Open Access

Pharmacological activities of ginsenoside Rg5 (Review)

  • Authors:
    • Ming-Yang Liu
    • Fei Liu
    • Yan-Li Gao
    • Jia-Ning Yin
    • Wei-Qun Yan
    • Jian-Guo Liu
    • Hai-Jun Li
  • View Affiliations

  • Published online on: June 6, 2021     https://doi.org/10.3892/etm.2021.10272
  • Article Number: 840
  • Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ginseng, a perennial plant belonging to genus Panax, has been widely used in traditional herbal medicine in East Asia and North America. Ginsenosides are the most important pharmacological component of ginseng. Variabilities in attached positions, inner and outer residues and types of sugar moieties may be associated with the specific pharmacological activities of each ginsenoside. Ginsenoside Rg5 (Rg5) is a minor ginsenoside synthesized during ginseng steaming treatment that exhibits superior pharmaceutical activity compared with major ginsenosides. With high safety and various biological functions, Rg5 may act as a potential therapeutic candidate for diverse diseases. To date, there have been no systematic studies on the activity of Rg5. Therefore, in this review, all available literature was reviewed and discussed to facilitate further research on Rg5.

1. Introduction

Ginseng, a perennial plant belonging to genus Panax, has been widely used in traditional herbal medicine in east Asia and North America for millennia to reinforce immunity, provide nutrition and reduce fatigue (1,2). Ginsenosides are unique triterpenoid saponins predominantly extracted from Panax ginseng C.A. Meyer that act as the main bioactive constituents of ginseng (3,4). To date, >100 ginsenosides have been extracted from the roots, leaves, stems, fruits and flower heads of ginseng (3). All ginsenosides share a common four-ring hydrophobic structure, but differ in the number and types of glycosyl (5). Ginsenosides are classified into 20(S)-protopanaxadiol, 20(S)-protopanaxatriol saponins and oleanoic acid ginsenosides (6,7). The variability of attached positions, inner and outer residues, and types of sugar moieties may be associated with the specific pharmacological activities of different ginsenosides (4). Glycosylated major ginsenosides, such as Rb1, Rb2, Rc, Rd, Re and Rg1, constitute >80% of the total ginsenosides in various parts of ginseng (8). Deglycosylated minor ginsenosides, which have fewer sugar moieties attached on aglycon, are absent or present in smaller amounts in wild ginseng (9,10).

Ginsenoside Rg5 (Rg5) is a minor ginsenoside synthesized during ginseng steaming treatment; the structural formula is displayed in Fig. 1 (11). It is obtained by the deglycosylation of ginsenoside Rb1 and dehydration of carbon at position 20 of ginsenoside Rg3, and exhibits superior pharmaceutical effect compared with major ginsenosides (12,13). In previous studies, Rg5 was found to exert multiple pharmacological effects, such as antitumor, anti-inflammatory, antidiabetic, anti-osteoarthritis (OA), neuroprotective and cardioprotective properties (14-20). With high safety and various biological functions, Rg5 has the potential to act as a potential therapeutic candidate for diverse diseases. The present article reviewed the Rg5 literature and summarized its pharmacological activities.

2. Pharmacological activities of Rg5

Anticancer effects

Cancer is a group of life-threatening diseases that are characterized by abnormal proliferation of cells with potential to invade and spread to surrounding and distant tissues. Conventional therapies for cancer include surgical resection, radiotherapy and chemotherapy (21,22). Chemotherapy serves an important role in the treatment of malignant tumors (23). However, the severe side effects of traditional chemotherapy, such as myelosuppression and immune suppression, hinder the therapeutic effects (24). The use of ginsenosides as alternative antitumor agents has gained increasing attention (25,26). Several studies have reported the antitumor effects on human gastric and breast cancer of Rg5, which are mainly associated with promoting apoptosis, autophagy and cell cycle arrest (15-18).

Breast cancer is a major health risk for the adult female population. The mechanism underlying the effects of Rg5 on breast cancer has been investigated in vivo and in vitro. Kim and Kim (15) demonstrated that Rg5 promoted breast cancer cell apoptosis via downregulation of the Bax/Bcl-2 pathway. It was also demonstrated that Rg5 inhibited breast cancer cell proliferation by arresting the cell cycle at the G0/G1 phase by promoting the expression of p53, p21WAF1/CIP1 and p15INK4B and inhibiting the expression levels of cyclin D1, cyclin E2 and CDK4(15). Moreover, it was discovered that Rg5 exhibited improved pro-apoptotic effects on human breast cancer cell lines compared with ginsenoside Rg3(15). Zou and Liu (16) reported that Rg5 exhibited antiproliferative effects against breast cancer cells by activating the AMPK pathway. More recently, Liu et al (14) discovered that Rg5 induced breast cancer cell apoptosis and autophagy by inhibiting the PI3K/Akt/mTOR signaling pathway. In addition, Kim et al (13) demonstrated that Rg5 inhibited the growth of tumors in breast cancer mouse models by promoting tumor cell autophagy and apoptosis without damaging the normal functions of major organs and immune cells, which indicated that Rg5 exerts effects against breast cancer in vivo (17).

The effects and associated mechanisms underlying Rg5 in the treatment of digestive system cancer have also been reported. Liu and Fan (18) investigated the anticancer activity of ginsenoside in human gastric cancer cell lines and suggested that Rg5 inhibited cell proliferation by inducing G2/M phase arrest, apoptosis and autophagy by activating reactive oxygen species (ROS)-mediated MAPK pathways. Moreover, in a human xenograft nude mouse model, Rg5 displayed significant effects against gastric cancer with few side effects (18). Zhang et al (27) revealed that Rg5 suppressed proliferation and promoted the apoptosis of human esophageal cancer cells, which was associated with inhibition of the PI3K/Akt signaling pathway. Wang et al (28) demonstrated that Rg5 bound to annexin A2, and inhibited the interaction between annexin A2 and NF-κB p50 subunit, which resulted in the promotion of caspase activation and NF-κB activity in human hepatoma HepG2 cells. Lee et al (29) reported that Rg5 arrested the cell cycle of human hepatoma SK-HEP-1 cells at the G1/S transition phase through cyclin E-dependent protein kinase activity by increasing the expression of p21Cip/WAF1 and reducing cyclin E.

Additionally, Rg5 promotes apoptosis in retinoblastoma cells by inhibiting the Akt signaling pathway and thereby downregulating Bcl-2 expression (30). Rg5 also promotes human cervical cancer cell apoptosis in concentration- and time-dependent manners by inducing DNA double-strand breaks and fragments (11). Rg5 has also been demonstrated to effectively overcome ATP binding cassette subfamily B member 1 (ABCB1)-mediated drug resistance by inhibiting ABCB1 transporter and blocking the activation of Akt/nuclear-related factor 2 pathways without affecting the expression of ABCB1 transporter (31). Therefore, Rg5 may serve as a chemosensitizer for reversing multidrug resistance.

With the development of nanomedicine, the antitumor effects of Rg5 through biocompatible nanoscale drug delivery systems has also been investigated (32). Dong et al (32) used folic acid (FA)-modified bovine serum albumin (BSA) nanoparticles (FA-Rg5-BSA NPs) as carriers to entrap Rg5. It was discovered that the FA-Rg5-BSA NPs exhibited superior anticancer activity compared with Rg5 in MCF-7 cells with low cytotoxicity to L929 cells. The FA-Rg5-BSA NPs facilitated cellular uptake and induced apoptosis in MCF-7 cells. Furthermore, an in vivo antitumor study demonstrated that FA-Rg5-BSA NPs were more effective in reducing tumor growth than Rg5 and Rg5-BSA NPs in an MCF-7 xenograft mouse model. This in vivo real-time bioimaging study demonstrated that the FA-Rg5-BSA NPs had an advanced ability to accumulate in tumors. These results indicated that FA-Rg5-BSA NPs has the potential to serve as an attractive therapeutic strategy for the management of cancer (32).

Anti-inflammatory effects

Inflammation is defined as a defensive mechanism characterized by the release of proinflammatory cytokines and the transmigration of inflammatory cells to protect human bodies from harmful stimuli (19,20). Excessive or abnormal inflammatory responses have the potential to lead to various diseases, such as organ failure, central nerve system injury, tissue damage and even death (33). Thus, regulating the expression of inflammatory mediators should be beneficial for the treatment of inflammation-related diseases (13,34). A number of studies have reported that Rg5 exerted anti-inflammatory effects in inflammatory responses by inhibiting the production of proinflammatory cytokines, such as TNF-α and IL-1β by interfering NF-κB or the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome pathways (35-40).

It has been demonstrated that Rg5 exhibited protective effects on major organs through its anti-inflammatory mechanisms. Kim et al (35) suggested that Rg5 ameliorates lung inflammation in mice by blocking the binding of lipopolysaccharide (LPS) to Toll-like receptor (TLR)-4 on macrophages, which are associated with inhibition of NF-κB activation. Park et al (36) demonstrated that Rg5 exerted protective effects against cisplatin-induced renal damage by attenuating JNK/p53/caspase-3 cascade-mediated inflammation. Li et al (37) demonstrated that Rg5 attenuated renal dysfunction by reducing the expression of inflammatory mediators, including NF-κB, p65 and cyclooxygenase-2 (COX-2). Similarly, Lee (38) reported that Rg5 also decreased the expression of NF-κB, inducible nitric oxide (NO) synthase (iNOS) and COX-2 in HepG2 cells treated with TNF-α and thereby acted as a potential anti-inflammatory agent against hepatitis. The anti-inflammatory activity of Rg5 was increased when compared with ginsenoside Rb1, Rd and Rg3 and this increased bioactivity of Rg5 was hypothesized to be due to the higher lipophilicity compared with Rb1, Rd and Rg3(38). Rg5 also ameliorated acetaminophen (APAP)-induced liver injury by suppressing APAP-induced expression of the inflammatory cytokines, TNF-α and IL-1β (39). Moreover, Rg5 demonstrated protective effects in high-fat diet/streptozotocin-induced diabetic nephropathy mice and improved renal injury by attenuating oxidative stress and inflammatory states by suppressing ROS-mediated activation of NLRP3 inflammasome, p38 MAPK and NF-κB signaling pathways in the kidneys of diabetic nephropathy mice (40).

Sepsis is a systemic inflammatory response syndrome caused by the body's response to infection (41). High mobility group box 1 (HMGB1) is regarded as a crucial mediator of sepsis (42). The suppression of HMGB1-induced inflammatory reactions and maintenance of endothelial integrity have served as promising therapeutic strategies for the treatment of sepsis (43). Kim et al (44) demonstrated that Rg5 suppressed the release of HMGB1 in LPS-activated human umbilical vein endothelial cells (HUVECs). Moreover, Rg5 inhibited the adhesion and migration of leukocytes toward HUVECs. The aforementioned study indicated that Rg5 may be a potential therapeutic option for the treatment of severe vascular inflammatory diseases, such as sepsis and septic shock.

Rg5 has also demonstrated anti-inflammatory properties in dermal diseases (45,46). Shin et al (45) demonstrated the inhibitory effects of Rg5 and its metabolite ginsenoside Rh3 in oxazolone-induced mouse ear contact dermatitis by inhibiting the expression of COX-2, TNF-α and IL-1β produced by macrophage cells and IFN-γ produced by Thl cells. Ahn et al (46) discovered that Rg5 had anti-inflammatory effects in two atopic dermatitis-related cell lines. LPS-induced production of NO and ROS was downregulated by Rg5 in RAW264.7 cells, indicating that Rg5 has the ability to improve chronic inflammatory skin disease by blocking the NF-κB/p38/MAPK/STAT1 signaling pathways.

Anti-inflammatory effects of Rg5 in the central neural system have also been demonstrated. Chu et al (47) reported that Rg5 significantly suppressed the expression of pro-inflammation-related cytokines, including IL-1β, TNF-α, COX-2 and iNOS, and thereby attenuated neuroinflammatory responses in STZ-induced memory-impaired rats. Rg5 was also revealed to relieve cerebral ischemic injury by decreasing NF-κB transcriptional activity and the expression of proinflammatory cytokines, such as IL-1β, TNF-α and IL-6, by activating TLR4/MyD88 and sirtuin 1 signaling pathways, contributing to reductions in cerebral ischemic injury (48). In addition, Lee et al (49) demonstrated that Rg5 suppressed neuroinflammation induced by LPS in BV2 microglial cells by inhibiting the MAPK and PI3K/Akt pathways.

Neuroprotective effects

Neurodegenerative diseases have become another category of health-threatening diseases (50). Rg5 exerts beneficial effects on nervous system diseases, such as Alzheimer's disease (AD) and Huntington's disease (HD) (51-53). AD is a multifactorial neurodegenerative disease featuring extracellular β-amyloid (Aβ) plaques and intracellular neurofibrillary tangles in the brain (51). Inhibition of cAMP response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF) has the potential to lead to memory deficits in patients with AD (52,53). Kim et al (54) revealed that Rg5 significantly reversed memory deficits induced by acetylcholinesterase using the passive avoidance, Y-maze and Morris water maze tasks in mice. The research revealed that treatment with Rg5 ameliorated the reduction of BDNF expression and CREB phosphorylation induced by scopolamine (54). Chu et al (47) demonstrated that Rg5 improves cognitive dysfunction in streptozotocin-induced AD rats by modulating the cholinergic system, decreasing Aβ deposition and promoting the expression levels of neurotrophic factors BDNF and insulin-like growth factor 1 (IGF-1). Moreover, Rg5 has significant ameliorative effects on STZ-induced neuroinflammatory responses (47). Choi et al (55) revealed that Rg5 suppressed thermal stress-induced cell cycle arrest at G1/S phase by activating p21 and poly(ADP-ribose) polymerase cleavage. CREB and BDNF were also increased by Rg5 in thermal stress-exposed HT22 cells (55). HD is an autosomal-dominant neurogenic disorder that leads to progressive nerve cell damage in the brain. Wu et al (56) demonstrated Rg5 attenuates neuronal apoptosis by inhibiting glutamate-induced increases of Ca2+ concentrations in cultured medium spiny neurons, which indicated Rg5 has the potential to be useful for HD therapy. Wu et al (56) further discovered that the inhibitory activity of Rg5 on glutamate-induced Ca2+ responses was similar to ginsenoside Rc and far greater than ginsenoside Re.

In addition, an in vivo study revealed that Rg5 may regulate nerve transmission by affecting neurotransmitter and neuroregulatory receptors (57). Glutamate (Glu) is known as a major excitatory neurotransmitter, whereas γ-aminobutyric acid (GABA) is well-known as a major inhibitory neurotransmitter in the CNS (58,59). Shao et al (57) suggested that Rg5 downregulates the GABA/Glu ratio, and augments the expression of GABAA and GABAB receptors. Serotonin (5-HT) is a neurotransmitter involved in sleep-wake cycle regulation (60). Shao et al (57) demonstrated that 5-hydroxytryptophan, together with a precursor of 5-HT, promotes the sleep effects of Rg5 in mice and Rg5 augments the expression of 5-HT1A. The results indicated that Rg5 exhibited hypotensive and sedative activities by modulating GABA and serotonin signaling in the nervous system, thus ameliorating sleep in mice models (57).

Cardioprotective effects

Studies into the therapeutic effects of Rg5 on cardiovascular diseases have also been reported. Cho et al (61) demonstrated that Rg5 regulates neovascularization and vasorelaxation by activating IGF-1 receptor (IGF-1R). The angiogenic activity of Rg5 is highly associated with a specific increase in IGF-1R phosphorylation and the subsequent activation of multiple angiogenic signals (61). Furthermore, the vasodilative activity of Rg5 is mediated by the endothelial NOS/NO/cGMP pathway (61). These findings offer a mechanistic explanation of the beneficial effects of Rg5 on neovascularization and endothelial function under pathological conditions. Yang et al (62) reported that Rg5 increased cardiomyocyte resistance to ischemic injury by regulating the translocation of two important enzymes, hexokinase-II (HK-II) and dynamin-related protein 1 (Drp1). Drp1 and HK-II exert opposite effects on mitochondrial function in cardiomyocytes by competing for binding to mitochondria (62). Rg5 protects mitochondrial morphological and functional integrity by suppressing Drp1 activation and increasing HK-II binding to cardiomyocyte mitochondria through Akt activation (62). These results provide a rationale for utilizing Rg5 for treating cardiovascular diseases.

Anti-OA and anti-osteoporosis (OP) effects

OA and OP are common bone diseases in middle-aged and elderly populations. OA involves a series of complicated processes characterized by the destruction of chondrocytes and remodeling of subchondral bones, resulting in progressive joint degeneration (63). The regulation of inflammatory cytokine networks by ginsenosides has attracted increased attention for the treatment of OA (64,65). Zhang (66) revealed that Rg5 prevented articular cartilage degradation and inhibited synovium disintegration in OA rat models. The level of OA-related enzyme metalloproteinase-13 decreased to 45% compared with controls; tissue inhibitors of metalloproteinase-1 increased by 67% after treatment with Rg5. The levels of inflammatory mediators, such as IL-1β, TNF-α, NO and iNOS, decreased by 67, 54, 32 and 49%, respectively, after 1 month of treatment with Rg5(66). The expression of bone morphogenetic protein-2 (BMP-2) and TGF-β1 increased to 67 and 52%, respectively, after treatment with Rg5. Therefore, Zhang (66) considered Rg5 useful for OA therapy.

OP systemically decreases bone mass and strength, and is characterized by the disturbance of osteoblast activity (67). Siddiqi et al (68) demonstrated that Rg5/Rk1 stimulates osteoblast cell growth and promotes the expression of osteoblastic markers, such as alkaline phosphatase activity and type I collagen content, BMP-2 and calcium deposition in dose-dependent manners. Moreover, Rg5/Rk1 also stimulates the mRNA expression of Runt-related transcription factor 2 (Runx2) and osteocalcin (68). These results indicate that Rg5/Rk1 has the potential to prevent OP by stimulating osteoblast proliferation and differentiation via the BMP-2/Runx2 signaling pathway.

Antidiabetic and anti-obesity effects

The antidiabetic effect of Rg5 can be attributed to the amelioration of insulin resistance and reduction of glucagon response (67-72). A high level of insulin is required to regulate blood glucose under insulin resistance conditions. During endoplasmic reticulum stress, the Rk1/Rg5 ginsenoside complex was found to improve insulin sensitivity and increase glucose uptake to exert protective effects in 3T3-L1 cells through CHOP-mediated glucose transporter 4 translocation (69). Furthermore, Xiao et al (71) discovered that Rg5 inhibited succinate-associated lipolysis by reducing cellular energy charge and effectively prevented insulin resistance in muscle by reducing lipid deposits. The inhibitory effects of Rg5 in hepatic glucagon response have also been demonstrated (70). Xiao et al (70) revealed that Rg5 decreased succinate accumulation by suppressing hepatic fatty acid oxidation and cAMP accumulation by blocking succinate/hypoxia-inducible factor-1α expression, leading to an attenuated hepatic glucagon response.

Ginsenosides have also been widely reported to have an anti-obesity effect (73-76) and the anti-obesity effect of Rg5 has been reported in vitro. Yesmin Simu et al (77) demonstrated that Rg5/Rk1 inhibited lipid droplet accumulation and decreased triglyceride content in 3T3-L1 adipocyte cells. The expression levels of STAT3, peroxisome proliferator-activated receptor (PPAR)γ, CCAAT/enhancer-binding protein (CEBP)α and adaptor protein complex were also reduced in dose-dependent manners after treatment with Rg5/Rk1. Furthermore, Yesmin Simu et al (77) reported no significant cytotoxicity effects on 3T3-L1 cells up to 100 µg/ml. Their results indicated that Rg5 may have therapeutic potential for treating obesity via the STAT3/PPARγ/CEBPα signaling pathway.

3. Conclusion

The current review summarized the pharmacological effects of Rg5. In general, Rg5 has substantial potential activity for use as a broad-spectrum anticancer and anti-inflammatory drug. Rg5 has been reported to exert several positive effects on the nervous system, which potentiate the clinical applications of Rg5 in the treatment of neurodegenerative diseases. Additional studies have investigated other pharmacological properties, such as cardioprotective, anti-OA, anti-OP, antidiabetic and anti-obesity effects. The biological activities of Rg5 have been widely investigated and the mechanisms underlying the actions of Rg5 based on the existing studies are summarized in Table I and Fig. 2. These optimized therapies should also be evaluated for their efficacies in vivo. It may be possible to develop novel Rg5 analogues with improved efficacy, pharmacokinetics and bioavailability profiles. Evidence of Rg5 efficacy has yet to be demonstrated in humans. There is a significant need to perform larger cohort clinical studies to confirm Rg5 efficacy for improved applications in the clinic. With this considered, further investigations in clinical trials are highly recommended to provide more reliable evidence for the clinical efficacy of Rg5.

Table I

Summary of the pharmacological activities of Rg5.

Table I

Summary of the pharmacological activities of Rg5.

A, Anti-inflammation
First author, yearModelEffects(Refs.)
Zhu et al, 2020DN miceRg5 attenuates oxidative stress and inflammatory states in HFD/STZ-induced DN mice by inactivating p38 MAPK and NF-κB signaling pathways(40)
Kim et al, 2019HUVECsRg5/Rk1 reduces the secretion of HMGB1, and the adhesion and migration of leukocytes toward HUVECs(44)
Kim et al, 2019Male C57BL/6 miceRg5 reduces CLP-induced mortality and pulmonary injury(44)
Yang et al, 2017I/R ratsRg5 reduces TNF-α, IL-6, and IL-1β tissue levels in I/R rats(62)
Wang et al, 2018Male ICR miceRg5 protects against oxidative/nitrative stress injury, inflammation and apoptosis in APAP-induced hepatotoxicity(28)
Li et al, 2016Male ICR miceRg5 attenuates oxidative stress, suppresses inflammation and inhibits apoptosis in cisplatin-treated kidney cells(37)
Ahn et al, 2016HaCaT cellsRg5/Rk1 suppresses NF-κB/p38 MAPK/STAT1 signaling(46)
Ahn et al, 2016RAW264.7 cellsRg5/Rk1 suppresses NF-κB/p38 MAPK/STAT1 signaling(46)
Park et al, 2015LLC-PK1 cellsRg5 ameliorates renal cell damage by inhibiting inflammation and preventing apoptosis(36)
Lee et al, 2013BV2 microglial cellsRg5 exhibits anti-inflammatory effects in LPS-stimulated microglia(49)
Kim et al, 2017Male C57BL/6 miceRg5 ameliorates lung inflammation via downregulation of NF-κB activation by inhibiting binding of LPS to TLR4 on macrophages(13)
Shin et al, 2006Female ICR miceRg5 improves chronic dermatitis or psoriasis in oxazolone-induced ICR mice via downregulation of IL-lβ, TNF-α and IFN-γ production(45)
Chu et al, 2014Male Wistar ratsRg5 induces amelioration of STZ-induced neuroinflammatory responses(47)
B, Neuroprotection
First author, yearModelEffects(Refs.)
Shao et al, 2018Male Kunming mice, male Wistar ratsRg5 exerts sedative and hypnotic effects by affecting GABA and serotonin signaling(57)
Choi et al, 1994HT22 cellsRg5 inhibits thermal stress-induced apoptosis in HT22 cells(55)
Chu et al, 2014Wistar ratsRg5 alleviates cognitive dysfunction in STZ-induced AD rats by regulating cholinergic signaling, attenuating Aβ deposition and increasing neurotrophic factor expression(47)
Kim et al, 2013Male ICR miceRg5/Rh3 protects against memory deficits by inhibiting AChE activity, and increasing BDNF expression and CREB activation(54)
Wu et al, 2009YAC128 miceRg5 protects striatal neurons via inhibition of Ca2+ signaling(56)
C, Cardioprotection
First author, yearModelEffects(Refs.)
Yang et al, 2017Male ICR miceRg5 protects mitochondrial morphological and functional integrity by regulating HK-II and Drp1 translocation via Akt activation(62)
Cho et al, 2015HUVECsRg5 promotes angiogenesis and vasorelaxation by activating signal transduction pathways downstream of IGF-1R(61)
Cho et al, 2015C57BL/6J miceRg5 promotes angiogenesis and vasorelaxation by activating signal transduction pathways downstream of IGF-1R(61)
D, Anti-osteoarthritis/anti-osteoporosis
First author, yearModelEffects(Refs.)
Zhang, 2017Male Wistar ratsRg5 prevents destruction of articular cartilage via inhibition of chondrocyte apoptosis and matrix damage in osteoarthritis rats(66)
Siddiqi et al, 2014MC3T3-E1Rg5/Rk1 promotes the function of MC3T3-E1 cells via BMP-2/Runx2 signaling(68)
E, Antidiabetes/anti-obesity
First author, yearModelEffects(Refs.)
Ponnuraj et al, 20143T3-L1 cellsRg5/Rk1 ameliorates insulin sensitivity in 3T3-L1 cells via CHOP signaling(69)
Xiao et al, 20173T3-L1 cellsRg5 inhibits succinate-associated lipolysis via reducing cellular energy charge, and effectively prevented insulin resistance by reducing lipid deposits(70)
Xiao et al, 2017Male ICR miceRg5 inhibits succinate-associated lipolysis and prevents insulin resistance by reducing lipid deposition(70)
Xiao et al, 2017Male C57BL/6J miceRg5 reduces succinate accumulation and inhibits hepatic cAMP accumulation(70)
Yesmin Simu et al, 20173T3-L1 cellsRg5/Rk1 exhibits anti-adipogenic activity via downregulation of STAT3/PPARγ/CEBPα signaling(77)

[i] Rg5, ginsenoside Rg5; DN, diabetic nephropathy; HFD, high-fat diet; STZ, streptozotocin; HUVEC, human umbilical vein endothelial cell; HMGB1, high mobility group box protein 1; CLP, cecal ligation and puncture; I/R, ischemia/reperfusion; APAP, acetaminophen; LPS, lipopolysaccharide; TLR, Toll-like receptor; GABA, γ-aminobutyric acid; AD, Alzheimer's disease; Aβ, β-amyloid; AChE, acetylcholinesterase; BDNF, brain-derived neurotrophic factor; CREB, cAMP response element-binding protein; IGF-1R, insulin-like growth factor-1 receptor; HK-II, hexokinase-II; Drp1, dynamin-related protein 1; BMP-2, bone morphogenic protein-2; Runx2, Runt-related transcription factor 2; PPARγ, peroxisome proliferator-activated receptor γ; CEBPα, CCAAT/enhancer-binding protein α.γ.

Acknowledgements

Not applicable.

Funding

Funding: This study was supported in part by the National Natural Science Foundation of China (grant nos. 81970529 and 81700018), the Jilin Provincial Natural Science Foundation of China (grant no. 20180101135JC), the Jilin Provincial Finance Foundation (grant no. 2018SCZWSZX-043), Jilin Provincial Health Commission Foundation (grant no. 2018Q020), the Interdisciplinary Chemistry and Medicine Foundation of Jilin University (grant no. JDYYJCHX004), the Norman Bethune Program of Jilin University (grant no. 2019007) and the Foundation for The Excellent Youth Scholars of Jilin University.

Availability of data and materials

Not applicable.

Authors' contributions

MYL and FL wrote the manuscript. YLG and JNY collected the references and produced the figure. HJL designed, interpreted and funded the study and revised the manuscript. WQY and JGL revised the manuscript. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

De Souza LR, Jenkins AL, Sievenpiper JL, Jovanovski E, Rahelić D and Vuksan V: Korean red ginseng (Panax ginseng C.A. Meyer) root fractions: Differential effects on postprandial glycemia in healthy individuals. J Ethnopharmacol. 137:245–250. 2011.PubMed/NCBI View Article : Google Scholar

2 

Zhou QL, Zhu DN, Yang YF, Xu W and Yang XW: Simultaneous quantification of twenty-one ginsenosides and their three aglycones in rat plasma by a developed UFLC-MS/MS assay: Application to a pharmacokinetic study of red ginseng. J Pharm Biomed Anal. 137:1–12. 2017.PubMed/NCBI View Article : Google Scholar

3 

Christensen LP: Ginsenosides chemistry, biosynthesis, analysis, and potential health effects. Adv Food Nutr Res. 55:1–99. 2009.PubMed/NCBI View Article : Google Scholar

4 

Shi W, Wang Y, Li J, Zhang H and Ding L: Investigation of ginsenosides in different parts and ages of Panax ginseng. Food Chem. 102:664–668. 2007.

5 

Chen RJ, Chung TY, Li FY, Lin NH and Tzen JTC: Effect of sugar positions in ginsenosides and their inhibitory potency on Na+/K+-ATPase activity. Acta Pharmacol Sin. 30:61–69. 2009.PubMed/NCBI View Article : Google Scholar

6 

Shin BK, Kwon SW and Park JH: Chemical diversity of ginseng saponins from Panax ginseng. J Ginseng Res. 39:287–298. 2015.PubMed/NCBI View Article : Google Scholar

7 

Shin KC and Oh DK: Classification of glycosidases that hydrolyze the specific positions and types of sugar moieties in ginsenosides. Crit Rev Biotechnol. 36:1036–1049. 2016.PubMed/NCBI View Article : Google Scholar

8 

Jin S, Jeon JH, Lee S, Kang WY, Seong SJ, Yoon YR, Choi MK and Song IS: Detection of 13 ginsenosides (Rb1, Rb2, Rc, Rd, Re, Rf, Rg1, Rg3, Rh2, F1, compound K, 20(S)-protopanaxadiol, and 20(S)-protopanaxatriol) in human plasma and application of the analytical method to human pharmacokinetic studies following two week-repeated administration of red ginseng extract. Molecules. 24(2618)2019.PubMed/NCBI View Article : Google Scholar

9 

Cui CH, Jeon BM, Fu Y, Im WT and Kim SC: High-density immobilization of a ginsenoside-transforming β-glucosidase for enhanced food-grade production of minor ginsenosides. Appl Microbiol Biotechnol. 103:7003–7015. 2019.PubMed/NCBI View Article : Google Scholar

10 

Noh KH and Oh DK: Production of the rare ginsenosides compound K, compound Y, and compound Mc by a thermostable beta-glycosidase from sulfolobus acidocaldarius. Biol Pharm Bull. 32:1830–1835. 2009.PubMed/NCBI View Article : Google Scholar

11 

Liang LD, He T, Du TW, Fan YG, Chen DS and Wang Y: Ginsenoside-Rg5 induces apoptosis and DNA damage in human cervical cancer cells. Mol Med Rep. 11:940–946. 2015.PubMed/NCBI View Article : Google Scholar

12 

Qi LW, Wang CZ and Yuan CS: Ginsenosides from American ginseng: Chemical and pharmacological diversity. Phytochemistry. 72:689–699. 2011.PubMed/NCBI View Article : Google Scholar

13 

Kim JH, Yi YS, Kim MY and Cho JY: Role of ginsenosides, the main active components of Panax ginseng, in inflammatory responses and diseases. J Ginseng Res. 41:435–443. 2017.PubMed/NCBI View Article : Google Scholar

14 

Liu Y and Fan D: The preparation of ginsenoside Rg5, its antitumor activity against breast cancer cells and its targeting of PI3K. Nutrients. 12(246)2020.PubMed/NCBI View Article : Google Scholar

15 

Kim SJ and Kim AK: Anti-breast cancer activity of fine black ginseng (Panax ginseng Meyer) and ginsenoside Rg5. J Ginseng Res. 39:125–134. 2015.PubMed/NCBI View Article : Google Scholar

16 

Zou Y and Liu P: Ginsenoside-Rg5 inhibits proliferation of the breast carcinoma cells through promotion of the proteins involved in AMP kinase pathway. Int J Clin Exp Med. 9:17664–17669. 2016.

17 

Liu Y and Fan D: Ginsenoside Rg5 induces apoptosis and autophagy via the inhibition of the PI3K/Akt pathway against breast cancer in a mouse model. Food Funct. 9:5513–5527. 2018.PubMed/NCBI View Article : Google Scholar

18 

Liu Y and Fan D: Ginsenoside Rg5 induces G2/M phase arrest, apoptosis and autophagy via regulating ROS-mediated MAPK pathways against human gastric cancer. Biochem Pharmacol. 168:285–304. 2019.PubMed/NCBI View Article : Google Scholar

19 

Jin HS, Suh HW, Kim SJ and Jo EK: Mitochondrial control of innate immunity and inflammation. Immune Netw. 17:77–88. 2017.PubMed/NCBI View Article : Google Scholar

20 

Medzhitov R: Origin and physiological roles of inflammation. Nature. 454:428–435. 2008.PubMed/NCBI View Article : Google Scholar

21 

Harrison DJ, Geller DS, Gill JD, Lewis VO and Gorlick R: Current and future therapeutic approaches for osteosarcoma. Expert Rev Anticancer Ther. 18:39–50. 2018.PubMed/NCBI View Article : Google Scholar

22 

Akram M, Iqbal M, Daniyal M and Khan AU: Awareness and current knowledge of breast cancer. Biol Res. 50(33)2017.PubMed/NCBI View Article : Google Scholar

23 

Sun M, Ye Y, Xiao L, Duan X, Zhang Y and Zhang H: Anticancer effects of ginsenoside Rg3 (Review). Int J Mol Med. 39:507–518. 2017.PubMed/NCBI View Article : Google Scholar

24 

Baldo BA and Pham NH: Adverse reactions to targeted and non-targeted chemotherapeutic drugs with emphasis on hypersensitivity responses and the invasive metastatic switch. Cancer Metastasis Rev. 32:723–761. 2013.PubMed/NCBI View Article : Google Scholar

25 

Majeed F, Malik FZ, Ahmed Z, Afreen A, Afzal MN and Khalid N: Ginseng phytochemicals as therapeutics in oncology: Recent perspectives. Biomed Pharmacother. 100:52–63. 2018.PubMed/NCBI View Article : Google Scholar

26 

Nakhjavani M, Hardingham JE, Palethorpe HM, Tomita Y, Smith E, Price TJ and Townsend AR: Ginsenoside Rg3: Potential molecular targets and therapeutic indication in metastatic breast cancer. Medicines (Basel). 6(17)2019.PubMed/NCBI View Article : Google Scholar

27 

Zhang D, Wang A, Feng J, Zhang Q, Liu L and Ren H: Ginsenoside Rg5 induces apoptosis in human esophageal cancer cells through the phosphoinositide-3 kinase/protein kinase B signaling pathway. Mol Med Rep. 19:4019–4026. 2019.PubMed/NCBI View Article : Google Scholar

28 

Wang YS, Li H, Li Y, Zhu H and Jin YH: Identification of natural compounds targeting annexin A2 with an anti-cancer effect. Protein Cell. 9:568–579. 2018.PubMed/NCBI View Article : Google Scholar

29 

Lee KH, Lee YH, Kim SI, Park JH and Lee SK: Ginsenoside-Rg5 suppresses cyclin E-dependent protein kinase activity via up-regulating p21Cip/WAF1 and down-regulating cyclin E in SK-HEP-1 cells. Anticancer Res. 17:1067–1072. 1997.PubMed/NCBI

30 

Cui Y, Su Y, Deng L and Wang W: Ginsenoside-Rg5 inhibits retinoblastoma proliferation and induces apoptosis through suppressing BCL2 expression. Chemotherapy. 63:293–300. 2018.PubMed/NCBI View Article : Google Scholar

31 

Feng SL, Luo HB, Cai L, Zhang J, Wang D, Chen YJ, Zhan HX, Jiang ZH and Xie Y: Ginsenoside Rg5 overcomes chemotherapeutic multidrug resistance mediated by ABCB1 transporter: In vitro and in vivo study. J Ginseng Res. 44:247–257. 2020.PubMed/NCBI View Article : Google Scholar

32 

Dong Y, Fu R, Yang J, Ma P, Liang L, Mi Y and Fan D: Folic acid-modified ginsenoside Rg5-loaded bovine serum albumin nanoparticles for targeted cancer therapy in vitro and in vivo. Int J Nanomedicine. 14:6971–6988. 2019.PubMed/NCBI View Article : Google Scholar

33 

Bortolotti P, Faure E and Kipnis E: Inflammasomes in tissue damages and immune disorders after trauma. Front Immunol. 9(1900)2018.PubMed/NCBI View Article : Google Scholar

34 

Yi YS: Roles of ginsenosides in inflammasome activation. J Ginseng Res. 43:172–178. 2019.PubMed/NCBI View Article : Google Scholar

35 

Kim TW, Joh EH, Kim B and Kim DH: Ginsenoside Rg5 ameliorates lung inflammation in mice by inhibiting the binding of LPS to toll-like receptor-4 on macrophages. Int Immunopharmacol. 12:110–116. 2012.PubMed/NCBI View Article : Google Scholar

36 

Park JY, Choi P, Kim T, Ko H, Kim HK, Kang KS and Ham J: Protective effects of processed ginseng and its active ginsenosides on cisplatin-induced nephrotoxicity: In vitro and in vivo studies. J Agric Food Chem. 63:5964–5969. 2015.PubMed/NCBI View Article : Google Scholar

37 

Li W, Yan M, Liu Y, Liu Z, Wang Z, Chen C, Zhang J and Sun YS: Ginsenoside Rg5 ameliorates cisplatin-induced nephrotoxicity in mice through inhibition of inflammation, oxidative stress, and apoptosis. Nutrients. 8(566)2016.PubMed/NCBI View Article : Google Scholar

38 

Lee SM: Anti-inflammatory effects of ginsenosides Rg5, Rz1, and Rk1: Inhibition of TNF-α-induced NF-κB, COX-2, and iNOS transcriptional expression. Phytother Res. 28:1893–1896. 2014.PubMed/NCBI View Article : Google Scholar

39 

Wang Z, Hu JN, Yan MH, Xing JJ, Liu WC and Li W: Caspase-mediated anti-apoptotic effect of ginsenoside Rg5, a main rare ginsenoside, on acetaminophen-induced hepatotoxicity in mice. J Agric Food Chem. 65:9226–9236. 2017.PubMed/NCBI View Article : Google Scholar

40 

Zhu Y, Zhu C, Yang H, Deng J and Fan D: Protective effect of ginsenoside Rg5 against kidney injury via inhibition of NLRP3 inflammasome activation and the MAPK signaling pathway in high-fat diet/streptozotocin-induced diabetic mice. Pharmacol Res. 155(104746)2020.PubMed/NCBI View Article : Google Scholar

41 

Cecconi M, Evans L, Levy M and Rhodes A: Sepsis and septic shock. Lancet. 392:75–87. 2018.PubMed/NCBI View Article : Google Scholar

42 

Andersson U, Wang H, Palmblad K, Aveberger AC, Bloom O, Erlandsson-Harris H, Janson A, Kokkola R, Zhang M, Yang H and Tracey KJ: High mobility group 1 protein (HMG-1) stimulates proinflammatory cytokine synthesis in human monocytes. J Exp Med. 192:565–570. 2000.PubMed/NCBI View Article : Google Scholar

43 

Bae JS: Role of high mobility group box 1 in inflammatory disease: Focus on sepsis. Arch Pharm Res. 35:1511–1523. 2012.PubMed/NCBI View Article : Google Scholar

44 

Kim JE, Lee W, Yang S, Cho SH, Baek MC, Song GY and Bae JS: Suppressive effects of rare ginsenosides, Rk1 and Rg5, on HMGB1-mediated septic responses. Food Chem Toxicol. 124:45–53. 2019.PubMed/NCBI View Article : Google Scholar

45 

Shin YW, Bae EA and Kim DH: Inhibitory effect of ginsenoside Rg5 and its metabolite ginsenoside Rh3 in an oxazolone-induced mouse chronic dermatitis model. Arch Pharm Res. 29:685–690. 2006.PubMed/NCBI View Article : Google Scholar

46 

Ahn S, Siddiqi MH, Aceituno VC, Simu SY, Zhang J, Jimenez Perez ZE, Kim YJ and Yang DC: Ginsenoside Rg5:Rk1 attenuates TNF-α/IFN-γ-induced production of thymus- and activation-regulated chemokine (TARC/CCL17) and LPS-induced NO production via downregulation of NF-κB/p38 MAPK/STAT1 signaling in human keratinocytes and macrophages. In Vitro Cell Dev Biol Anim. 52:287–295. 2016.PubMed/NCBI View Article : Google Scholar

47 

Chu S, Gu J, Feng L, Liu J, Zhang M, Jia X, Liu M and Yang D: Ginsenoside Rg5 improves cognitive dysfunction and beta-amyloid deposition in STZ-induced memory impaired rats via attenuating neuroinflammatory responses. Int Immunopharmacol. 19:317–326. 2014.PubMed/NCBI View Article : Google Scholar

48 

Cheng Z, Zhang M, Ling C, Zhu Y, Ren H, Hong C, Qin J, Liu T and Wang J: Neuroprotective effects of ginsenosides against cerebral ischemia. Molecules. 24(1102)2019.PubMed/NCBI View Article : Google Scholar

49 

Lee YY, Park JS, Jung JS, Kim DH and Kim HS: Anti-inflammatory effect of ginsenoside Rg5 in lipopolysaccharide-stimulated BV2 microglial cells. Int J Mol Sci. 14:9820–9833. 2013.PubMed/NCBI View Article : Google Scholar

50 

Liu FF, Zhang Z, Chen W, Gu HY and Yan QJ: Regulatory mechanism of microRNA-377 on CDH13 expression in the cell model of Alzheimer's disease. Eur Rev Med Pharmacol Sci. 22:2801–2808. 2018.PubMed/NCBI View Article : Google Scholar

51 

Bolognin S, Blanchard J, Wang X, Basurto-Islas G, Tung YC, Kohlbrenner E, Grundke-Iqbal I and Iqbal K: An experimental rat model of sporadic Alzheimer's disease and rescue of cognitive impairment with a neurotrophic peptide. Acta Neuropathol. 123:133–151. 2012.PubMed/NCBI View Article : Google Scholar

52 

Tao X, Finkbeiner S, Arnold D, Shaywitz A and Greenberg M: Ca2+ influx regulates BDNF transcription by a CREB family transcription factor-dependent mechanism. Neuron. 20:709–726. 1998.PubMed/NCBI View Article : Google Scholar

53 

Bourtchuladze R, Frenguelli B, Blendy J, Cioffi D, Schutz G and Silva A: Deficient long-term memory in mice with a targeted mutation of the cAMP-responsive element-binding protein. Cell. 79:59–68. 1994.PubMed/NCBI View Article : Google Scholar

54 

Kim EJ, Jung IH, Van Le TK, Jeong JJ, Kim NJ and Kim DH: Ginsenosides Rg5 and Rh3 protect scopolamine-induced memory deficits in mice. J Ethnopharmacol. 146:294–299. 2013.PubMed/NCBI View Article : Google Scholar

55 

Choi SY, Kim KJ, Song JH and Lee BY: Ginsenoside Rg5 prevents apoptosis by modulating heme-oxygenase-1/nuclear factor E2-related factor 2 signaling and alters the expression of cognitive impairment-associated genes in thermal stress-exposed HT22 cells. J Ginseng Res. 42:225–228. 2018.PubMed/NCBI View Article : Google Scholar

56 

Wu J, Jeong HK, Bulin SE, Kwon SW, Park JH and Bezprozvanny I: Ginsenosides protect striatal neurons in a cellular model of Huntington's disease. J Neurosci Res. 87:1904–1912. 2009.PubMed/NCBI View Article : Google Scholar

57 

Shao J, Zheng X, Qu L, Zhang H, Yuan H, Hui J, Mi Y, Ma P and Fan D: Ginsenoside Rg5/Rk1 ameliorated sleep via regulating the GABAergic/serotoninergic signaling pathway in a rodent model. Food Funct. 11:1245–1257. 2020.PubMed/NCBI View Article : Google Scholar

58 

Yuan CS, Attele AS, Wu JA and Liu D: Modulation of American ginseng on brainstem GABAergic effects in rats. J Ethnopharmacol. 62:215–222. 1998.PubMed/NCBI View Article : Google Scholar

59 

Sattler R and Tymianski M: Molecular mechanisms of glutamate receptor-mediated excitotoxic neuronal cell death. Mol Neurobiol. 24:107–129. 2001.PubMed/NCBI View Article : Google Scholar

60 

Yuan Q, Joiner WJ and Sehgal A: A sleep-promoting role for the Drosophila serotonin receptor 1A. Curr Biol. 16:1051–1062. 2006.PubMed/NCBI View Article : Google Scholar

61 

Cho YL, Hur SM, Kim JY, Kim JH, Lee DK, Choe J, Won MH, Ha KS, Jeoung D, Han S, et al: Specific activation of insulin-like growth factor-1 receptor by ginsenoside Rg5 promotes angiogenesis and vasorelaxation. J Biol Chem. 290:467–477. 2015.PubMed/NCBI View Article : Google Scholar

62 

Yang YL, Li J, Liu K, Zhang L, Liu Q, Liu B and Qi LW: Ginsenoside Rg5 increases cardiomyocyte resistance to ischemic injury through regulation of mitochondrial hexokinase-II and dynamin-related protein 1. Cell Death Dis. 8(e2625)2017.PubMed/NCBI View Article : Google Scholar

63 

Wojdasiewicz P, Poniatowski Ł and Szukiewicz D: The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of osteoarthritis. Mediators Inflamm. 2014(561459)2014.PubMed/NCBI View Article : Google Scholar

64 

Cheng W, Jing J, Wang Z, Wu D and Huang Y: Chondroprotective effects of ginsenoside Rg1 in human osteoarthritis chondrocytes and a rat model of anterior cruciate ligament transection. Nutrients. 9(263)2017.PubMed/NCBI View Article : Google Scholar

65 

Chen Y, Lin S, Sun Y, Pan X, Xiao L, Zou L, Ho KW and Li G: Translational potential of ginsenoside Rb1 in managing progression of osteoarthritis. J Orthop Translat. 6:27–33. 2016.PubMed/NCBI View Article : Google Scholar

66 

Zhang P: Ginsenoside-Rg5 treatment inhibits apoptosis of chondrocytes and degradation of cartilage matrix in a rat model of osteoarthritis. Oncol Rep. 37:1497–1502. 2017.PubMed/NCBI View Article : Google Scholar

67 

Wang T, Liu Q, Tjhioe W, Zhao J, Lu A, Zhang G, Tan RX, Zhou M, Xu J and Feng HT: Therapeutic potential and outlook of alternative medicine for osteoporosis. Current Drug Targets. 18:1051–1068. 2017.PubMed/NCBI View Article : Google Scholar

68 

Siddiqi MH, Siddiqi MZ, Ahn S, Kang S, Kim YJ, Veerappan K, Yang DU and Yang DC: Stimulative effect of ginsenosides Rg5:Rk1 on murine osteoblastic MC3T3-E1 cells. Phytother Res. 28:1447–1455. 2014.PubMed/NCBI View Article : Google Scholar

69 

Ponnuraj SP, Siraj F, Kang S, Noh HY, Min JW, Kim YJ and Yang DC: Amelioration of insulin resistance by Rk1 + Rg5 complex under endoplasmic reticulum stress conditions. Pharmacognosy Res. 6:292–296. 2014.PubMed/NCBI View Article : Google Scholar

70 

Xiao N, Lou MD, Lu YT, Yang LL, Liu Q, Liu B, Qi LW and Li P: Ginsenoside Rg5 attenuates hepatic glucagon response via suppression of succinate-associated HIF-1α induction in HFD-fed mice. Diabetologia. 60:1084–1093. 2017.PubMed/NCBI View Article : Google Scholar

71 

Xiao N, Yang LL, Yang YL, Liu LW, Li J, Liu B, Liu K, Qi LW and Li P: Ginsenoside Rg5 inhibits succinate-associated lipolysis in adipose tissue and prevents muscle insulin resistance. Front Pharmacol. 8(43)2017.PubMed/NCBI View Article : Google Scholar

72 

Bai L, Gao J, Wei F, Zhao J, Wang D and Wei J: Therapeutic potential of ginsenosides as an adjuvant treatment for diabetes. Front Pharmacol. 9(423)2018.PubMed/NCBI View Article : Google Scholar

73 

Park HJ, Kim JH and Shim I: Anti-obesity effects of ginsenosides in high-fat diet-fed rats. Chin J Integr Med. 25:895–901. 2019.PubMed/NCBI View Article : Google Scholar

74 

Liu H, Liu M, Jin Z, Yaqoob S, Zheng M, Cai D, Liu J and Guo S: Ginsenoside Rg2 inhibits adipogenesis in 3T3-L1 preadipocytes and suppresses obesity in high-fat-diet-induced obese mice through the AMPK pathway. Food Funct. 10:3603–3614. 2019.PubMed/NCBI View Article : Google Scholar

75 

Liu H, Wang J, Liu M, Zhao H, Yaqoob S, Zheng M, Cai D and Liu J: Antiobesity effects of ginsenoside Rg1 on 3T3-L1 preadipocytes and high fat diet-induced obese mice mediated by AMPK. Nutrients. 10(830)2018.PubMed/NCBI View Article : Google Scholar

76 

Zhang L, Zhang L, Wang X and Si H: Anti-adipogenic effects and mechanisms of ginsenoside Rg3 in pre-adipocytes and obese mice. Front Pharmacol. 8(113)2017.PubMed/NCBI View Article : Google Scholar

77 

Yesmin Simu S, Ahn S, Castro-Aceituno V and Yang DC: Ginsenoside Rg5: Rk1 exerts an anti-obesity effect on 3T3-L1 Cell Line by the downregulation of PPARγ and CEBPα. Iran J Biotechnol. 15:252–259. 2017.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

August-2021
Volume 22 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu M, Liu F, Gao Y, Yin J, Yan W, Liu J and Li H: Pharmacological activities of ginsenoside Rg5 (Review). Exp Ther Med 22: 840, 2021
APA
Liu, M., Liu, F., Gao, Y., Yin, J., Yan, W., Liu, J., & Li, H. (2021). Pharmacological activities of ginsenoside Rg5 (Review). Experimental and Therapeutic Medicine, 22, 840. https://doi.org/10.3892/etm.2021.10272
MLA
Liu, M., Liu, F., Gao, Y., Yin, J., Yan, W., Liu, J., Li, H."Pharmacological activities of ginsenoside Rg5 (Review)". Experimental and Therapeutic Medicine 22.2 (2021): 840.
Chicago
Liu, M., Liu, F., Gao, Y., Yin, J., Yan, W., Liu, J., Li, H."Pharmacological activities of ginsenoside Rg5 (Review)". Experimental and Therapeutic Medicine 22, no. 2 (2021): 840. https://doi.org/10.3892/etm.2021.10272