb1 is a major component of ginseng [53], which is frequently used as a natural medication in diabetic patients. Altogether, GRb1 has the potential to be used as an anti-obesity, anti-hyperglycemic, and anti-diabetic drug that affects multiple targets.

9. Ginsenoside Rb2 (GRb2)

It is a PPD-type saponin abundant in the stems and leaves of ginseng [54]. GRb2 significantly enhanced the viability of HT22 murine hippocampal neuronal cells [55] and inhibited the growth, migration, and invasion of colorectal cancer cells (HT29 and SW620 cell lines) [56]. GRb2 has been used to manage atherosclerosis [57], insulin resistance and obesity [58], endothelial cell senescence [59], and suppression of glutamate-induced neurotoxicity [55]. Thus, GRb2 appears to have potential for treating diabetes, obesity, tumors, viral infections, and cardiovascular conditions.

10. Ginsenoside Rb3 (GRb3)

GRb3 is a ginseng-derived natural product with cardioprotective properties [60] that can reduce the risk of myocardial infarction by inhibiting oxidative stress and suppressing inflammation [61]. It was found in an in vivo study to reduce the levels of the inflammatory markers NF-κB and CD45 and enhance the activities of crucial proteins of the contraction unit (cardiac troponin protein I (cTnI) and α-actinin) to recover cardiac function [62]. Furthermore, in vivo and in vitro studies have shown GRb3 mitigates oxidative stress by triggering the anti-oxidation signaling of PERK/Nrf2/HMOX1 [63]. Studies indicate that GRb3 may be helpful for treating heart-related disorders.

11. Ginsenoside Rc (GRc)

GRc has been reported to enhance bone development in ovariectomy-induced osteoporotic mice and to stimulate osteogenic differentiation in vitro through the Wnt/β-catenin signaling pathway [64]. Additionally, an in vivo study demonstrated that GRc administration significantly attenuated acetaminophen-induced hepatotoxicity, repaired liver damage, and improved survival [65]. In addition, in a dose-dependent manner, GRc reduced the proliferation and viability of 3T3L1 preadipocytes, adipocyte numbers, and lipid accumulation in maturing 3T3L1 preadipocytes, indicating it inhibited lipogenesis [66]. Collectively, it would appear that GRc has potential utility for managing several diseases.

12. Ginsenoside Rd (GRd)

The leading causes of muscle wasting are aging and cancer, and there are no effective cures for these conditions. However, GRd has been shown to alleviate muscle wasting. In mice, GRd administration suppressed age- and cancer-induced muscle atrophy and improved grip strength, hanging times, muscle mass, and muscle tissue cross-sectional areas; at the molecular level, GRd inhibited STAT3 phosphorylation and suppressed atrogin-1, muscle RING-finger protein-1 (MuRF-1), and myostatin levels [67]. Myostatin is a well-known inhibitor of muscle development [68]. MuRF1 is a key factor in the SM atrophy process that occurs during catabolic conditions, making MuRF1 a promising target for pharmaceutical therapies for muscle-wasting conditions [69]. SM improvement is necessary for healthy life [70,71]. Furthermore, GRd improved ischemic stroke-induced damage by suppressing oxidative stress and inflammation, prolonging neural cell survival by upregulating the endogenous anti-oxidant system and phosphoinositide-3-kinase/AKT signaling [72]. Thus, GRd may be an innovative natural product for treating muscle-wasting conditions, act as an anti-diabetic therapy by improving muscle health, and have anti-inflammatory, neuroprotective, and cardioprotective properties.

13. Ginsenoside Rf (GRf)

GRf is a constituent of Korean ginseng and upregulates markers of myoblast differentiation and mitochondrial biogenesis. GRf improves exercise tolerance in mice, possibly by enhancing mitochondrial biogenesis and myoblast differentiation via AMPK and p38 MAPK signaling pathways, suggesting that GRf boosts energy production to meet the increased demands of working muscle cells [73]. In addition, GRf has been reported to have neuroprotective and anti-inflammatory effects under hypoxic conditions. The binding of GRf at the active site of PPARγ suggests that it binds at the position used by known agonists [74]. In 3T3L1 adipocytes, GRf treatment downregulated PPARγ and perilipin (lipid droplet-associated protein) levels and reduced lipid accumulation [75]. These observations suggest GRf might be useful for treating obesity.

14. Ginsenoside Rg2 (GRg2)

GRg2 promotes porcine mesenchymal stem cell (pMSC) proliferation, prevents D-galactose-induced oxidative stress and senescence, and increases autophagic activity through the AMPK signaling pathway. Furthermore, long-term culture with GRg2 promoted pMSC proliferation, prevented replicative senescence, and preserved stemness [76]. In addition, the ability of GRg2 to exert its anti-atherosclerotic effects at the cellular and animal levels supports ginseng’s role as a functional dietary regulator [77]. Additionally, GRg2 was found to decrease mRNA levels of the inflammatory factors TNF, IL-6, and IL-8, and at 20 μM, it considerably suppressed IL-6, IL-8, and IL-1 [77]. These findings suggest a strategy for muscle regeneration based on the in vitro expansion of pMSCs.

15. Ginsenoside Rg3 (GRg3)

Acute pancreatitis (AP) is a systemic inflammatory response syndrome. In a study performed using a cerulein-induced murine model of AP to investigate the effect of GRg3, cerulein increased serum amylase, TNFα, IL-6, IL-1β, ROS, and Fe2+ levels, and GRg3 co-treatment decreased cerulein-induced ROS buildup and cell death in pancreatic tissues [78]. Due to the lack of an effective delivery approach, it is difficult to deliver GRg3 to body organs due to its hydrophobic nature. However, the intramyocardial injection of GRg3-loaded PEG-b-PPS nanoparticles in a rat ischemia–reperfusion model improved cardiac functions and reduced infarct sizes [79].

16. Ginsenoside Rg5 (GRg5)

GRg5 was administered to nude mice bearing A549/T tumors to combat multidrug resistance. Treatment with GRg5 and docetaxel considerably suppressed the growth of drug-resistant tumors without increasing toxicity compared to docetaxel alone at the same dose [80]. In another study, GRg5 remarkably suppressed breast cancer cell propagation by inducing mitochondria-mediated apoptosis and autophagic cell death. It was also found that GRg5 decreased the phosphorylation of PI3K, Akt, and mTOR and attenuated PI3K/Akt signaling in breast cancer [81]. Thus, GRg5 has therapeutic potential as a breast cancer treatment. Furthermore, GRg5 [82] and GRh1 [83] can both alleviate cisplatin-induced nephrotoxicity [82], presumably due to their anti-oxidant, anti-apoptotic, and anti-inflammatory effects.

17. Ginsenoside Rh1 (GRh1)

GRh1 is obtained from red ginseng and used to improve physical fitness. In a cisplatin-induced injury model, GRh1 enhanced the vitality of HK-2 cells and inhibited ROS production and apoptosis [83], which suggested that GRh1 has potential use for alleviating cisplatin-induced nephrotoxicity in cancer patients. In addition, GRh1 was found to have an anti-cancer effect on breast cancer by inhibiting the ROS-mediated PI3K/Akt pathway and causing cell cycle arrest, apoptosis, and autophagy [84]. An in vitro study reported that GRh1 (at 100 µM) significantly inhibited cell migration and invasion and effectively inhibited colorectal cancer development [85]. Currently, GRh1 is mostly being used as an anticancer agent and could be further explored for its use against various other cancers.

18. Ginsenoside Rh2 (GRh2)

GRh2 is obtained from the roots of P. ginseng and has been reported to have anti-tumor effects by immunomodulating the tumor microenvironment (TME) [86], regulating HMGB1/NF-κB signaling, and improving the oxygen–glucose deprived environment of cardiomyocytes [87]. However, the in vivo effects of GRh2 have not yet been well explored, and its therapeutic effects are unknown.

19. Ginsenoside Rh3 (GRh3)

Lung cancer is the second most common cause of cancer-related death after breast cancer [88]. In vitro, GRh3 (at 50 μM) inhibited the proliferation of A549 and PC9 cells, and in another in vitro study, GRh3 inhibited tumor growth by causing cell arrest in the G1 phase. In vivo, GRh3 at 50 and 100 mg/kg significantly inhibited lung cancer metastasis [89]. GRh3 also inhibited HCT116 (colon cancer) cell proliferation, invasion, migration, and arrested cells in the G1 phase by downregulating genes related to DNA replication [90]. In addition, GRh3 significantly ameliorated myocardial necrosis and caspase 3 levels in male Sprague Dawley rat myocardial tissues by hindering the p38 MAPK pathway [91].

20. Ginsenoside Rh4 (GRh4)

GRh4 significantly inhibited the production of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) in RAW264.7 cells (macrophages). JAK-STAT, TNF, NF-κB, and PI3K-Akt were identified as the main pathways used by GRh4 to reduce inflammation [92]. It has also been reported that GRh4 protects kidneys from cisplatin-induced oxidative injury [93]. GRh4 has been shown to have a high anti-lung adenocarcinoma efficacy in vitro and in vivo. Lung adenocarcinoma is a typical cellular breakdown in the lungs with a high harm that desperately should be treated [94]. Further, it is reported that GRh4 inhibits colorectal cancer cell proliferation [95], breast cancer growth [96], and delays SM aging through the SIRT1 pathway [97].

21. Ginsenoside Rh7 (GRh7)

GRh7 has been reported to inhibit H1299 (a lymph node-derived human non-small cell lung cancer cell line) growth (by 83%) and proliferation. In the same study, A549 (also an NSCLC cell line) and H1299 cells were used to study the time-dependent effects of GRh7 on cell growth. After treatment with 25 μM GRh7 for 4 days, A549 growth was inhibited by 72% and H1299 growth by 75% compared to non-treated controls [98]. These results suggest GRh7 has anti-cancerous properties that warrant further investigation.

22. Ginsenoside Rk1 (GRk1)

GRk1 is produced by thermally dehydrating GRg3, a saponin present in Panax ginseng Meyer. GRk1 effectively inhibited N-methyl-d-aspartate receptors in cultured hippocampal neurons [99] and protected human melanocytes from H2O2-induced death. PIG1 melanocytes were pretreated with GRk1 at 0, 0.1, 0.2, or 0.4 mM for 2 h and then exposed to H2O2 under cytotoxic conditions (at 1.0 mM for 24 h). GRk1 pretreatment at 0.2 or 0.4 mM for 2 h considerably improved cell viability and decreased cell shrinkage versus H2O2 treatment controls [100]. In another study, GRk1 treatment reversed cisplatin-induced increases in the protein levels of Bax, cleaved caspase 3 and 9, and Bcl-2 [101]. GRk1 was also reported to have anti-tumor activity against lung squamous cell carcinoma [102], and at 30 mg/kg, GRk1 injections markedly inhibited tumor xenograft growth [103].

23. Ginsenoside Rk3 (GRk3)

GRk3 is a key bioactive constituent in ginseng and has robust anti-oxidant properties. GRk3 was found to enhance neuronal apoptosis, decrease intracellular ROS production, and restore mitochondrial membrane potentials in PC12 and primary neuronal cells. GRk3 also improved spatial learning and reduced memory deficits in an amyloid precursor protein (APP)/presenilin 1 (PS1) double transgenic mouse model of Alzheimer’s disease (AD) [104]. AD has devastating effects on society with a limited number of approaches for its treatment [105]. AD is caused by mutations in one of the genes that codes for APP and presenilins 1 and 2. The majority of these gene mutations boost Aβ42 production [106]. Parkinson’s disease (PD) is the second most common ND without any proper cure [107] and is categorized as a movement disorder. Natural products for the management of PD have also been reported [108]. Additionally, several other natural products have been reported for the management of AD and other NDs [109,110]. The anti-cancer effects of GRk3 were also checked in Eca109 and KYSE150 cell lines (both esophageal squamous carcinoma cell lines), and it was observed that GRk3 suppressed proliferation and colony formation for both cell types. This inhibition was ascribed to blocking of the PI3K/Akt/mTOR pathway and consequent activation of apoptosis and autophagy [111]. GRk3 might be an effective anti-tumor agent for esophageal cancer and for the treatment of renal dysfunctions caused by cisplatin-induced oxidative injury [93]. Furthermore, GRk3 improved hematopoietic function in myelosuppressed mice [112] and inhibited the proliferation, migration, and invasion associated with the extramedullary infiltration of leukemia [113].

24. Ginsenoside Ro (GRo)

GRo is a primary saponin in P. ginseng C.A. Meyer with several biological actions. In B16F10 tumor-bearing mice, GRo significantly inhibited tumor growth [114], LPS-induced lung damage, and TNF, IL-6, and IL-1 transcript levels in tumor tissues. Furthermore, in a dose-dependent manner, GRo suppressed the phosphorylation of NF-κB and MAPKs and the nuclear translocation of the p65 subunit. These findings imply that GRo targets inflammation by directly inhibiting the TLR4 signaling pathway [115]. The anti-inflammatory effects of GRo have been linked to a significant decrease in levels of pro-inflammatory cytokines generated by lipopolysaccharides. In a dose-dependent manner, GRo enhances cell survival while lowering reactive oxygen species (ROS) and nitric oxide generation produced by lipopolysaccharides [116]. Further, GRo improves obesity and insulin resistance in mice [117].

25. Floralginsenoside A (FGA)

Melanin provides UV protection and removes ROS from the skin. However, excessive melanin production and its accumulation in the skin can result in pigmentation disorders (e.g., solar lentigo, melasma, and freckles) [118]. Melan-a cells, an immortalized C57BL/6 mouse melanocyte cell line containing high levels of melanin, were used to investigate the melanin-inhibitory activity of FGA. At a concentration of 160 μM, FGA inhibited melanin activity by 23.9% without causing cytotoxicity. Tyrosinase is a key player in the biosynthesis of melanin, and the main mechanism underlying the anti-melanogenesis effects of inhibitory agents involves the downregulation of MITF (microphthalmia-related transcription factor) due to the ERK-induced phosphorylation of MITF at serine-73, which triggers the ubiquitination of MITF and its subsequent degradation. The phosphorylation of MITF at serine 29 activates Akt signaling and inhibits melanin production. FGA can block MITF–tyrosinase signaling and/or activate ERK–Akt signaling, which are both involved in melanogenesis. In addition, FGA treatment dose-dependently decreases the expressions of tyrosinase and MITF. Furthermore, FGA (at 160 μM) significantly and dose-dependently augmented phospho-ERK and Akt signaling pathways [119]. Tyrosinase activity and melanin content inhibition may result in skin whitening. However, the potential carcinogenic side effects of the agent currently used (kojic acid) to whiten skin [120] necessitates the development of new, safer, more effective depigmenting agents, and natural products feature prominently in these studies. Makeup/cosmetic production is expanding, especially in South Korea. Thus, the identification of natural makeup agents is likely to result in commercially attractive health and skin care products.

26. Future Perspectives

This study was performed to summarize what is known about the influence of ginseng and its derived natural products on diseases. It is well known that synthetic drugs are associated with adverse effects and that herbal remedies have been used for millennia and are relatively free of side effects. As a result, perceptions are changing in favor of natural therapies and traditional medicines. In particular, ginseng has been administered as an herbal medicine for thousands of years and is now commercially available in pill and tea forms. Intriguingly, one clinical study reported that patients who took ginseng after curative surgery had a 38% higher overall survival rate and a 35% higher 5-year disease-free rate [19,121].
The roles and functions of a number of ginseng compounds are listed in Table 2. Several of the natural products isolated from ginseng are therapeutically beneficial. Natural products have been thoroughly investigated in vitro and in vivo, and we suggest in silico studies be undertaken to aid in the primary screening of ginseng natural products. It has been reported that in silico-screened compounds produce better results during subsequent in vitro or in vivo testing [122,123,124]. Most of the natural products mentioned have anti-diabetic, anti-neuroprotective, anti-cancer, anti-oxidant, and anti-inflammatory effects. We suggest that the repurposing of these compounds be attempted to improve their therapeutic effects.

27. Conclusions

Ginseng and ginseng-derived natural products are attractive candidates for the treatment of several disease. Furthermore, this study showed that ginseng and its derived natural products are powerful therapeutic agents/supplements that improve health and increase energy. Therefore, we suggest that clinical trials be performed to confirm the therapeutic efficacies of ginsenosides in cancer, stroke, obesity, aging, and NDs.

Author Contributions

S.S.A., K.A. and Y.C.H.: conceptualization; writing—original draft preparation; S.S.A. and K.A. data curation, E.J.L. and I.C.: supervision, writing—reviewing and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Basic Science Research Program of the National Research Foundation of Korea (NRF), funded by the Ministry of Education (2020R1A6A1A03044512), the National Research Foundation of Korea (NRF), and the Korean government (NRF-2021R1A2C2004177). In addition, the study was supported by the Forestry (IPET) through High Value-added Food Technology Development Program of the Korea Institute of Planning and Evaluation for Technology in Food and Agriculture, funded by the Ministry of Agriculture, Food, and Rural Affairs (MAFRA) (Grant nos. 322008-5).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Ji, H.F.; Li, X.J.; Zhang, H.Y. Natural products and drug discovery. Can thousands of years of ancient medical knowledge lead us to new and powerful drug combinations in the fight against cancer and dementia? EMBO Rep. 2009, 10, 194–200. [Google Scholar] [CrossRef] [PubMed]
  2. Mathur, S.; Hoskins, C. Drug development: Lessons from nature. Biomed. Rep. 2017, 6, 612–614. [Google Scholar] [CrossRef]
  3. Oshiro, B.T. The semisynthetic penicillins. Prim. Care Update OB/GYNS 1999, 6, 56–60. [Google Scholar] [CrossRef]
  4. Lahlou, M. The success of natural products in drug discovery. Pharmacol. Pharmacy 2013, 4, 17–31. [Google Scholar] [CrossRef]
  5. Cragg, G.M.; Newman, D.J. Natural products: A continuing source of novel drug leads. Biochim. Biophys. Acta 2013, 1830, 3670–3695. [Google Scholar] [CrossRef] [PubMed]
  6. Tu, Y. Artemisinin-A Gift from Traditional Chinese Medicine to the World (Nobel Lecture). Angew. Chem. Int. Ed. Engl. 2016, 55, 10210–10226. [Google Scholar] [CrossRef]
  7. Bai, L.; Gao, J.; Wei, F.; Zhao, J.; Wang, D.; Wei, J. Therapeutic Potential of Ginsenosides as an Adjuvant Treatment for Diabetes. Front. Pharmacol. 2018, 9, 423. [Google Scholar] [CrossRef]
  8. He, N.W.; Zhao, Y.; Guo, L.; Shang, J.; Yang, X.B. Antioxidant, antiproliferative, and pro-apoptotic activities of a saponin extract derived from the roots of Panax notoginseng (Burk.) F.H. Chen. J. Med. Food 2012, 15, 350–359. [Google Scholar] [CrossRef] [PubMed]
  9. Lee, I.S.; Uh, I.; Kim, K.S.; Kim, K.H.; Park, J.; Kim, Y.; Jung, J.H.; Jung, H.J.; Jang, H.J. Anti-Inflammatory Effects of Ginsenoside Rg3 via NF-κB Pathway in A549 Cells and Human Asthmatic Lung Tissue. J. Immunol. Res. 2016, 2016, 7521601. [Google Scholar] [CrossRef]
  10. Lu, M.; Fei, Z.; Zhang, G. Synergistic anticancer activity of 20(S)-Ginsenoside Rg3 and Sorafenib in hepatocellular carcinoma by modulating PTEN/Akt signaling pathway. Biomed. Pharmacother. 2018, 97, 1282–1288. [Google Scholar] [CrossRef]
  11. Lee, M.H.; Lee, B.H.; Jung, J.Y.; Cheon, D.S.; Kim, K.T.; Choi, C. Antiviral effect of Korean red ginseng extract and ginsenosides on murine norovirus and feline calicivirus as surrogates for human norovirus. J. Ginseng Res. 2011, 35, 429–435. [Google Scholar] [CrossRef]
  12. Wang, Y.; Yang, G.; Gong, J.; Lu, F.; Diao, Q.; Sun, J.; Zhang, K.; Tian, J.; Liu, J. Ginseng for Alzheimer’s Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Curr. Top. Med. Chem. 2016, 16, 529–536. [Google Scholar] [CrossRef]
  13. Kim, K.H.; Lee, D.; Lee, H.L.; Kim, C.E.; Jung, K.; Kang, K.S. Beneficial effects of Panax ginseng for the treatment and prevention of neurodegenerative diseases: Past findings and future directions. J. Ginseng Res. 2018, 42, 239–247. [Google Scholar] [CrossRef]
  14. Park, S.H.; Chung, S.; Chung, M.Y.; Choi, H.K.; Hwang, J.T.; Park, J.H. Effects of Panax ginseng on hyperglycemia, hypertension, and hyperlipidemia: A systematic review and meta-analysis. J. Ginseng Res. 2022, 46, 188–205. [Google Scholar] [CrossRef]
  15. Li, Z.; Ji, G.E. Ginseng and obesity. J. Ginseng Res. 2018, 42, 1–8. [Google Scholar] [CrossRef]
  16. Lee, C.H.; Kim, J.H. A review on the medicinal potentials of ginseng and ginsenosides on cardiovascular diseases. J. Ginseng Res. 2014, 38, 161–166. [Google Scholar] [CrossRef] [PubMed]
  17. Cho, D.E.; Choi, G.M.; Lee, Y.S.; Hong, J.P.; Yeom, M.; Lee, B.; Hahm, D.H. Long-term administration of red ginseng non-saponin fraction rescues the loss of skeletal muscle mass and strength associated with aging in mice. J. Ginseng Res. 2022, 46, 657–665. [Google Scholar] [CrossRef] [PubMed]
  18. Kim, T.J.; Pyun, D.H.; Kim, M.J.; Jeong, J.H.; Abd El-Aty, A.M.; Jung, T.W. Ginsenoside compound K ameliorates palmitate-induced atrophy in C2C12 myotubes via promyogenic effects and AMPK/autophagy-mediated suppression of endoplasmic reticulum stress. J. Ginseng Res. 2022, 46, 444–453. [Google Scholar] [CrossRef] [PubMed]
  19. Ahmad, S.S.; Chun, H.J.; Ahmad, K.; Choi, I. Therapeutic applications of ginseng for skeletal muscle-related disorder management. J. Ginseng Res. 2023; in press. [Google Scholar] [CrossRef]
  20. Ahmad, K.; Shaikh, S.; Ahmad, S.S.; Lee, E.J.; Choi, I. Cross-Talk Between Extracellular Matrix and Skeletal Muscle: Implications for Myopathies. Front. Pharmacol. 2020, 11, 142. [Google Scholar] [CrossRef]
  21. Kim, R.; Kim, J.W.; Lee, S.J.; Bae, G.U. Ginsenoside Rg3 protects glucocorticoid-induced muscle atrophy in vitro through improving mitochondrial biogenesis and myotube growth. Mol. Med. Rep. 2022, 25, 94. [Google Scholar] [CrossRef]
  22. Ahmad, S.S.; Ahmad, K.; Shaikh, S.; You, H.J.; Lee, E.Y.; Ali, S.; Lee, E.J.; Choi, I. Molecular Mechanisms and Current Treatment Options for Cancer Cachexia. Cancers 2022, 14, 2107. [Google Scholar] [CrossRef]
  23. Lee, E.J.; Ahmad, S.S.; Lim, J.H.; Ahmad, K.; Shaikh, S.; Lee, Y.S.; Park, S.J.; Jin, J.O.; Lee, Y.H.; Choi, I. Interaction of Fibromodulin and Myostatin to Regulate Skeletal Muscle Aging: An Opposite Regulation in Muscle Aging, Diabetes, and Intracellular Lipid Accumulation. Cells 2021, 10, 2083. [Google Scholar] [CrossRef]
  24. Ginseng. Drugs and Lactation Database (LactMed®); Bethesda: Rockville, MD, USA, 2006. [Google Scholar]
  25. Yun, T.K. Brief introduction of Panax ginseng C.A. Meyer. J. Korean Med. Sci. 2001, 16, S3–S5. [Google Scholar] [CrossRef] [PubMed]
  26. Zhang, H.; Abid, S.; Ahn, J.C.; Mathiyalagan, R.; Kim, Y.J.; Yang, D.C.; Wang, Y. Characteristics of Panax ginseng Cultivars in Korea and China. Molecules 2020, 25, 2635. [Google Scholar] [CrossRef] [PubMed]
  27. Baeg, I.H.; So, S.H. The world ginseng market and the ginseng (Korea). J. Ginseng Res. 2013, 37, 1–7. [Google Scholar] [CrossRef] [PubMed]
  28. Wan, Y.; Wang, J.; Xu, J.F.; Tang, F.; Chen, L.; Tan, Y.Z.; Rao, C.L.; Ao, H.; Peng, C. Panax ginseng and its ginsenosides: Potential candidates for the prevention and treatment of chemotherapy-induced side effects. J. Ginseng Res. 2021, 45, 617–630. [Google Scholar] [CrossRef]
  29. Lai, Z.Z.; Yang, H.L.; Shi, J.W.; Shen, H.H.; Wang, Y.; Chang, K.K.; Zhang, T.; Ye, J.F.; Sun, J.S.; Qiu, X.M.; et al. Protopanaxadiol improves endometriosis associated infertility and miscarriage in sex hormones receptors-dependent and independent manners. Int. J. Biol. Sci. 2021, 17, 1878–1894. [Google Scholar] [CrossRef]
  30. Zhou, C.; Gong, T.; Chen, J.; Chen, T.; Yang, J.; Zhu, P. Production of a Novel Protopanaxatriol-Type Ginsenoside by Yeast Cell Factories. Bioengineering 2023, 10, 463. [Google Scholar] [CrossRef]
  31. Zhang, Y.; Yu, L.; Cai, W.; Fan, S.; Feng, L.; Ji, G.; Huang, C. Protopanaxatriol, a novel PPARgamma antagonist from Panax ginseng, alleviates steatosis in mice. Sci. Rep. 2014, 4, 7375. [Google Scholar] [CrossRef]
  32. Ahmad, S.S.; Ahmad, K.; Lee, E.J.; Lee, Y.H.; Choi, I. Implications of Insulin-like Growth Factor-1 in Skeletal Muscle and Various Diseases. Cells 2020, 9, 1773. [Google Scholar] [CrossRef]
  33. Zhang, J.; Liu, M.; Huang, M.; Chen, M.; Zhang, D.; Luo, L.; Ye, G.; Deng, L.; Peng, Y.; Wu, X.; et al. Ginsenoside F1 promotes angiogenesis by activating the IGF-1/IGF1R pathway. Pharmacol. Res. 2019, 144, 292–305. [Google Scholar] [CrossRef]
  34. Yun, Y.J.; Park, B.H.; Hou, J.; Oh, J.P.; Han, J.H.; Kim, S.C. Ginsenoside F1 Protects the Brain against Amyloid Beta-Induced Toxicity by Regulating IDE and NEP. Life 2022, 12, 58. [Google Scholar] [CrossRef] [PubMed]
  35. Han, J.; Oh, J.P.; Yoo, M.; Cui, C.H.; Jeon, B.M.; Kim, S.C.; Han, J.H. Minor ginsenoside F1 improves memory in APP/PS1 mice. Mol. Brain 2019, 12, 77. [Google Scholar] [CrossRef]
  36. Kim, S.J.; Lee, J.; Choi, W.S.; Kim, H.J.; Kim, M.Y.; Kim, S.C.; Kim, H.S. Ginsenoside F1 Attenuates Eosinophilic Inflammation in Chronic Rhinosinusitis by Promoting NK Cell Function. J. Ginseng Res. 2021, 45, 695–705. [Google Scholar] [CrossRef] [PubMed]
  37. Kim, M.H.; Kim, H.H.; Jeong, J.M.; Shim, Y.R.; Lee, J.H.; Kim, Y.E.; Ryu, T.; Yang, K.; Kim, K.R.; Jeon, B.M.; et al. Ginsenoside F2 attenuates chronic-binge ethanol-induced liver injury by increasing regulatory T cells and decreasing Th17 cells. J. Ginseng Res. 2020, 44, 815–822. [Google Scholar] [CrossRef] [PubMed]
  38. Shin, H.S.; Park, S.Y.; Hwang, E.S.; Lee, D.G.; Mavlonov, G.T.; Yi, T.H. Ginsenoside F2 reduces hair loss by controlling apoptosis through the sterol regulatory element-binding protein cleavage activating protein and transforming growth factor-beta pathways in a dihydrotestosterone-induced mouse model. Biol. Pharm. Bull. 2014, 37, 755–763. [Google Scholar] [CrossRef] [PubMed]
  39. Yoshizaki, K.; Yahara, S. New triterpenoid saponins from leaves of Panax japonicus (3). Saponins of the specimens collected in Miyazaki prefecture. Nat. Prod. Commun. 2012, 7, 491–493. [Google Scholar] [CrossRef] [PubMed]
  40. Yu, J.L.; Dou, D.Q.; Chen, X.H.; Yang, H.Z.; Guo, N.; Cheng, G.F. Immunoenhancing activity of protopanaxatriol-type ginsenoside-F3 in murine spleen cells. Acta Pharmacol. Sin. 2004, 25, 1671–1676. [Google Scholar]
  41. Zhao, Y.; Liu, Y.; Deng, J.; Zhu, C.; Ma, X.; Jiang, M.; Fan, D. Ginsenoside F4 Alleviates Skeletal Muscle Insulin Resistance by Regulating PTP1B in Type II Diabetes Mellitus. J. Agric. Food Chem. 2023, 71, 14263–14275. [Google Scholar] [CrossRef]
  42. Chen, B.; Shen, Y.P.; Zhang, D.F.; Cheng, J.; Jia, X.B. The apoptosis-inducing effect of ginsenoside F4 from steamed notoginseng on human lymphocytoma JK cells. Nat. Prod. Res. 2013, 27, 2351–2354. [Google Scholar] [CrossRef] [PubMed]
  43. Liu, J.; Nile, S.H.; Xu, G.; Wang, Y.; Kai, G. Systematic exploration of Astragalus membranaceus and Panax ginseng as immune regulators: Insights from the comparative biological and computational analysis. Phytomedicine 2021, 86, 153077. [Google Scholar] [CrossRef] [PubMed]
  44. Sim, U.; Sung, J.; Lee, H.; Heo, H.; Jeong, H.S.; Lee, J. Effect of calcium chloride and sucrose on the composition of bioactive compounds and antioxidant activities in buckwheat sprouts. Food Chem. 2020, 312, 126075. [Google Scholar] [CrossRef]
  45. Park, H.W.; In, G.; Han, S.T.; Lee, M.W.; Kim, S.Y.; Kim, K.T.; Cho, B.G.; Han, G.H.; Chang, I.M. Simultaneous determination of 30 ginsenosides in Panax ginseng preparations using ultra performance liquid chromatography. J. Ginseng Res. 2013, 37, 457–467. [Google Scholar] [CrossRef]
  46. Dou, D.Q.; Zhang, Y.W.; Zhang, L.; Chen, Y.J.; Yao, X.S. The inhibitory effects of ginsenosides on protein tyrosine kinase activated by hypoxia/reoxygenation in cultured human umbilical vein endothelial cells. Planta Med. 2001, 67, 19–23. [Google Scholar] [CrossRef]
  47. Zhou, P.; Xie, W.; He, S.; Sun, Y.; Meng, X.; Sun, G.; Sun, X. Ginsenoside Rb1 as an Anti-Diabetic Agent and Its Underlying Mechanism Analysis. Cells 2019, 8, 204. [Google Scholar] [CrossRef] [PubMed]
  48. Song, B.; Ding, L.; Zhang, H.; Chu, Y.; Chang, Z.; Yu, Y.; Guo, D.; Zhang, S.; Liu, X. Ginsenoside Rb1 increases insulin sensitivity through suppressing 11beta-hydroxysteroid dehydrogenase type I. Am. J. Transl. Res. 2017, 9, 1049–1057. [Google Scholar] [PubMed]
  49. Zhong, Z.D.; Wang, C.M.; Wang, W.; Shen, L.; Chen, Z.H. Major hypoglycemic ingredients of Panax notoginseng saponins for treating diabetes. Sichuan Da Xue Xue Bao Yi Xue Ban. 2014, 45, 235–239. [Google Scholar]
  50. Yu, X.; Ye, L.; Zhang, H.; Zhao, J.; Wang, G.; Guo, C.; Shang, W. Ginsenoside Rb1 ameliorates liver fat accumulation by upregulating perilipin expression in adipose tissue of db/db obese mice. J. Ginseng Res. 2015, 39, 199–205. [Google Scholar] [CrossRef] [PubMed]
  51. Wu, Y.; Yu, Y.; Szabo, A.; Han, M.; Huang, X.F. Central inflammation and leptin resistance are attenuated by ginsenoside Rb1 treatment in obese mice fed a high-fat diet. PLoS ONE 2014, 9, e92618. [Google Scholar] [CrossRef]
  52. Xiong, Y.; Shen, L.; Liu, K.J.; Tso, P.; Xiong, Y.; Wang, G.; Woods, S.C.; Liu, M. Antiobesity and antihyperglycemic effects of ginsenoside Rb1 in rats. Diabetes 2010, 59, 2505–2512. [Google Scholar] [CrossRef]
  53. Tabandeh, M.R.; Jafari, H.; Hosseini, S.A.; Hashemitabar, M. Ginsenoside Rb1 stimulates adiponectin signaling in C2C12 muscle cells through up-regulation of AdipoR1 and AdipoR2 proteins. Pharm. Biol. 2015, 53, 125–132. [Google Scholar] [CrossRef] [PubMed]
  54. Miao, L.; Yang, Y.; Li, Z.; Fang, Z.; Zhang, Y.; Han, C.C. Ginsenoside Rb2: A review of pharmacokinetics and pharmacological effects. J. Ginseng Res. 2022, 46, 206–213. [Google Scholar] [CrossRef] [PubMed]
  55. Kim, D.H.; Kim, D.W.; Jung, B.H.; Lee, J.H.; Lee, H.; Hwang, G.S.; Kang, K.S.; Lee, J.W. Ginsenoside Rb2 suppresses the glutamate-mediated oxidative stress and neuronal cell death in HT22 cells. J. Ginseng Res. 2019, 43, 326–334. [Google Scholar] [CrossRef] [PubMed]
  56. Phi, L.T.H.; Wijaya, Y.T.; Sari, I.N.; Yang, Y.G.; Lee, Y.K.; Kwon, H.Y. The anti-metastatic effect of ginsenoside Rb2 in colorectal cancer in an EGFR/SOX2-dependent manner. Cancer Med. 2018, 7, 5621–5631. [Google Scholar] [CrossRef]
  57. Wang, S.; Yang, S.; Chen, Y.; Chen, Y.; Li, R.; Han, S.; Kamili, A.; Wu, Y.; Zhang, W. Ginsenoside Rb2 Alleviated Atherosclerosis by Inhibiting M1 Macrophages Polarization Induced by MicroRNA-216a. Front. Pharmacol. 2021, 12, 764130. [Google Scholar] [CrossRef] [PubMed]
  58. Lin, Y.; Hu, Y.; Hu, X.; Yang, L.; Chen, X.; Li, Q.; Gu, X. Ginsenoside Rb2 improves insulin resistance by inhibiting adipocyte pyroptosis. Adipocyte 2020, 9, 302–312. [Google Scholar] [CrossRef] [PubMed]
  59. Chen, Y.; Wang, S.; Yang, S.; Li, R.; Yang, Y.; Chen, Y.; Zhang, W. Inhibitory role of ginsenoside Rb2 in endothelial senescence and inflammation mediated by microRNA-216a. Mol. Med. Rep. 2021, 23, 1–11. [Google Scholar] [CrossRef]
  60. Heras, G.; Namuduri, A.V.; Traini, L.; Shevchenko, G.; Falk, A.; Bergstrom Lind, S.; Jia, M.; Tian, G.; Gastaldello, S. Muscle RING-finger protein-1 (MuRF1) functions and cellular localization are regulated by SUMO1 post-translational modification. J. Mol. Cell Biol. 2019, 11, 356–370. [Google Scholar] [CrossRef]
  61. Chen, X.; Liu, T.; Wang, Q.; Wang, H.; Xue, S.; Jiang, Q.; Li, J.; Li, C.; Wang, W.; Wang, Y. Synergistic Effects of Ginsenoside Rb3 and Ferruginol in Ischemia-Induced Myocardial Infarction. Int. J. Mol. Sci. 2022, 23, 15935. [Google Scholar] [CrossRef]
  62. Shao, M.; Gao, P.; Cheng, W.; Ma, L.; Yang, Y.; Lu, L.; Li, C.; Wang, W.; Wang, Y. Ginsenoside Rb3 upregulates sarcoplasmic reticulum Ca2+-ATPase expression and improves the contractility of cardiomyocytes by inhibiting the NF-κB pathway. Biomed. Pharmacother. 2022, 154, 113661. [Google Scholar] [CrossRef]
  63. Sun, J.; Yu, X.; Huangpu, H.; Yao, F. Ginsenoside Rb3 protects cardiomyocytes against hypoxia/reoxygenation injury via activating the antioxidation signaling pathway of PERK/Nrf2/HMOX1. Biomed. Pharmacother. 2019, 109, 254–261. [Google Scholar] [CrossRef] [PubMed]
  64. Yang, N.; Zhang, X.; Li, L.; Xu, T.; Li, M.; Zhao, Q.; Yu, J.; Wang, J.; Liu, Z. Ginsenoside Rc Promotes Bone Formation in Ovariectomy-Induced Osteoporosis In Vivo and Osteogenic Differentiation In Vitro. Int. J. Mol. Sci. 2022, 23, 6187. [Google Scholar] [CrossRef] [PubMed]
  65. Zhong, Y.; Chen, Y.; Pan, Z.; Tang, K.; Zhong, G.; Guo, J.; Cui, T.; Li, T.; Duan, S.; Yang, X.; et al. Ginsenoside Rc, as an FXR activator, alleviates acetaminophen-induced hepatotoxicity via relieving inflammation and oxidative stress. Front. Pharmacol. 2022, 13, 1027731. [Google Scholar] [CrossRef] [PubMed]
  66. Yang, J.W.; Kim, S.S. Ginsenoside Rc promotes anti-adipogenic activity on 3T3-L1 adipocytes by down-regulating C/EBPalpha and PPARgamma. Molecules 2015, 20, 1293–1303. [Google Scholar] [CrossRef] [PubMed]
  67. Wijaya, Y.T.; Setiawan, T.; Sari, I.N.; Park, K.; Lee, C.H.; Cho, K.W.; Lee, Y.K.; Lim, J.Y.; Yoon, J.K.; Lee, S.H.; et al. Ginsenoside Rd ameliorates muscle wasting by suppressing the signal transducer and activator of transcription 3 pathway. J. Cachexia Sarcopenia Muscle 2022, 13, 3149–3162. [Google Scholar] [CrossRef] [PubMed]
  68. Baig, M.H.; Ahmad, K.; Moon, J.S.; Park, S.Y.; Ho Lim, J.; Chun, H.J.; Qadri, A.F.; Hwang, Y.C.; Jan, A.T.; Ahmad, S.S.; et al. Myostatin and its Regulation: A Comprehensive Review of Myostatin Inhibiting Strategies. Front. Physiol. 2022, 13, 876078. [Google Scholar] [CrossRef]
  69. Peris-Moreno, D.; Taillandier, D.; Polge, C. MuRF1/TRIM63, Master Regulator of Muscle Mass. Int. J. Mol. Sci. 2020, 21, 6663. [Google Scholar] [CrossRef]
  70. Ahmad, K.; Shaikh, S.; Lim, J.H.; Ahmad, S.S.; Chun, H.J.; Lee, E.J.; Choi, I. Therapeutic application of natural compounds for skeletal muscle-associated metabolic disorders: A review on diabetes perspective. Biomed. Pharmacother. 2023, 168, 115642. [Google Scholar] [CrossRef] [PubMed]
  71. Ahmad, K.; Shaikh, S.; Chun, H.J.; Ali, S.; Lim, J.H.; Ahmad, S.S.; Lee, E.J.; Choi, I. Extracellular matrix: The critical contributor to skeletal muscle regeneration-a comprehensive review. Inflamm. Regen. 2023, 43, 58. [Google Scholar] [CrossRef]
  72. Nabavi, S.F.; Sureda, A.; Habtemariam, S.; Nabavi, S.M. Ginsenoside Rd and ischemic stroke; a short review of literatures. J. Ginseng Res. 2015, 39, 299–303. [Google Scholar] [CrossRef] [PubMed]
  73. Lim, W.C.; Shin, E.J.; Lim, T.G.; Choi, J.W.; Song, N.E.; Hong, H.D.; Cho, C.W.; Rhee, Y.K. Ginsenoside Rf Enhances Exercise Endurance by Stimulating Myoblast Differentiation and Mitochondrial Biogenesis in C2C12 Myotubes and ICR Mice. Foods 2022, 11, 1709. [Google Scholar] [CrossRef] [PubMed]
  74. Song, H.; Park, J.; Choi, K.; Lee, J.; Chen, J.; Park, H.J.; Yu, B.I.; Iida, M.; Rhyu, M.R.; Lee, Y. Ginsenoside Rf inhibits cyclooxygenase-2 induction via peroxisome proliferator-activated receptor gamma in A549 cells. J. Ginseng Res. 2019, 43, 319–325. [Google Scholar] [CrossRef] [PubMed]
  75. Siraj, F.M.; Natarajan, S.; Huq, M.A.; Kim, Y.J.; Yang, D.C. Structural investigation of ginsenoside Rf with PPARgamma major transcriptional factor of adipogenesis and its impact on adipocyte. J. Ginseng Res. 2015, 39, 141–147. [Google Scholar] [CrossRef]
  76. Che, L.; Zhu, C.; Huang, L.; Xu, H.; Ma, X.; Luo, X.; He, H.; Zhang, T.; Wang, N. Ginsenoside Rg2 Promotes the Proliferation and Stemness Maintenance of Porcine Mesenchymal Stem Cells through Autophagy Induction. Foods 2023, 12, 1075. [Google Scholar] [CrossRef]
  77. Xue, Q.; Yu, T.; Wang, Z.; Fu, X.; Li, X.; Zou, L.; Li, M.; Cho, J.Y.; Yang, Y. Protective effect and mechanism of ginsenoside Rg2 on atherosclerosis. J. Ginseng Res. 2023, 47, 237–245. [Google Scholar] [CrossRef] [PubMed]
  78. Shan, Y.; Li, J.; Zhu, A.; Kong, W.; Ying, R.; Zhu, W. Ginsenoside Rg3 ameliorates acute pancreatitis by activating the NRF2/HO-1-mediated ferroptosis pathway. Int. J. Mol. Med. 2022, 50, 89. [Google Scholar] [CrossRef]
  79. Li, L.; Wang, Y.; Guo, R.; Li, S.; Ni, J.; Gao, S.; Gao, X.; Mao, J.; Zhu, Y.; Wu, P.; et al. Ginsenoside Rg3-loaded, reactive oxygen species-responsive polymeric nanoparticles for alleviating myocardial ischemia-reperfusion injury. J. Control Release 2020, 317, 259–272. [Google Scholar] [CrossRef] [PubMed]
  80. Feng, S.L.; Luo, H.B.; Cai, L.; Zhang, J.; Wang, D.; Chen, Y.J.; Zhan, H.X.; Jiang, Z.H.; Xie, Y. Ginsenoside Rg5 overcomes chemotherapeutic multidrug resistance mediated by ABCB1 transporter: In vitro and in vivo study. J. Ginseng Res. 2020, 44, 247–257. [Google Scholar] [CrossRef]
  81. Liu, Y.; Fan, D. The Preparation of Ginsenoside Rg5, Its Antitumor Activity against Breast Cancer Cells and Its Targeting of PI3K. Nutrients 2020, 12, 246. [Google Scholar] [CrossRef] [PubMed]
  82. Li, W.; Yan, M.H.; Liu, Y.; Liu, Z.; Wang, Z.; Chen, C.; Zhang, J.; Sun, Y.S. Ginsenoside Rg5 Ameliorates Cisplatin-Induced Nephrotoxicity in Mice through Inhibition of Inflammation, Oxidative Stress, and Apoptosis. Nutrients 2016, 8, 566. [Google Scholar] [CrossRef]
  83. Yang, Q.; Qian, L.; Zhang, S. Ginsenoside Rh1 Alleviates HK-2 Apoptosis by Inhibiting ROS and the JNK/p53 Pathways. Evid. Based Complement. Alternat Med. 2020, 2020, 3401067. [Google Scholar] [CrossRef] [PubMed]
  84. Huynh, D.T.N.; Jin, Y.; Myung, C.S.; Heo, K.S. Ginsenoside Rh1 Induces MCF-7 Cell Apoptosis and Autophagic Cell Death through ROS-Mediated Akt Signaling. Cancers 2021, 13, 1892. [Google Scholar] [CrossRef] [PubMed]
  85. Lyu, X.; Xu, X.; Song, A.; Guo, J.; Zhang, Y.; Zhang, Y. Ginsenoside Rh1 inhibits colorectal cancer cell migration and invasion in vitro and tumor growth in vivo. Oncol. Lett. 2019, 18, 4160–4166. [Google Scholar] [CrossRef] [PubMed]
  86. Xiaodan, S.; Ying, C. Role of ginsenoside Rh2 in tumor therapy and tumor microenvironment immunomodulation. Biomed. Pharmacother. 2022, 156, 113912. [Google Scholar] [CrossRef] [PubMed]
  87. Qi, Z.; Yan, Z.; Wang, Y.; Ji, N.; Yang, X.; Zhang, A.; Li, M.; Xu, F.; Zhang, J. Ginsenoside Rh2 Inhibits NLRP3 Inflammasome Activation and Improves Exosomes to Alleviate Hypoxia-Induced Myocardial Injury. Front. Immunol. 2022, 13, 883946. [Google Scholar] [CrossRef] [PubMed]
  88. Thandra, K.C.; Barsouk, A.; Saginala, K.; Aluru, J.S.; Barsouk, A. Epidemiology of lung cancer. Contemp. Oncol. 2021, 25, 45–52. [Google Scholar] [CrossRef]
  89. Xue, X.; Liu, Y.; Qu, L.; Fan, C.; Ma, X.; Ouyang, P.; Fan, D. Ginsenoside Rh3 Inhibits Lung Cancer Metastasis by Targeting Extracellular Signal-Regulated Kinase: A Network Pharmacology Study. Pharmaceuticals 2022, 15, 758. [Google Scholar] [CrossRef]
  90. Teng, S.; Lei, X.; Zhang, X.; Shen, D.; Liu, Q.; Sun, Y.; Wang, Y.; Cong, Z. Transcriptome Analysis of the Anti-Proliferative Effects of Ginsenoside Rh3 on HCT116 Colorectal Cancer Cells. Molecules 2022, 27, 5002. [Google Scholar] [CrossRef] [PubMed]
  91. Cao, L.; Gao, Y.; Zhu, J.; Zhang, J.; Dong, M.; Mao, Y. Protective action of the ginsenoside Rh3 in a rat myocardial ischemia-reperfusion injury model by inhibition of apoptosis induced via p38 mitogen-activated protein kinase/caspase-3 signaling. J. Int. Med. Res. 2020, 48, 300060520969090. [Google Scholar] [CrossRef]
  92. To, K.I.; Zhu, Z.X.; Wang, Y.N.; Li, G.A.; Sun, Y.M.; Li, Y.; Jin, Y.H. Integrative network pharmacology and experimental verification to reveal the anti-inflammatory mechanism of ginsenoside Rh4. Front. Pharmacol. 2022, 13, 953871. [Google Scholar] [CrossRef] [PubMed]
  93. Baek, S.H.; Shin, B.K.; Kim, N.J.; Chang, S.Y.; Park, J.H. Protective effect of ginsenosides Rk3 and Rh4 on cisplatin-induced acute kidney injury in vitro and in vivo. J. Ginseng Res. 2017, 41, 233–239. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, Y.; Ma, P.; Duan, Z.; Liu, Y.; Mi, Y.; Fan, D. Ginsenoside Rh4 Suppressed Metastasis of Lung Adenocarcinoma via Inhibiting JAK2/STAT3 Signaling. Int. J. Mol. Sci. 2022, 23, 2018. [Google Scholar] [CrossRef] [PubMed]
  95. Wu, Y.; Pi, D.; Chen, Y.; Zuo, Q.; Zhou, S.; Ouyang, M. Ginsenoside Rh4 Inhibits Colorectal Cancer Cell Proliferation by Inducing Ferroptosis via Autophagy Activation. Evid. Based Complement. Alternat Med. 2022, 2022, 6177553. [Google Scholar] [CrossRef]
  96. Dong, F.; Qu, L.; Duan, Z.; He, Y.; Ma, X.; Fan, D. Ginsenoside Rh4 inhibits breast cancer growth through targeting histone deacetylase 2 to regulate immune microenvironment and apoptosis. Bioorg. Chem. 2023, 135, 106537. [Google Scholar] [CrossRef]
  97. Zhu, A.; Duan, Z.; Chen, Y.; Zhu, C.; Fan, D. Ginsenoside Rh4 delays skeletal muscle aging through SIRT1 pathway. Phytomedicine 2023, 118, 154906. [Google Scholar] [CrossRef]
  98. Chen, X.; Liu, W.; Liu, B. Ginsenoside Rh7 Suppresses Proliferation, Migration and Invasion of NSCLC Cells through Targeting ILF3-AS1 Mediated miR-212/SMAD1 Axis. Front. Oncol. 2021, 11, 656132. [Google Scholar] [CrossRef] [PubMed]
  99. Ryoo, N.; Rahman, M.A.; Hwang, H.; Ko, S.K.; Nah, S.Y.; Kim, H.C.; Rhim, H. Ginsenoside Rk1 is a novel inhibitor of NMDA receptors in cultured rat hippocampal neurons. J. Ginseng Res. 2020, 44, 490–495. [Google Scholar] [CrossRef]
  100. Xiong, J.; Yang, J.; Yan, K.; Guo, J. Ginsenoside Rk1 protects human melanocytes from H2O2-induced oxidative injury via regulation of the PI3K/AKT/Nrf2/HO-1 pathway. Mol. Med. Rep. 2021, 24, 1–9. [Google Scholar] [CrossRef]
  101. Hu, J.N.; Xu, X.Y.; Jiang, S.; Liu, Y.; Liu, Z.; Wang, Y.P.; Gong, X.J.; Li, K.K.; Ren, S.; Li, W. Protective effect of ginsenoside Rk1, a major rare saponin from black ginseng, on cisplatin-induced nephrotoxicity in HEK-293 cells. Kaohsiung J. Med. Sci. 2020, 36, 732–740. [Google Scholar] [CrossRef]
  102. An, X.; Fu, R.; Ma, P.; Ma, X.; Fan, D. Ginsenoside Rk1 inhibits cell proliferation and promotes apoptosis in lung squamous cell carcinoma by calcium signaling pathway. RSC Adv. 2019, 9, 25107–25118. [Google Scholar] [CrossRef]
  103. Oh, J.M.; Lee, J.; Im, W.T.; Chun, S. Ginsenoside Rk1 Induces Apoptosis in Neuroblastoma Cells through Loss of Mitochondrial Membrane Potential and Activation of Caspases. Int. J. Mol. Sci. 2019, 20, 1213. [Google Scholar] [CrossRef]
  104. She, L.; Xiong, L.; Li, L.; Zhang, J.; Sun, J.; Wu, H.; Ren, J.; Wang, W.; Zhao, X.; Liang, G. Ginsenoside Rk3 ameliorates Abeta-induced neurotoxicity in APP/PS1 model mice via AMPK signaling pathway. Biomed. Pharmacother. 2023, 158, 114192. [Google Scholar] [CrossRef]
  105. Ahmad, S.S.; Khalid, M.; Kamal, M.A.; Younis, K. Study of Nutraceuticals and Phytochemicals for the Management of Alzheimer’s Disease: A Review. Curr. Neuropharmacol. 2021, 19, 1884–1895. [Google Scholar] [CrossRef] [PubMed]
  106. Ahmad, S.S.; Kamal, M.A. Current Updates on the Regulation of Beta-Secretase Movement as a Potential Restorative Focus for Management of Alzheimer’s Disease. Protein Pept. Lett. 2019, 26, 579–587. [Google Scholar] [CrossRef] [PubMed]
  107. Corona, J.C. Natural Compounds for the Management of Parkinson’s Disease and Attention-Deficit/Hyperactivity Disorder. Biomed. Res. Int. 2018, 2018, 4067597. [Google Scholar] [CrossRef] [PubMed]
  108. Zhong, Z.; He, X.; Ge, J.; Zhu, J.; Yao, C.; Cai, H.; Ye, X.Y.; Xie, T.; Bai, R. Discovery of small-molecule compounds and natural products against Parkinson’s disease: Pathological mechanism and structural modification. Eur. J. Med. Chem. 2022, 237, 114378. [Google Scholar] [CrossRef] [PubMed]
  109. Ahmad, S.S.; Khatoon, A.; Khan, M.S.; Khalid, M.; Alharbi, A.M.; Siddiqui, M.H. Evaluation of vincamine against acetylcholinesterase enzyme. Cell. Mol. Biol. 2022, 68, 14–21. [Google Scholar] [CrossRef] [PubMed]
  110. Abitbol, A.; Mallard, B.; Tiralongo, E.; Tiralongo, J. Mushroom Natural Products in Neurodegenerative Disease Drug Discovery. Cells 2022, 11, 3938. [Google Scholar] [CrossRef]
  111. Liu, H.; Zhao, J.; Fu, R.; Zhu, C.; Fan, D. The ginsenoside Rk3 exerts anti-esophageal cancer activity in vitro and in vivo by mediating apoptosis and autophagy through regulation of the PI3K/Akt/mTOR pathway. PLoS ONE 2019, 14, e0216759. [Google Scholar] [CrossRef]
  112. Han, J.; Xia, J.; Zhang, L.; Cai, E.; Zhao, Y.; Fei, X.; Jia, X.; Yang, H.; Liu, S. Studies of the effects and mechanisms of ginsenoside Re and Rk(3) on myelosuppression induced by cyclophosphamide. J. Ginseng Res. 2019, 43, 618–624. [Google Scholar] [CrossRef] [PubMed]
  113. Ma, S.; Huang, Q.; Hu, Q.; Gao, R.; Lan, J.; Yu, X.; Zhao, Y.; Shen, F.; Mi, A.; Wang, B. Ginsenoside Rk3 Inhibits the Extramedullary Infiltration of Acute Monocytic Leukemia Cell via miR-3677-5p/CXCL12 Axis. Evid. Based Complement. Alternat Med. 2022, 2022, 3065464. [Google Scholar] [CrossRef]
  114. Zheng, S.W.; Xiao, S.Y.; Wang, J.; Hou, W.; Wang, Y.P. Inhibitory Effects of Ginsenoside Ro on the Growth of B16F10 Melanoma via Its Metabolites. Molecules 2019, 24, 2985. [Google Scholar] [CrossRef] [PubMed]
  115. Xu, H.L.; Chen, G.H.; Wu, Y.T.; Xie, L.P.; Tan, Z.B.; Liu, B.; Fan, H.J.; Chen, H.M.; Huang, G.Q.; Liu, M.; et al. Corrigendum to “Ginsenoside Ro, an oleanolic saponin of Panax ginseng, exerts anti-inflammatory effect by direct inhibiting toll like receptor 4 signaling pathway” [J Ginseng Res 46 (2022) 156–166]. J. Ginseng Res. 2022, 46, 315–319. [Google Scholar] [CrossRef] [PubMed]
  116. Kim, S.; Oh, M.H.; Kim, B.S.; Kim, W.I.; Cho, H.S.; Park, B.Y.; Park, C.; Shin, G.W.; Kwon, J. Upregulation of heme oxygenase-1 by ginsenoside Ro attenuates lipopolysaccharide-induced inflammation in macrophage cells. J. Ginseng Res. 2015, 39, 365–370. [Google Scholar] [CrossRef]
  117. Jiang, L.S.; Li, W.; Zhuang, T.X.; Yu, J.J.; Sun, S.; Ju, Z.C.; Wang, Z.T.; Ding, L.L.; Yang, L. Ginsenoside Ro Ameliorates High-Fat Diet-Induced Obesity and Insulin Resistance in Mice via Activation of the G Protein-Coupled Bile Acid Receptor 5 Pathway. J. Pharmacol. Exp. Ther. 2021, 377, 441–451. [Google Scholar] [CrossRef]
  118. Chiang, H.M.; Chien, Y.C.; Wu, C.H.; Kuo, Y.H.; Wu, W.C.; Pan, Y.Y.; Su, Y.H.; Wen, K.C. Hydroalcoholic extract of Rhodiola rosea L. (Crassulaceae) and its hydrolysate inhibit melanogenesis in B16F0 cells by regulating the CREB/MITF/tyrosinase pathway. Food Chem. Toxicol. 2014, 65, 129–139. [Google Scholar] [CrossRef]
  119. Lee, D.Y.; Lee, J.; Jeong, Y.T.; Byun, G.H.; Kim, J.H. Melanogenesis inhibition activity of floralginsenoside A from Panax ginseng berry. J. Ginseng Res. 2017, 41, 602–607. [Google Scholar] [CrossRef]
  120. Kim, S.; Jung, S.H.; Cho, C.W. Physicochemical studies of a newly synthesized molecule, 6-methyl-3-phenethyl-3,4-dihydro-1H-quinazoline-2-thione (JSH18) for topical formulations. Arch. Pharm. Res. 2008, 31, 1363–1368. [Google Scholar] [CrossRef]
  121. Zhang, S.; Chen, C.; Lu, W.; Wei, L. Phytochemistry, pharmacology, and clinical use of Panax notoginseng flowers buds. Phytother. Res. 2018, 32, 2155–2163. [Google Scholar] [CrossRef]
  122. Ahmad, S.S.; Ahmad, K.; Lee, E.J.; Shaikh, S.; Choi, I. Computational Identification of Dithymoquinone as a Potential Inhibitor of Myostatin and Regulator of Muscle Mass. Molecules 2021, 26, 5407. [Google Scholar] [CrossRef] [PubMed]
  123. Shaikh, S.; Ali, S.; Lim, J.H.; Chun, H.J.; Ahmad, K.; Ahmad, S.S.; Hwang, Y.C.; Han, K.S.; Kim, N.R.; Lee, E.J.; et al. Dipeptidyl peptidase-4 inhibitory potentials of Glycyrrhiza uralensis and its bioactive compounds licochalcone A and licochalcone B: An in silico and in vitro study. Front. Mol. Biosci. 2022, 9, 1024764. [Google Scholar] [CrossRef] [PubMed]
  124. Lee, E.J.; Shaikh, S.; Baig, M.H.; Park, S.Y.; Lim, J.H.; Ahmad, S.S.; Ali, S.; Ahmad, K.; Choi, I. MIF1 and MIF2 Myostatin Peptide Inhibitors as Potent Muscle Mass Regulators. Int. J. Mol. Sci. 2022, 23, 4222. [Google Scholar] [CrossRef] [PubMed]
Table 1. List of several known compounds of ginseng along with their molecular formula and weight. The compounds known in the different parts of the ginseng plant, such as root and flower bud, having higher molecular weight as compared to other parts of the ginseng plant.
Table 1. List of several known compounds of ginseng along with their molecular formula and weight. The compounds known in the different parts of the ginseng plant, such as root and flower bud, having higher molecular weight as compared to other parts of the ginseng plant.
Part of GinsengCompounds NamePubChem IDMolecular FormulaMolecular Weight (g/mol)
HydrolysisProtopanaxadiol (PPD)9920281C30H52O3460.7
Protopanaxatriol (PPT)9847853C30H52O4476.7
Panaxadiol73498C30H52O3460.7
Panaxatriol73599C30H52O4476.7
LeavesGinsenoside F19809542C36H62O9638.9
Ginsenoside F29918692C42H72O13785.0
Ginsenoside F346887678C41H70O13771.0
Ginsenoside F4102004835C42H70O12767.0
Ginsenoside Ki102294899C37H64O10668.9
Ginsenoside Km102294900C37H64O10668.9
Ginsenoside Rh6131752646C36H62O11670.9
Ginsenoside Rh7101096472C36H60O9636.9
Ginsenoside Rh885245726C36H60O9636.9
RootsGinsenoside Ra1100941542C58H98O261211.4
Ginsenoside Ra2100941543C58H98O261211.4
Ginsenoside Ra373157064C59H100O271241.4
Ginsenoside Rb19898279C54H92O231109.3
Malonylginsenoside Rb1118987129C57H94O261195.3
Ginsenoside Rb26917976C53H90O221079.3
Ginsenoside Rb312912363C53H90O221079.3
Ginsenoside Rc12855889C53H90O221079.3
Ginsenoside Rd11679800C48H82O18947.2
Ginsenoside Rf441922C42H72O14801.0
20-Glucoginsenoside Rf3052077C48H82O19963.2
Ginsenoside Rg1441923C42H72O14801.0
Ginsenoside Rg221599924C42H72O13785.0
Ginsenoside Ro11815492C48H76O19957.1
Ginsenoside Rs185044013C55H92O231121.3
Ginsenoside Rs2162343294C55H92O231121.3
Steamed rootsGinsenoside Rg39918693C42H72O13785.0
Ginsenoside Rg511550001C42H70O12767.0
Ginsenoside Rg691895489C42H70O12767.0
Ginsenoside Rh112855920C36H62O9638.9
Ginsenoside Rh2119307C36H62O8622.9
Ginsenoside Rh320839223C36H60O7604.9
Ginsenoside Rh421599928C36H60O8620.9
Ginsenoside Rh510699455C37H64O9652.9
Ginsenoside Rk111499198C42H70O12767.0
Ginsenoside Rk290472238C36H60O7604.9
Ginsenoside Rk375412555C36H60O8620.9
Ginsenoside Rs3100937823C44H74O14827.0
Ginsenoside Rs5102021585C44H72O13809.0
Flower budsFloralginsenoside A16655581C42H72O16833.0
Floralginsenoside B101423532C50H84O211021.2
Floralginsenoside C16655212C42H72O15817.0
Floralginsenoside D16655213C42H72O15817.0
Floralginsenoside E101423533C41H70O15803.0
Floralginsenoside F101423534C48H82O20979.2
Floralginsenoside G101423535C48H82O21995.2
Floralginsenoside H101423536C53H90O251127.3
Floralginsenoside I16655580C42H72O16833.0
Floralginsenoside J101423537C41H70O15803.0
Floralginsenoside K101423538C50H84O211021.2
Floralginsenoside Lb102512867C48H82O19963.2
Floralginsenoside M101423540C48H82O19963.2
Floralginsenoside N101423541C53H90O221079.3
Floralginsenoside O101423542C53H90O221079.3
Floralginsenoside P101423543C53H90O231095.3
Floralginsenoside Ta46224641C36H60O10652.9
Floralginsenoside Tb46224642C35H62O11658.9
Floralginsenoside Tc46224643C53H90O251127.3
Floralginsenoside Td46224646C53H90O251127.3
Ginsenoside I102050355C48H82O20979.2
Ginsenoside II101717751C48H82O19963.2
Fruits25-Hydroxyprotopanaxadiol158501C30H54O4478.7
SeedsPanaxadione25233029C30H48O5488.7
Table 2. Therapeutic application of different ginseng compounds in disease management.
Table 2. Therapeutic application of different ginseng compounds in disease management.
Compound NameFunctionModel/Object/ExperimentsReference
Protopanaxadiolrecovery from endometriosismice[29]
Ginsenoside F2alcoholic liver damage improvementC57BL/6J WT or IL-10 knockout mice[37]
Ginsenoside F1repair the vascular defects caused by axitinib in zebrafishin vivo tests in zebrafish[33]
reduce Aβ-induced cytotoxicityneuroblastoma neuro-2a (mouse) and neuroblastoma SH-SY5Y (human)[34]
Ginsenoside Rh7anticancerous propertiesA549 and H1299 cell line[98]
Ginsenoside Rb1decrease adipose tissue and leptin levelsKK-Ay DM mice[49]
reduce hepatic fat formationobese diabetic db/db mice[50]
reduce body weight gainHFD-induced obese mice[51]
increase GLUT4 translocationC2C12 and 3T3-L1 cells[53]
Ginsenoside Rb2improve cell viabilityHT22 murine hippocampal neuronal cells[55]
inhibit the growth of colorectal cancer cellsHT29 and SW620 cell lines[56]
Ginsenoside Rcenhance bone developmentovariectomy-induced osteoporosis mice[64]
reduce the proliferation and viability process3T3L1[66]
Ginsenoside Rdenhance hypertrophy aged mice[67]
Ginsenoside Rg2encourage pMSC proliferationMTT assay[76]
Ginsenoside Roinhibit tumor growthB16F10 tumor-bearing mice[114]
Ginsenoside Rg3decrease ROS buildupmice[78]
Ginsenoside Rh2improve the oxygen–glucose deprivation environment of cardiomyocytesregulate the HMGB1/NF-κB signaling[87]
Ginsenoside Rh1anticancer effect on breast cancer cellsinhibition of the ROS-mediated PI3K/Akt pathway[84]
Ginsenoside Rh3inhibit proliferationA549 and PC9 cells[89]
Ginsenoside Rk3improve neuronal apoptosisPC12 and primary neuronal cells[104]
Floralginsenoside Amelanin inhibitory activityC57BL/6 mouse melanocyte cell line[119]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ahmad, S.S.; Ahmad, K.; Hwang, Y.C.; Lee, E.J.; Choi, I. Therapeutic Applications of Ginseng Natural Compounds for Health Management. Int. J. Mol. Sci. 2023, 24, 17290. https://doi.org/10.3390/ijms242417290

AMA Style

Ahmad SS, Ahmad K, Hwang YC, Lee EJ, Choi I. Therapeutic Applications of Ginseng Natural Compounds for Health Management. International Journal of Molecular Sciences. 2023; 24(24):17290. https://doi.org/10.3390/ijms242417290

Chicago/Turabian Style

Ahmad, Syed Sayeed, Khurshid Ahmad, Ye Chan Hwang, Eun Ju Lee, and Inho Choi. 2023. "Therapeutic Applications of Ginseng Natural Compounds for Health Management" International Journal of Molecular Sciences 24, no. 24: 17290. https://doi.org/10.3390/ijms242417290

APA Style

Ahmad, S. S., Ahmad, K., Hwang, Y. C., Lee, E. J., & Choi, I. (2023). Therapeutic Applications of Ginseng Natural Compounds for Health Management. International Journal of Molecular Sciences, 24(24), 17290. https://doi.org/10.3390/ijms242417290

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