Skip to main content
Journal of Ginseng Research logoLink to Journal of Ginseng Research
. 2021 Dec 22;46(6):711–721. doi: 10.1016/j.jgr.2021.12.007

Ginsenosides are active ingredients in Panax ginseng with immunomodulatory properties from cellular to organismal levels

Long You a, Seunghwa Cha b, Mi-Yeon Kim b,∗∗, Jae Youl Cho a,c,
PMCID: PMC9597430  PMID: 36312737

Abstract

The immune system is one of the most important parts of the human body and immunomodulation is the major function of the immune system. In response to outside pathogens or high inflammation, the immune system is stimulated or suppressed. Thus, identifying effective and potent immunostimulants or immunosuppressants is critical. Ginsenosides are a type of steroid saponin derived from ginseng. Most are harmless to the body and even have tonic effects. In this review, we mainly focus on the immunostimulatory and immunosuppressive roles of two types ginsenosides: the protopanaxadiol (PPD)-type and protopanaxatriol (PPT)-type. PPT-type ginsenosides include Rg1, Rg2, Rh4, Re and notoginsenoside R1, and PPD-type ginsenosides include Rg3, Rh2, Rb1, Rb2, Rc, Rd, compound K (CK) and PPD, which activate the immune responses. In addition, Rg1 and Rg6 belong to PPT-type ginsenosides and together with Rg3, Rb1, Rd, CK show immunosuppressive properties. Current explorations of ginsenosides in immunological areas are in the preliminary stages. Therefore, this review may provide some novel ideas to researchers who study the immunoregulatory roles of ginsenosides.

Keywords: Ginsenoside, Immunomodulation, Immunostimulant, Immunosuppressant, Immune response

Graphical abstract

Image 1

1. Introduction

After the worldwide pandemic caused by COVID-19, increasing numbers of people have realized that innate body immunity is the first barrier to safeguard their health. Thus, boosting human immunity is an important issue. Immunity is the body's ability to defend against the invasion of outside pathogens including virus, bacteria, or other microorganisms. Of course, for general people, immune boosting is a common saying in immune system in the narrower sense. There are two types of immunity, innate immunity and adaptive immunity [1]. Innate immunity is considered to be the first barrier of defense against pathogens, though this kind of protection is fundamental [2]. Adaptive immunity is activated when innate immunity is insufficient against the pathogen. Therefore, adaptive immunity is more specific than the innate component [3]. The adaptive immune response is mainly carried out by two kinds of specialized cells, T-cells and B-cells. T-cells are in charge of the cell-mediated immune response, whereas B-cells control the humoral immune response [3]. Although adaptive immunity can specifically attack pathogens, this kind of response takes three to five days, even weeks, to produce sufficient lymphocytes to kill the pathogens. If there is no innate response, most pathogens have enough time to damage the host during this period. Because innate immunity can be rapidly activated, control the infection, and gather pathogen information, it is considered as the one of main protective functions in the immune system [[1], [2], [3], [4]]. The role of memory in adaptive immunity can prevent re-infection in the long term. If the host is exposed to the same type of pathogen, the adaptive immunity can respond and quickly eliminate the threat [3,5].

The response of the immune system can categorized into two categories: immunostimulatory (increasing immunity) and immunosuppressive (decreasing immunity) response [6]. Immunostimulatory activity means that the immune response is enhanced against many disorders, such as infection of pathogenic microorganisms, immunodeficiency or even cancers [[7], [8], [9]]. Many drugs, called immunostimulants, such as lipopolysaccharides (LPS), can up-regulate the immune system. LPS is present in almost all Gram-negative bacteria. It is one of the exceedingly strong activators for innate immunity that rapidly activates macrophages and B lymphocytes [[10], [11], [12]]. The main functions of immunostimulants are to induce the immune system of humans and animals to improve the body's resistance and prevent disease.

Drugs that down-regulate the immune responses are called immunosuppressants. As the name suggests, immunosuppressants inhibit the immune response and are prescribed to prevent immune rejection in organ or tissue transplantation and to treat autoimmune diseases such as allergies, pemphigus, and lupus [6,[13], [14], [15]]. For example, glucocorticoids have been used in the clinic for many years and show a high success in treating acute immune rejection of organ transplants or severe autoimmune disorder [[15], [16], [17]]. They act as transcription factors to directly bind to DNA through combining with receptors and also bind and interfere with other transcription factors [18]. Immunomodulation drugs contain immunostimulants and immunosuppressants, which have their own capabilities. To maintain homeostasis or return to a normal state, suitable immunostimulants or immunosuppressants must be selected and administered.

As more and more immunomodulators have been discovered, researchers found that these immunomodulators have numerous functions. To study the mechanisms of immunomodulation, multiple cells that are related with immune systems have been studied. These cells include neutrophils, monocytes or macrophages, and lymphocytes, which include T-cells, B-cells and natural killer (NK) cells [19,20]. Neutrophils, namely human neutrophilic polymorphonuclear leukocytes [21], are produced in the bone marrow and are located in the bloodstream to provide a potent defense against infections through rapidly migrating to the sites of infections [[21], [22], [23]]. Neutrophils are a pivotal component in the immune response that not only recruit, activate, and program antigen-presenting cells (APCs), but also produce tumor-necrosis factor (TNF) and other cytokines such as interleukin (IL)-1α, IL-1β, and IL-6 to stimulate the differentiation and activation of dendritic cells (DCs) and macrophages [23,24]. Neutrophils have a close relationship with monocytes and macrophages. Monocytes are derived from peripheral blood mononuclear cells (PBMCs). They also shift in the bloodstream and capture the pathogenic microorganisms. Once macrophages mature and migrate into tissues, they differentiate into DCs and macrophages [25,26]. As the first barrier to defend against pathogens, macrophages ingest microorganisms and directly deliver noxious components outside [27]. Lymphocytes are also extremely crucial immune cells in the immune system. They are developed from stem cells that are located in the bone marrow and work in the blood and lymph tissues. If these cells move to the thymus, they become T-cells. Lymphocytes that remain in the bone marrow are called B-cells [28,29]. Based on their functions, T-cells are classified into three types: helper T-cells, killer T-cells, and regulatory T-cells. These cells express different cell surface proteins, CD4, CD8 and CD4 FOXP3 [[30], [31], [32]]. B-cells also have three subsets, which includes B-1, follicular and marginal zone B cells. Depending on the condition, they can be activated in the presence or the absence of T-cells [33]. NK cells, which comprise a minor part of total lymphocytes, are distributed in both lymphoid and nonlymphoid tissues [31,34]. Based on the difference of homing properties, NK cells in humans are divided into two subsets, CD56dim and CD56bright.

We next discuss the immune system based on its different components, including white blood cells, antibodies, complement system, lymphatic system, spleen, bone marrow, and thymus. White blood cells are the crucial player in our immune system. They are produced by the thymus and contain two main types, granulocytes and agranulocytes. Granulocytes include neutrophils, eosinophils and basophils, and agranulocytes include lymphocytes and monocytes [35]. Neutrophils are the major component of our white blood cells and play a role as body scavengers that get rid of many bacteria and fungus. Eosinophils respond to a variety of infection and inflammation and basophils are highly present in the allergic reaction. The function of monocytes is to target and fight off chronic infections.

Immune antibodies come from T-cells and B-cells. They recognize the antigens produced by outside microbes or toxins. Once the antibodies find the invader, the cells will quickly defend against the threat. Vaccines are designed based on this mechanism. Vaccines stimulate our immune system and produce antibodies. There are five major immunoglobulins (Ig) antibodies: IgA, IgG, IgM, IgD and IgE. These antibodies are all generated by B cells. IgA, IgM and IgG are commonly used in diagnosis. When our bodies are infected by organisms, IgA and IgM are quickly made by our immune systems and act as the earliest antibodies to defend or delay the infection. Within several weeks, IgG is produced in our immune system, and IgA and IgM are gradually broken down. Thus, IgA and IgM act in the short-term whereas IgG are involved in a long-lasting response. The complement system consists of some proteins that complement the functions of antibodies.

The lymphatic system contains lymph nodes, lymph vessels and lymphocytes [36]. During infection, lymph nodes can catch the microbes. Lymphocytes are carried by lymph vessels to fight against the infection. The spleen acts as a filter of blood to removes damage and bone marrow serves as a factory to produce red blood cells, white blood cells and platelets [37].

Ginseng is a traditional herb that has been used for various conventional medical therapies for more than 5000 years. Historically, people believed that ginseng could prolong human life. Current studies have shown that ginseng exhibits multiple therapeutic properties against various diseases, like physical and infectious diseases and diverse cancers. Ginseng includes Panax ginseng Meyer (Korean ginseng), P. quinquefolius L. (American ginseng), P. japonicus Meyer (Japanese ginseng) and P. notoginseng (Burk) F.H. Chen (Sanqi-ginseng) [38,39]. Korean ginseng is the most studied and widely used in all over the world, and its roots are a main focus of research [[40], [41]]. Based on the water content and processing method, Korean ginseng, especially the roots, can be categorized as fresh ginseng, white ginseng and red ginseng [42]. Red ginseng is incubated in 100 °C steam for more than 2 h until the moisture content is less than 15%.

Ginsenosides, which belong to steroid saponins, are the major components of ginseng. Over 100 ginsenosides have been separated from the extractions. These ginsenosides are named as Rx: the ‘x’ represents the polarity of each ginsenoside on the thin-layer chromatography, in which the most polar component is named as Ra, while the least polar one is addressed as Rh [42]. According to the difference of backbone [43], ginsenosides can be classified into four types, the PPD-, PPT-, ocotillol- and oleanolic acid-types (Fig. 1). PPD-type ginsenosides include Ra1, Ra2, Ra3, Rb1, Rb2, Rb3, Rc, Rd, F2, Rg3, Rg5, Rh2, Rh3, CK and PPD. PPT-type ginsenosides include Re, Rg1, Rg2, Rg4, Rh1, Rh4, Rf and PPT [[44], [45], [46]]. Ocotillol-type ginsenosides contain 24(R)-pseudoginsenoside-F11, 24(R)-pseudoginsenoside-RT5, Majonoside R1 and R2 [47], and Ro is an oleanolic acid-type ginsenoside.

Fig. 1.

Fig. 1

Types of ginsenosides.

Ginseng has been used as an immunomodulatory herb for a long time. Ginseng is used to reduce inflammation induced by infection by outside microorganisms and can also be applied for boosting human immunity. However, these activities are not paradoxical. Because of its complicated properties, ginseng has attracted attention from researchers. However, the immunoregulatory role of ginseng is not well understood. Thus, in this review, we specifically focus on the immunomodulatory roles of ginsenosides and discuss the functions of ginsenosides from the cell to body levels. Other components have similar functions such as ginseng polysaccharides. The use of ginsenosides as immunomodulators may be useful for further studies and also as a guideline for commercial development.

2. Main body

2.1. The immunostimulatory role of ginsenosides

In research on the immunostimulatory functions of some drug, the Th1 and Th2 responses will be considered. Th1 and Th2 are T helper type 1 and 2, respectively, which are associated with helper T-cells. Helper T-cells are the T lymphocytes that express CD4. The Th1 immune response is also called cellular immune response. In the Th1 response, Th1 cells act as an activator to enhance human immunity and induce phagocyte-dependent inflammation [48]. The Th2 response, also known as the humoral immune response, is related with the promotion of the responses of antibody like immunoglobulin E (IgE) and the accumulation of eosinophils, as well as inactivation of phagocytic cells [48]. Th1 cells secrete interferon (IFN)-γ, IL-2 and TNF-β, whereas Th2 cells produce IL-4, IL-5, IL-6, IL-13 and other cytokines. Because of this, Th2 is mostly like to counteract excessive Th1 response, which will cause uncontrolled damage [49]. In the investigation of immunostimulatory roles of ginsenosides, more attention can be paid to the relationships between ginsenosides and Th1 and Th2 responses.

2.1.1. The mechanisms of immunostimulatory role of ginsenosides in vitro

As one of the most important components in ginseng, ginsenosides are a focus of much attention. Previously, ginseng was known to boost human immunity. However, research is ongoing into the mechanisms of the immunostimulatory role of ginseng. Therefore, the ginsenosides have been isolated and studied. Shin et al. compared the immune stimulating role of white ginseng and heat-processed ginseng (HPG) and found that HPG, which contains Rg3, Rg5 and Rk1 complex, can enhance the activation of macrophages through inducing the mitogen-activated protein kinase (MAPK)/activator protein 1 (AP-1) and nuclear factor-κB (NF-κB) signaling pathways, as indicated in Fig. 2. In addition, they found that extracellular signal-regulated kinase (ERK)/c-Jun is the main mechanism for this activation and the cytokines TNF-α and IL-6 were produced [50]. Rg3 was also studied for its effects in macrophage phagocytosis. Rg3 promotes phagocytosis of IgG-opsonized Escherichia coli and IgG-opsonized beads (IgG beads) through activating ras-related C3 botulinum toxin substrate 1 (Rac1) and cell division cycle 42 (CDC42) and up-regulating the phosphorylation of ERK and p38 MAPK. IL-6 and TNF-α were also induced [51]. Kim et al. compared red ginseng (RG) and fermented RG (FRG) and found that the content of Rd, Rh1, F2 and Rg3 were increased. FRG up-regulated the activity of macrophages much more significantly than RG through MAPK and NF-κB pathways. In addition, FRG can boost the immunostimulatory function in central immune cells which contains mouse splenocytes and bone marrow-derived macrophages by promoting the proliferation and production of TNF-α and IL-6 [52]. Another two PPD-types ginsenosides, 20S-dihydroprotopanaxadiol (2H-PPD) and CK, reported by Kim et al. [53] and Yang et al. [54] respectively, showed an immunostimulatory role on the innate immune response. Both activate monocytes and macrophages through increasing phagocytic uptake, cell to cell aggregation, and the surface level of costimulatory molecules CD80 and CD86. Surface level CD43 was also enhanced by these two compounds whereas CD29 was increased by 2H-PPD. Some cytokines such as TNF-α can be promoted by CK through NF-κB and AP-1 pathways. However, 2H-PPD cannot induce the adhesion of cells to the extracellular matrix and CK suppressed the interactions of cell to tissue. CD82, which is the marker of monocyte differentiation, was also inhibited by CK.

Fig. 2.

Fig. 2

The immunomodulatory role of ginsenosides in vitro.

Although most in vitro experiments were performed by PPD-type ginsenosides, some other ginsenoside which derived from PPT-type were also used in vitro. Rg1, which is a major component of PPT-type ginsenoside, was studied by Wang et al. [55]. Rg1 can dramatically boost the innate immune response in macrophages. In response to LPS-activated macrophages, Rg1 enhanced the production of TNF-α through up-regulating phosphatidylinositol-3-kinase (PI3K)/Akt and the mammalian target of rapamycin (mTOR) signaling pathways and down-regulating the secretion of IL-6 via suppressing the NF-κB pathway. In addition, Rg1 was also tested in the splenocytes of BALB/c mice to find the mechanisms of enhancement of activity of CD4+ T cells [56]. Rg1 increased the immune activity of CD4+ T cells through accelerating their proliferation. This activation owed to the enhancements of CD3, CD28 and CD69. Apart from that, Rg1 suppressed the Th1 immune response by restraining IFN-γ whereas enhance the Th2 response by increasing IL-4 in purified CD4+ T cells, which adjusted the balance of Th1 and Th2 and corrected the Th1-dominant pathological disorders (Table 1).

Table 1.

The immunomodulatory role of ginsenosides in vitro.

The immunostimulatory role of ginsenosides in vitro
Type of ginsenosides Ginsenosides Target cells Signaling pathways Cytokine/molecules Ref.
FRG (Rd, Rh1, F2) Macrophages
Splenocytes
MAPKs
NF-κB
TNF-α↑
IL-6↑
[52]
PPD-type HPG (Rg3, Rg5, Rk1) Macrophages MAPKs/AP-1
NF-κB
TNF-α↑
IL-6↑
[50]
Rg3 Macrophages Rac1/CDC42
ERK1/2 and p38
TNF-α↑
IL-6↑
[51]
2H-PPD Macrophages
Monocytes
CD29, CD43↑
CD80, CD86↑
[53]
CK Macrophages
Monocytes
AP-1
NF-κB
CD43, CD69↑
CD80, CD86↑
TNF-α↑
[54]
PPT-type Rg1 Macrophages NF-κB
PI3K/AKT/mTOR
TNF-α↑
IL-6↑
[55]
T helper cells IL-6, IL-4↑
CD3, CD28, CD69↑
[56]
The immunosuppressive role of ginsenosides in vitro
PPD-type 6″-O-acetylginsenoside Rb1 Macrophages TNF-α↓
IL-6↓
[82]
Rg3 NSCLC cells AKT, ERK and NF-κB/HIF-1α [83]
PPT-type Rg1 Macrophages NF-κB TNF-α↓
IL-6↓
[79]
Rg6 BMDMs NF-κB
MAPKs
TNF-α, IL-1β↓
IL-12p40↓
IL-6↓
[81]

2.1.2. The immunostimulatory applications of ginsenosides in vivo

2.1.2.1. The immunostimulatory mechanism of PPT-type ginsenoside in vivo

As a natural tonic herb, ginseng has been used as an immune booster for more than thousands of years. Ginsenosides, the major component of ginseng, has been tested in many animal models for research on their mechanisms of immunomodulatory action (Fig. 3A). Currently, the major role of ginsenosides is acting as adjuvant, as well as a few cases were still applied in vivo alone. Rg1 was used as adjuvant in BALB/c mice. Sun et al. screened many ginsenosides containing both PPT- and PPD-types and found that Rg1 is the most potent and ideal adjuvant that when co-administrated with ovalbumin (OVA) exhibited immunostimulatory properties [57]. Combining Rg1 with OVA caused a huge promotion of IgG1 and IgG2a in mouse blood and led to a great proliferation of lymphocytes. All the enhancements are ascribable to IL-5/IFN-γ production and Th1/Th2 activation. Rg2 and Re also exhibited similar characteristics as Rg1v [57]. Sun et al. investigated co-treatment of Rg1 and aluminum hydroxide to check the immune response to OVA [58]. Similar results were obtained as well as delayed-type hypersensitivity and less hemolytic activity. Another study in BALB/c mice examined co-treatment Rg1 with hepatitis B surface antigen (HBsAg) [59]. Rg1 not only increased the Th1 responses including IgG2b and IFN- γ but also enhanced the Th2 responses containing IgG1 and IL-4 through TLR4 signaling pathway. Qu et al. and Su et al. also studied the Rg1 combination with T. gondii recombinant surface antigen 1 (rSAG1) and inactivated porcine reproductive and respiratory syndrome virus (PRRSV) vaccine, respectively, in ICR mice. Co-administration of Rg1 and rSAG1 significantly enhanced the activities of NK cells and T helper cells and stimulated higher expressions of IL-4 and IFN- γ, which indicating the promising adjuvant together with rSAG1 against toxoplasmosis [60]. PRRSV vaccine combined with Rg1 activated IgG and IgA antibodies and neutralizing antibody titer, increased the proliferation of T and B lymphocytes as well as the CD107a expression in CD4+ T cells. In addition, many cytokines which related to Th1 (IFN-γ and IL-2), Th2 (IL-5 and IL-10), and Th17 (IL-6 and IL-17A) were accumulated by stimulating type I interferon signaling pathway [61]. Chickens can also be used to test the association of Rg1 and infectious bursal disease vaccine. These chickens underwent the oxidative stress which caused by cyclophosphamide (CP). This combination not only relieved the oxidative stress but also raised the proliferation of lymphocyte and the cytokines of immune response like IFN-γ and IL-6 [62].

Fig. 3.

Fig. 3

The immunomodulatory role of ginsenosides in vivo. (A) The immunostimulatory mechanism of PPT- and PPD-type ginsenosides in vivo. (B) The immunosuppressive mechanism of PPT- and PPD-type ginsenosides in vivo.

We previously introduced Rg1, which acts as an adjuvant against OVA in BALB/c mice. Another animal model, ICR mice, was used to examine the immune response against OVA together with various ginsenosides, such as Re, notoginsenoside R1 and Rh4 [63,64]. Similar to Rg1, these three ginsenosides dramatically promoted the proliferation of splenocytes induced by concanavalin A (Con A), LPS and OVA. Additionally, the specific antibodies IgG, IgG1 and IgG2b were also increased in serum. Moreover, there were none or low hemolytic effects of these three ginsenosides, indicating they show safe properties as an adjuvant.

Several vaccines were tested on the ICR mice with the addition of Re. H3N2, an inactivated influenza virus antigen, is one of the vaccines was evaluated together with Re. IgG, IgG1, IgG2a, IgG2b and HI titers, which were derived from serum, were significantly amplified. Both Th1 and Th2 cytokines IFN-γ and IL-5 were highly secreted with the proliferation of lymphocytes, which showed mixed immune responses for enhancing the activities of vaccine [65]. Two other studies examined recombinant Toxoplasma gondii ROP18 antigen [66] and inactivated rabies virus vaccine (RV) [67]. Beside the enhancements of IgG production and Th1/Th2 cytokines (IFN-γ, IL-4, IL-10 and IL-12), RV vaccine with Re significantly increased the ratio of CD4+ and CD8+. These studies demonstrated that PPT-type ginsenoside can improve the efficiency of vaccines in activating the immune responses of Th1 and Th2.

2.1.2.2. The immunostimulatory mechanism of PPD-type ginsenoside in vivo

Rg3 effectively induces macrophage phagocytosis in mouse lung through activating the phosphorylation of ERK1/2 and p38 [51], and also significantly promotes the cellular immunity of KM mice that were transplanted with the hepatocellular carcinoma cell line H22 [68]. After Rg3 treatment, lymphocytes induced by Con A were highly proliferated and two cytokines, IFN-γ and IL-2, which derived from the Th1 immune response, were accumulated (Fig. 3A). This immunostimulatory role caused the suppression of xenografted H22 tumors could provide guidance for clinical tumor immunity. CP is a chemotherapeutic agent that caused immune suppression. With CP injury, macrophage phagocytosis is inhibited by decreased activities of lactate dehydrogenase and acid phosphatase as well as the production of IgG, IL-2 and granulocyte colony-stimulating factor. In addition, the disordered balance of Th1 and Th2 resulted from the suppression of T-Cell-Specific T-Box Transcription Factor T-bet and IFN-γ and the enhancement of trans-acting T-cell-specific transcription actor GATA-3 and IL-4. Rg3 partially recovered the immunity of immunodeficient BALB/c mice caused by CP, including the activation of macrophages and T cells as well as the balance of the Th1 and Th2 immune response [69]. Although CP is used to treat non-small cell lung cancer (NSCLC) as a chemotherapeutic drug, it causes the severe immune suppression. Qian et al. reported that this serious immune deficiency can be improved by Rh2 through down-regulation of fatty acid metabolism [70]. Rh2 induced the oxidative decomposition of fatty acid and attenuated the protein expression of fatty acid synthase, which caused immune enhancement in CP-treated mice.

Interestingly, Rh2 also showed a similar immune response against tumor [71]. Wang et al. established a melanoma C57BL/6 mouse model by injecting B16F10 melanoma cells. The administration of Rh2 caused inhibition of tumor growth and induced the infiltration of CD4+ and CD8+ lymphocytes. Rh2 increased the cytotoxicity of lymphocytes to suppress B16F10 cells, which promotes the immunological response against melanoma.

In screening the effects that enhance the immune responses to OVA in BALB/c mice, some PPD-type ginsenosides like Rg3 and Rb1 also displayed properties of immunostimulants [57,72]. Song et al. co-administered the nanoparticles of PPD-type ginsenosides (Rb2, Rc, Rb1 and Rd) with OVA in ICR mice [73]. Compared with OVA administration alone, co-treatment these PPD-type nanoparticles can remarkably enhance the levels of IgG1, IgG2a, IgG2b and IgG3. In addition, the proliferation of B and T lymphocytes was accelerated. The productions of cytokines IFN-γ and IL-5, which belongs to Th1 and Th2 immune responses, were also higher than those with OVA injected alone. Yang et al. reported that Rd not only can increase the cytokines above, but it also can induce the expressions of IL-2, IL-4 and IL-10 in ICR mice to resist OVA [74]. Han et al. used Rd to assist Candida albicans surface mannan (CASM) to produce antibody against candidiasis [75]. BALB/c mice that were intraperitoneally injected with CASM and Rd induced the Th1 immune response by strongly producing IFN-γ. Moreover, the high increase of IL-4 and the ratio of IgG2a to IgG showed the effective Th2 response, which suggested that Rd can be a useful immunostimulatory adjuvant.

Rb1 was used as an immunostimulatory adjuvant to test the porcine parvovirus (PPV) vaccine [76]. Cytokines were collected in serum or the cultured supernatants of lymphocyte of BALB/c mice and expression levels were measured by ELISA. The results demonstrated that the vaccine containing Rb1 resulted in dramatically enhanced secretion of cytokines such as TNF-α, IFN-γ, IL-2, IL-4 and IL-10 from the Th1 and Th2 immune response. These increased cytokines together with the stimulation of specific IgG1, IgG2a and IgG2b revealed that Rb1 can balance the immune response of Th1 and Th2. However, as the immunostimulatory agent, Rb1 displayed an antiviral role to treat enterovirus 71 (EV71) infection [77]. Suckling ICR mice were infected by EV71 and treated with Rb1. The results showed that Rb1 reduced the cytopathic effect and the protein level of viral protein-1 (VP-1) caused by EV71. Immune cytokines such as TNF-α, IFN-γ and IL-10 and specific antigens like IgA, IgG1 and IgG2a were significantly increased, indicating enhanced immune responses by Rb1. In addition, Rb1 strengthened the expression of IFN-β. Upon knockdown of IFN-β, VP-1 was not significantly be inhibited by Rb1 although it was remarkably suppressed by Rb1. This finding demonstrated that the antiviral function of Rb1 is directly related with host immune responses, which provides a link between immune responses and antiviral therapeutics (Table 2).

Table 2.

The immunomodulatory role of ginsenosides in vivo.

The immunostimulatory role of ginsenosides in vivo
Type of ginsenosides Ginsenosides Applications Animal models Signaling pathways/mechanisms Cytokine/molecules Ref.
PPD-type Rg3 Mouse macrophages Lung injury in BALB/c mice ERK1/2 p38 TNF-α↑
IL-6↑
[51]
Lymphocytes H22 transplanted KM mice Th1 IFN-γ↑
IL-2↑
[68]
Mouse macrophages and Lymphocytes CP-injury
In BALB/c mice
Th1/Th2 IgG, IL-2↑
T-bet, IFN-γ↑
GATA-3↑
IL-4↑
[69]
Rh2 CP-treated
C57BL/6J
BALB/c mice
Fatty acid metabolism [70]
Lymphocytes B16F10-injected C57BL/6 mice CD4+ and CD8a+ T-lymphocytes' infiltration [71]
nanoparticles (Rb2, Rc, Rb1 and Rd) Lymphocytes Against OVA in ICR mice Th1/Th2 IgG↑
IFN-γ, IL-5↑
[73]
Rd adjuvant Against OVA in ICR mice Th1/Th2 IL-2, IL4↑
IL-10↑
[73]
Adjuvant with CASM Candidiasis-infected BALB/c mice Th1/Th2 IFN-γ↑
IL-4, IgG2a↑
[75]
Rb1 Adjuvant with PPV vaccine PPV infected BALB/c mice Th1/Th2 TNF-α, IFN-γ IL-2, IL-4,
IL-10, IgG↑
[76]
Adjuvant with EV71 EV71 infected ICR mice Th1/Th2 TNF-α, IFN-γ, IL-10, IgA,
IgG↑
[77]
PPT-type Rg1, Rg2 Adjuvant Against OVA in BALB/c mice Th1/Th2 IgG1, IgG2a↑
IFN-γ, IL-5↑
[57]
Rg1 Adjuvant with aluminum hydroxide Against OVA in BALB/c mice Th1/Th2 IgG1, IgG2a↑
IFN-γ, IL-5↑
[58]
Adjuvant with HBsAg Hepatitis B infected BALB/c mice Th1/Th2
TLR4
IFN-γ, IgG2b↑
IL-4, IgG1↑
[59]
Adjuvant with rSAG1 T. gondii infected ICR mice NK cells T helper cells IFN-γ, IL-4↑ [60]
Adjuvant with PRRSV vaccine PRRSV infected ICR mice Th1/Th2/Th17 type I interferon IFN-γ, IL-2↑
IL-5, IL-10↑
IL-6, IL-17A↑
[61]
Adjuvant with bursal disease vaccine CP infected chicken Th1 IFN-γ, IL-6↑ [62]
Notoginsenoside R1, Rh4 Adjuvant Against OVA in ICR mice Th1 IgG, IgG1, IgG2b↑ [63]
[64]
Re Adjuvant with H3N2 Inactivated influenza virus infected ICR mice Th1/Th2 IgG, IgG1, IgG2a, IgG2b, HI titer↑
IFN-γ, IL-5↑
[65]
Adjuvant with ROP18 antigen Toxoplasma gondi infected ICR mice Th1/Th2 IFN-γ, IL-4, IL-10, IL-12↑ [66]
Adjuvant with RV Inactivated rabies virus infected ICR mice Th1/Th2 IFN-γ, IL-4, IL-10, IL-12↑ ratio of CD4+ and CD8+ [67]
The immunosuppressive role of ginsenosides in vivo
PPD-type Rg3 C57BL/6 mice with RA Th1 TNF-α, IL-6↓ [85]
CK Lymphocytes Acute cardiac allograft rejection in C57BL/6, BALB/c mice Th1 IL-2, IFN-γ↓ [86]
Lymphocytes STZ-induced diabetic mice with islet transplantation CD4+, CD8+ T cells↓ [87]
Rd Allo-skin transplanted C57BL/6 and BALB/c mice Th1 TNF-α, IL-12↓ [88]
PPT-type Rg1 Lymphocytes CLP-induced C57BL/6 induced Th1/Th2 TNF-α, IL-6 IL-10↓ [78]
Lymphocytes CFA-induced AIA 3D rats NF-κB TNF-α
IL-6↓
[79]
Lymphocytes type 2 PRRSV-infected piglets Th1 TNF-α, IL-1β, IL-6,
IL-8↓
[80]
Rg6 Lymphocytes LPS-induced C57BL/6 mice (Septic shock) NF-κB MAPKs TNF-α, IL-1β, IL-6,
IL-12p40↓
[81]

2.2. The immunosuppressive role of ginsenosides

Since the immunostimulatory mechanisms of ginsenosides both in vitro and in vivo have been introduced, next we will discuss the immunosuppressive application of each ginsenoside according to the classification. Kim et al. [43] reviewed the immunosuppressive roles of ginsenosides prior to 2016 very well. Therefore, here we summarize studies related to immunosuppression of ginsenosides from 2016 and onwards. Many studies examined both in vitro and in vivo findings and we will also combine them here (Table 1, Table 2).

2.2.1. The immunosuppressive mechanisms and applications of PPT-type ginsenosides

People with sepsis have a high mortality rate because these individuals show an unbalanced immune response and apoptotic lymphocytes. The effects of Rg1, a PPT-type ginsenoside, on sepsis C57BL/6 mice induced by cecal ligation and puncture (CLP) were examined [78]. Rg1 remarkably reversed the high expressions of TNF-α, IL-6 and IL-10 induced by CLP and limited the apoptotic splenocytes by decreasing the highly apoptotic lymphocytes in the thymus (Fig. 3B). Rg1 also inhibited the inflammation of RAW264.7 cells stimulated by LPS and relieved the damage of adjuvant-induced arthritis (AIA) 3D rats induced by Complete Freund's adjuvant (CFA) [79]. The immune cytokines (TNF-α, IL-6) and NF-κB signaling pathway were dramatically inhibited whereas the expression of peroxisome proliferator-activated receptor gamma (PPAR-γ) was enhanced. This finding could provide good guidance for treating AIA. Additionally, Rg1 can alleviate the threat of type 2 PRRSV in piglets. Rg1 decreased the productions of cytokines (TNF-α, IL-1β, IL-6 and IL-8) and the activations of NF-κB pathway in Marc-145 cells and porcine alveolar macrophages (PAMs) infected by PRRSV. Rg1 not only relieved lung injury and virus in serum and tissues, but it also promoted the survival rate of piglets [80]. The rare ginsenoside Rg6 was also found to have immunosuppressive functions [81]. As an inhibitor of the inflammatory response of Toll-like receptor-4, Rg6 relieved septic shock in C57BL/6 mice induced by LPS and sepsis induced by CLP. Rg6 suppressed the production of TNF-α, IL-1β, IL-6 and IL-12p40 and augmented the expression of IL-10, and these results were confirmed in bone marrow-derived macrophages (BMDMs) through down-regulating NF-κB and MAPKs signaling pathways. Moreover, Rg6 triggered miR-146a, which is an operator miRNA in BMDMs with anti-inflammation functions. Hence, Rg6 elicited an immunosuppressive role through inhibiting NF-κB and MAPKs signaling pathways and stimulating IL-10 and miR-146a.

2.2.2. The immunosuppressive mechanisms and applications of PPD-type ginsenosides

The 6″-O-acetylginsenoside Rb1 isolated from North American ginseng by Samimi et al. [82] showed immunosuppressive activity in LPS-induced macrophage cells. Gemcitabine (GEM) is a chemotherapeutic drug used to treat NSCLC. GEM treatment increased the expression of pentraxin (PTX)-3 in lung cancer cells. PTX-3 is an gene related to pro-inflammation and is extremely highly expressed in lung cancer cells [83]. Yan et al. discovered that Rg3 can attenuate PTX-3 induced by GEM by down-regulating AKT, ERK and NF-κB/hypoxia-inducible factor 1-alpha (HIF-1α) pathways [84]. Rg3 relieves rheumatoid arthritis (RA) in mice. C57BL/6 mice administrated with CFA exhibited severe inflammation (Fig. 3B). The highly expressed pro-inflammatory cytokines TNF-α and IL-6 induced by CFA were suppressed. Moreover, the anti-inflammatory cytokines IL-10 and TGF-β, which were inhibited by CFA, were reversed after Rg3 treatment. CD4+CD25+Foxp3+Treg cells were enhanced as well, which showed that Rg3 has immunosuppressive and immune tolerance effects in RA mice [85].

In some patients who receive cardiac transplantation, acute cardiac allograft rejection can occur. Acute rejection can stimulate cellular immunity, which leads to the failure of transplantation. CK, a potent immunosuppressive adjuvant, was tested by Wang et al. [86] for its ability to inhibit acute rejection in both C57BL/6 and BALB/c mice. The results showed that CK decreased the proliferation of lymphocytes. Moreover, CK dramatically inhibited the production of IL-2 and IFN-γ but not IL-10. Islet transplantation was studied by Ma et al. [87] to assess the immunosuppressive role of CK in streptozotocin (STZ)-induced diabetic mice. In addition to the suppression of the same cytokines, CK not only decreased the occupancy of CD4+ and CD8+ T cells in the lymphatic system, but it also enhanced the expressions of TGF-β and Foxp3, which triggered the generation of Tregs. Allo-skin transplantation was conducted by Wang et al. [88] to examine the suppression effect of Rd on skin allograft rejection. Rd exhibited a similar characteristic with CK: Rd also inhibited TNF-α, IL-12 and up-regulated IL-10, which was thought to treat Th1-related transplant rejection.

3. Discussion

The coronavirus disease-19, caused by a globally rampant virus called COVID-19, represents an ongoing threat to human health. The global pandemic has caused thousands of deaths, with impacts on the global economy, resulting in a severe recession. Researchers around the world have developed several approaches to defend against the virus. Pharmacologists found that favipiravir [89] and Liahua Qingwen capsule [90] could be used to treat the COVID-19. Other studies have explored the use of ginseng on treating or preventing COVID-19 [91].

Ginseng is one of the ancient herbs in the world, as it has been used as medicine or a food ingredient for over 4000 years. However, with ongoing research, novel compounds of ginseng continue to be identified and isolated. In addition, both former and fresh compositions exhibited the infinite potentials in pharmacology, such as anti-inflammation, immunoregulatory, and anti-cancer activities. The saponins of ginseng are called ginsenosides, which exhibit many functions. Ginsenosides are classified into least four types according to their backbones. In this review, we mainly focused the immunomodulatory roles of ginsenosides in vitro and in vivo. It is worth noting that some ginsenosides are versatile. Many ginseng-derived saponins have both immunostimulatory and immunosuppressive functions, such as Rg1 in PPT-type ginsenosides as well as Rg3 and compound K in PPD-type ginsenosides. In vitro, Rg1 can stimulate innate immunity on macrophages induced by LPS and enhance the Th2 immune response through PI3K/AKT and mTOR signaling pathways. However, Rg1 down-regulates Th1 immunity by suppressing the NF-κB pathway. In vivo, Rg1 acts as an adjuvant in many vaccines like HBsAg, eSAG1 antigen or the PRRSV vaccine. In this case, Rg1 can promote the immune responses through both Th1 and Th2. However, the mechanisms in vivo are extremely complex. Rg1 was also used to treat CLP-induced sepsis mice or AIA rats through relieving Th1 and Th2 immunities. Rg1 even decreased the Th1 response in type 2 RPPSV-induced piglets. Rg3 and CK showed similar properties as Rg1. In addition, CK is applied to treat transplant rejection including cardiac, islet and skin transplantation. All these studies proved that ginsenosides perform different immune responses according to the chemicals or pathogens. COVID-19 impairs innate immunity and causes pneumonia. Ginseng has been proven to have anti-inflammation and anti-bacteria or -virus effects and boost human immunity. Some researchers investigated whether ginseng could alleviate respiration disease or co-infection caused by COVID-19 [92]. Park et al. used PEGylated nanoparticle albumin-bound (PNAB) ginsenosides, PNAB-RG6 and PNAB-Rgx365 and showed that PNAB-steroidal ginsenosides alleviated the inflammation and cytokine storm in an animal model, as well as remarkably reduced the severe conditions of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in ICU patients [92]. Rg3 was also found to efficiently block the entry of pseudovirus [93].

The discussion of immunity usually involves inflammation. Immunity is classified into two categories: the nonspecific and specific reactions. The nonspecific reaction, which is called innate immunity, includes barriers or eliminators that clear the invader without specificity. The specific reaction, named adaptive immunity, removes the pathogens through a precisely targeted approach. Inflammation serves as an alarm to indicate the infection [94,95]. Although some inflammation is not detrimental, exploring whether something can boost immunity without inflammation is important. Several immunostimulants were discovered and applied into clinical trials. For example, bropirimine is an immunostimulant that was clinically used to treat the transitional bladder [96]. However, bropirimine can easily induce headache, fever, and vomiting. In some conditions of transplantation or cancer, immunosuppressants, like corticosteroids, are required. However, corticosteroids also have some side effects, such as hoarse voice and agitation [97]. Therefore, ginseng has attracted more and more attention because of its mild and tonic properties. The immunomodulatory functions of ginsenoside, the major component of ginseng, are gradually becoming more well understood. However, although some papers reported that ginsenosides have immunostimulatory and immunosuppressive functions, the mechanisms of each ginsenoside remain unclear. Numerous papers showed that ginsenosides can trigger the Th1 and Th2 immune responses but did not conduct further investigation on the underlying mechanism. On the other hand, no clear mechanism of ginsenosides in human immune system is also a big possibility for investigators because the immunological studies on ginsenosides are vast uncultivated lands. Korea and China are the top two countries in ginseng production [98]. Further investigation into the mechanisms of ginsenosides in immunology will bring tremendous benefits in not only improving public health but also boosting individual immunity.

Data availability statement

Datasets related to this article can be found at https://www.ncbi.nlm.nih.gov/pubmed, hosted at the U.S. National Institutes of Health's National Library of Medicine (NIH/NLM).

Declaration of competing interest

The authors declare that no conflicts of interest exist.

Acknowledgments

This research was funded by the Basic Science Research Program through the National Research Foundation of Korea (NRF), the Ministry of Science and ICT, Republic of Korea (2017R1A6A1A03015642).

Contributor Information

Mi-Yeon Kim, Email: kimmy@ssu.ac.kr.

Jae Youl Cho, Email: jaecho@skku.edu.

References

  • 1.Medzhitov R., Janeway C.J. Advances in immunology: innate immunity. N Engl J Med. 2000;343:338–344. doi: 10.1056/NEJM200008033430506. [DOI] [PubMed] [Google Scholar]
  • 2.Černý J., Stříž I. Adaptive innate immunity or innate adaptive immunity? Clin Sci. 2019;133:1549–1565. doi: 10.1042/CS20180548. [DOI] [PubMed] [Google Scholar]
  • 3.Iwasaki A., Medzhitov R. Control of adaptive immunity by the innate immune system. Nat Immunol. 2015;16:343–353. doi: 10.1038/ni.3123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Medzhitov R., Janeway C.A. Innate immunity: impact on the adaptive immune response. Curr Opin Immunol. 1997;9:4–9. doi: 10.1016/s0952-7915(97)80152-5. [DOI] [PubMed] [Google Scholar]
  • 5.Bonilla F.A., Oettgen H.C. Adaptive immunity. J Allergy Clin Immunol. 2010;125:S33–S40. doi: 10.1016/j.jaci.2009.09.017. [DOI] [PubMed] [Google Scholar]
  • 6.Bascones-Martinez A., Mattila R., Gomez-Font R., Meurman J.H. Immunomodulatory drugs: oral and systemic adverse effects. Med Oral Patol Oral Cir Bucal. 2014;19:e24–e31. doi: 10.4317/medoral.19087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Melero I., Hervas-Stubbs S., Glennie M., Pardoll D.M., Chen L. Immunostimulatory monoclonal antibodies for cancer therapy. Nat Rev Cancer. 2007;7:95–106. doi: 10.1038/nrc2051. [DOI] [PubMed] [Google Scholar]
  • 8.Paulsen B. Plant polysaccharides with immunostimulatory activities. Curr Org Chem. 2005;5:939–950. [Google Scholar]
  • 9.Shahbazi S., Bolhassani A. Immunostimulants: types and functions. J Med Microbiol Infect Dis. 2016;4:45–51. [Google Scholar]
  • 10.Alexander C., Rietschel E.T. Bacterial lipopolysaccharides and innate immunity. J Endotoxin Res. 2001;7:167–202. [PubMed] [Google Scholar]
  • 11.Erridge C., Bennett-Guerrero E., Poxton I.R. Structure and function of lipopolysaccharides. Microb Infect. 2002;4:837–851. doi: 10.1016/s1286-4579(02)01604-0. [DOI] [PubMed] [Google Scholar]
  • 12.Silipo A., De Castro C., Lanzetta R., Parrilli M., Molinaro A. Springer; 2010. Lipopolysaccharides. Prokaryotic cell wall Compd; pp. 133–153. [Google Scholar]
  • 13.Fireman M., DiMartini A.F., Armstrong S.C., Cozza K.L. Immunosuppressants. Psychosomatics. 2004;45:354–360. doi: 10.1176/appi.psy.45.4.354. [DOI] [PubMed] [Google Scholar]
  • 14.Suthanthiran M., Morris R.E., Strom T.B. Immunosuppressants: cellular and molecular mechanisms of action. Am J Kidney Dis. 1996;28:159–172. doi: 10.1016/s0272-6386(96)90297-8. [DOI] [PubMed] [Google Scholar]
  • 15.Krensky M.A., Bennett M.W., Vincenti F. Immunosuppressants, tolerogens and immunostimulants. Brunt LL. 2011;1005–31 [Google Scholar]
  • 16.Thompson E.B., Lippman M.E. Mechanism of action of glucocorticoids. Metabolism. 1974;23:159–202. doi: 10.1016/0026-0495(74)90113-9. [DOI] [PubMed] [Google Scholar]
  • 17.Barnes P.J. Anti-inflammatory actions of glucocorticoids: molecular mechanisms. Clin Sci. 1998;94:557–572. doi: 10.1042/cs0940557. [DOI] [PubMed] [Google Scholar]
  • 18.Saklatvala J. Glucocorticoids: do we know how they work? Arthritis Res Ther. 2002;4:146. doi: 10.1186/ar398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kurosawa S., Kato M. Anesthetics, immune cells, and immune responses. J Anesth. 2008;22:263–277. doi: 10.1007/s00540-008-0626-2. [DOI] [PubMed] [Google Scholar]
  • 20.Gajewski T.F., Schreiber H., Fu Y.-X. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 2013;14:1014–1022. doi: 10.1038/ni.2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Malech H.L., Gallin J.I. Neutrophils in human diseases. N Engl J Med. 1987;317:687–694. doi: 10.1056/NEJM198709103171107. [DOI] [PubMed] [Google Scholar]
  • 22.Witko-Sarsat V., Rieu P., Descamps-Latscha B., Lesavre P., Halbwachs-Mecarelli L. Neutrophils: molecules, functions and pathophysiological aspects. Lab Invest. 2000;80:617–654. doi: 10.1038/labinvest.3780067. [DOI] [PubMed] [Google Scholar]
  • 23.Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol. 2006;6:173–182. doi: 10.1038/nri1785. [DOI] [PubMed] [Google Scholar]
  • 24.Kobayashi S.D., Voyich J.M., Burlak C., DeLeo F.R. Neutrophils in the innate immune response. Arch Immunol Ther Exp. 2005;53:505–517. [PubMed] [Google Scholar]
  • 25.Geissmann F., Manz M.G., Jung S., Sieweke M.H., Merad M., Ley K. Development of monocytes, macrophages, and dendritic cells. Science. 2010;327:656–661. doi: 10.1126/science.1178331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cline M.J., Lehrer R.I., Territo M.C., Golde D.W. Monocytes and macrophages: functions and diseases. Ann Intern Med. 1978;88:78–88. doi: 10.7326/0003-4819-88-1-78. [DOI] [PubMed] [Google Scholar]
  • 27.Franken L., Schiwon M., Kurts C. Macrophages: sentinels and regulators of the immune system. Cell Microbiol. 2016;18:475–487. doi: 10.1111/cmi.12580. [DOI] [PubMed] [Google Scholar]
  • 28.LaRosa D.F., Orange J.S. 1. Lymphocytes. J Allergy Clin Immunol. 2008;121:S364–S369. doi: 10.1016/j.jaci.2007.06.016. [DOI] [PubMed] [Google Scholar]
  • 29.Hauser A.E., Höpken U.E. In: Reth MBT-MB of BC (second E. second ed. Alt F.W., Honjo T., Radbruch A., editors. Academic Press; London: 2015. B cell localization and migration in health and disease; pp. 187–214. (Mol. Biol. B cells). [Google Scholar]
  • 30.Zhu J., Paul W.E. CD4 T cells: fates, functions, and faults. Blood. 2008;112:1557–1569. doi: 10.1182/blood-2008-05-078154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Parnham M.J., Nijkamp F.P., Rossi A.G. Springer Nature; 2019. Nijkamp and Parnham's principles of immunopharmacology. [Google Scholar]
  • 32.Narsale A., Moya R., Davies J.D. Human CD4+CD25+CD127hi cells and the Th1/Th2 phenotype. Clin Immunol. 2018;188:103–112. doi: 10.1016/j.clim.2018.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Allman D., Pillai S. Peripheral B cell subsets. Curr Opin Immunol. 2008;20:149–157. doi: 10.1016/j.coi.2008.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Vivier E., Tomasello E., Baratin M., Walzer T., Ugolini S. Functions of natural killer cells. Nat Immunol. 2008;9:503–510. doi: 10.1038/ni1582. [DOI] [PubMed] [Google Scholar]
  • 35.Lv M., Zhao X., Chen F., Yu M., Li C., Sun J. A rapid white blood cell classification system based on multimode imaging technology. J Biophot. 2020;13:1–10. doi: 10.1002/jbio.202000197. [DOI] [PubMed] [Google Scholar]
  • 36.Swartz M.A. The physiology of the lymphatic system. Adv Drug Deliv Rev. 2001;50:3–20. doi: 10.1016/s0169-409x(01)00150-8. [DOI] [PubMed] [Google Scholar]
  • 37.Boes K.M., Durham A.C. Bone marrow, blood cells, and the lymphoid/lymphatic system. Pathol Basis Vet Dis. 2017:724–804. [Google Scholar]
  • 38.Yun T.K., Choi S.Y. Non-organ specific cancer prevention of ginseng: a prospective study in Korea. Int J Epidemiol. 1998;27:359–364. doi: 10.1093/ije/27.3.359. [DOI] [PubMed] [Google Scholar]
  • 39.Zhao Y. Springer; 2015. Panax notoginseng (burk.) F.H. Chen sanqi (sanqi, notoginseng). Diet. Chinese herbs; pp. 185–193. [Google Scholar]
  • 40.Lee S.M., Bae B.S., Park H.W., Ahn N.G., Cho B.G., Cho Y.L., et al. Characterization of Korean red ginseng (Panax ginseng Meyer): history, preparation method, and chemical composition. J Ginseng Res. 2015;39:384–391. doi: 10.1016/j.jgr.2015.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.You L., Cho J.Y. The regulatory role of Korean ginseng in skin cells. J Ginseng Res. 2021;45:363–370. doi: 10.1016/j.jgr.2020.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chang-Xiao L., Pei-Gen X. Recent advances on ginseng research in China. J Ethnopharmacol. 1992;36:27–38. doi: 10.1016/0378-8741(92)90057-x. [DOI] [PubMed] [Google Scholar]
  • 43.Kim J.H., Yi Y.S., Kim M.Y., Cho J.Y. Role of ginsenosides, the main active components of Panax ginseng, in inflammatory responses and diseases. J Ginseng Res. 2017;41:435–443. doi: 10.1016/j.jgr.2016.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Yu H., Zhang C., Lu M., Sun F., Fu Y., Jin F. Purification and characterization of new special ginsenosidase hydrolyzing multi-glycisides of protopanaxadiol ginsenosides, ginsenosidase type I. Chem Pharm Bull. 2007;55:231–235. doi: 10.1248/cpb.55.231. [DOI] [PubMed] [Google Scholar]
  • 45.Fuzzati N. Analysis methods of ginsenosides. J Chromatogr B Anal Technol Biomed Life Sci. 2004;812:119–133. doi: 10.1016/j.jchromb.2004.07.039. [DOI] [PubMed] [Google Scholar]
  • 46.Yu J., Niu Y., Li R., Liao J., Zhang F., Yu B. Synthetic access toward the diverse ginsenosides. Chem Sci. 2013;4:3899–3905. [Google Scholar]
  • 47.Ma X., Xiao H., Liang X. Identification of ginsenosides in Panax quinquefolium by LC-MS. Chromatographia. 2006;64:31–36. [Google Scholar]
  • 48.Romagnani S. T-cell subsets (Th1 versus Th2) Ann Allergy Asthma Immunol. 2000;85:9–18. doi: 10.1016/S1081-1206(10)62426-X. [DOI] [PubMed] [Google Scholar]
  • 49.Berger A. Science commentary: Th1 and Th2 responses: what are they? Br Med J. 2000;321:424. doi: 10.1136/bmj.321.7258.424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Shin M.S., Song J.H., Choi P., Lee J.H., Kim S.Y., Shin K.S., et al. Stimulation of innate immune function by Panax ginseng after heat processing. J Agric Food Chem. 2018;66:4652–4659. doi: 10.1021/acs.jafc.8b00152. [DOI] [PubMed] [Google Scholar]
  • 51.Xin C., Kim J., Quan H., Yin M., Jeong S., Choi J Il, et al. Ginsenoside Rg3 promotes Fc gamma receptor-mediated phagocytosis of bacteria by macrophages via an extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase-dependent mechanism. Int Immunopharm. 2019;77:105945. doi: 10.1016/j.intimp.2019.105945. [DOI] [PubMed] [Google Scholar]
  • 52.Kim J.H., Doo E.H., Jeong M., Kim S., Lee Y.Y., Yang J., et al. Enhancing immunomodulatory function of red ginseng through fermentation using Bifidobacterium animalis subsp. Lactis LT 19-2. Nutrients. 2019:11071481. doi: 10.3390/nu11071481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kim M.Y., Cho J.Y. 20S-dihydroprotopanaxadiol, a ginsenoside derivative, boosts innate immune responses of monocytes and macrophages. J Ginseng Res. 2013;37:293–299. doi: 10.5142/jgr.2013.37.293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Yang W.S., Yi Y.S., Kim D., Kim M.H., Park J.G., Kim E., et al. Nuclear factor kappa-B- and activator protein-1-mediated immunostimulatory activity of compound K in monocytes and macrophages. J Ginseng Res. 2017;41:298–306. doi: 10.1016/j.jgr.2016.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wang Y., Liu Y., Zhang X.Y., Xu L.H., Ouyang D.Y., Liu K.P., et al. Ginsenoside Rg1 regulates innate immune responses in macrophages through differentially modulating the NF-κB and PI3K/Akt/mTOR pathways. Int Immunopharm. 2014;23:77–84. doi: 10.1016/j.intimp.2014.07.028. [DOI] [PubMed] [Google Scholar]
  • 56.Lee E.J., Ko E., Lee J., Rho S., Ko S., Shin M.K., et al. Ginsenoside Rg1 enhances CD4+ T-cell activities and modulates Th1/Th2 differentiation. Int Immunopharm. 2004;4:235–244. doi: 10.1016/j.intimp.2003.12.007. [DOI] [PubMed] [Google Scholar]
  • 57.Sun J., Hu S., Song X. Adjuvant effects of protopanaxadiol and protopanaxatriol saponins from ginseng roots on the immune responses to ovalbumin in mice. Vaccine. 2007;25:1114–1120. doi: 10.1016/j.vaccine.2006.09.054. [DOI] [PubMed] [Google Scholar]
  • 58.Sun J., Song X., Hu S. Ginsenoside Rg1 and aluminum hydroxide synergistically promote immune responses to ovalbumin in BALB/c mice. Clin Vaccine Immunol. 2008;15:303–307. doi: 10.1128/CVI.00448-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Yuan D., Yuan Q., Cui Q., Liu C., Zhou Z., Zhao H., et al. Vaccine adjuvant ginsenoside Rg1 enhances immune responses against hepatitis B surface antigen in mice. Can J Physiol Pharmacol. 2016;94:676–681. doi: 10.1139/cjpp-2015-0528. [DOI] [PubMed] [Google Scholar]
  • 60.Qu D.F., Yu H.J., Liu Z., Zhang D.F., Zhou Q.J., Zhang H.L., et al. Ginsenoside Rg1 enhances immune response induced by recombinant Toxoplasma gondii SAG1 antigen. Vet Parasitol. 2011;179:28–34. doi: 10.1016/j.vetpar.2011.02.008. [DOI] [PubMed] [Google Scholar]
  • 61.Su F., Wu Y., Li J., Huang Y., Yu B., Xu L., et al. Escherichia coli heat-labile enterotoxin B subunit combined with ginsenoside Rg1 as an intranasal adjuvant triggers type I interferon signaling pathway and enhances adaptive immune responses to an inactivated PRRSV vaccine in ICR mice. Vaccines. 2021:9030266. doi: 10.3390/vaccines9030266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bi S., Chi X., Zhang Y., Ma X., Liang S., Wang Y., et al. Ginsenoside Rg1 enhanced immune responses to infectious bursal disease vaccine in chickens with oxidative stress induced by cyclophosphamide. Poultry Sci. 2018;97:2698–2706. doi: 10.3382/ps/pey132. [DOI] [PubMed] [Google Scholar]
  • 63.Sun H.X., Chen Y., Ye Y. Ginsenoside Re and notoginsenoside R1: immunologic adjuvants with low haemolytic effect. Chem Biodivers. 2006;3:718–726. doi: 10.1002/cbdv.200690074. [DOI] [PubMed] [Google Scholar]
  • 64.Yang Z.G., Ye Y.P., Sun H.X. Immunological adjuvant effect of ginsenoside Rh4 from the roots of Panax notoginseng on specific antibody and cellular response to ovalbumin in mice. Chem Biodivers. 2007;4:232–240. doi: 10.1002/cbdv.200790028. [DOI] [PubMed] [Google Scholar]
  • 65.Song X., Chen J., Sakwiwatkul K., Li R., Hu S. Enhancement of immune responses to influenza vaccine (H3N2) by ginsenoside Re. Int Immunopharm. 2010;10:351–356. doi: 10.1016/j.intimp.2009.12.009. [DOI] [PubMed] [Google Scholar]
  • 66.Qu D., Han J., Du A. Enhancement of protective immune response to recombinant Toxoplasma gondii ROP18 antigen by ginsenoside Re. Exp Parasitol. 2013;135:234–239. doi: 10.1016/j.exppara.2013.07.013. [DOI] [PubMed] [Google Scholar]
  • 67.Su X., Pei Z., Hu S. Ginsenoside Re as an adjuvant to enhance the immune response to the inactivated rabies virus vaccine in mice. Int Immunopharm. 2014;20:283–289. doi: 10.1016/j.intimp.2014.03.008. [DOI] [PubMed] [Google Scholar]
  • 68.Wu R., Ru Q., Chen L., Ma B., Li C. Stereospecificity of ginsenoside Rg3 in the promotion of cellular immunity in hepatoma H22-bearing mice. J Food Sci. 2014;79:H1430–H1435. doi: 10.1111/1750-3841.12518. [DOI] [PubMed] [Google Scholar]
  • 69.Liu X., Zhang Z., Liu J., Wang Y., Zhou Q., Wang S., et al. Ginsenoside Rg3 improves cyclophosphamide-induced immunocompetence in Balb/c mice. Int Immunopharm. 2019;72:98–111. doi: 10.1016/j.intimp.2019.04.003. [DOI] [PubMed] [Google Scholar]
  • 70.Qian Y., Huang R., Li S., Xie R., Qian B., Zhang Z., et al. Ginsenoside Rh2 reverses cyclophosphamide-induced immune deficiency by regulating fatty acid metabolism. J Leukoc Biol. 2019;106:1089–1100. doi: 10.1002/JLB.2A0419-117R. [DOI] [PubMed] [Google Scholar]
  • 71.Wang M., Yan S.J., Zhang H.T., Li N., Liu T., Zhang Y.L., et al. Ginsenoside Rh2 enhances the antitumor immunological response of a melanoma mice model. Oncol Lett. 2017;13:681–685. doi: 10.3892/ol.2016.5490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wei X., Chen J., Su F., Su X., Hu T., Hu S. Stereospecificity of ginsenoside Rg3 in promotion of the immune response to ovalbumin in mice. Int Immunol. 2012;24:465–471. doi: 10.1093/intimm/dxs043. [DOI] [PubMed] [Google Scholar]
  • 73.Song X., Zang L., Hu S. Amplified immune response by ginsenoside-based nanoparticles (ginsomes) Vaccine. 2009;27:2306–2311. doi: 10.1016/j.vaccine.2009.02.040. [DOI] [PubMed] [Google Scholar]
  • 74.Yang Z., Chen A., Sun H., Ye Y., Fang W. Ginsenoside Rd elicits Th1 and Th2 immune responses to ovalbumin in mice. Vaccine. 2007;25:161–169. doi: 10.1016/j.vaccine.2006.05.075. [DOI] [PubMed] [Google Scholar]
  • 75.Han Y., Rhew K.Y. Ginsenoside Rd induces protective anti-Candida albicans antibody through immunological adjuvant activity. Int Immunopharm. 2013;17:651–657. doi: 10.1016/j.intimp.2013.08.003. [DOI] [PubMed] [Google Scholar]
  • 76.Rivera E., Pettersson F.E., Inganäs M., Paulie S., Grönvik K.O. The Rb1 fraction of ginseng elicits a balanced Th1 and Th2 immune response. Vaccine. 2005;23:5411–5419. doi: 10.1016/j.vaccine.2005.04.007. [DOI] [PubMed] [Google Scholar]
  • 77.Kang N., Gao H., He L., Liu Y., Fan H., Xu Q., et al. Ginsenoside Rb1 is an immune-stimulatory agent with antiviral activity against enterovirus 71. J Ethnopharmacol. 2021;266:113401. doi: 10.1016/j.jep.2020.113401. [DOI] [PubMed] [Google Scholar]
  • 78.Zou Y., Tao T., Tian Y., Zhu J., Cao L., Deng X., et al. Ginsenoside Rg1 improves survival in a murine model of polymicrobial sepsis by suppressing the inflammatory response and apoptosis of lymphocytes. J Surg Res. 2013;183:760–766. doi: 10.1016/j.jss.2013.01.068. [DOI] [PubMed] [Google Scholar]
  • 79.Zhang L., Zhu M., Li M., Du Y., Duan S., Huang Y., et al. Ginsenoside Rg1 attenuates adjuvant-induced arthritis in rats via modulation of PPAR-γ/NF-κB signal pathway. Oncotarget. 2017;8:55384–55393. doi: 10.18632/oncotarget.19526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Yu Z.Q., Yi H.Y., Ma J., Wei Y.F., Cai Meng-Kai, Li Q., et al. Ginsenoside Rg1 suppresses type 2 prrsv infection via NF-κB signaling pathway in vitro, and provides partial protection against HP-PRRSV in piglet. Viruses. 2019:1045. doi: 10.3390/v11111045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Paik S., Choe J.H., Choi G.E., Kim J.E., Kim J.M., Song G.Y., et al. Rg6, a rare ginsenoside, inhibits systemic inflammation through the induction of interleukin-10 and microRNA-146a. Sci Rep. 2019;9:4342. doi: 10.1038/s41598-019-40690-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Samimi R., Xu W.Z., Lui E.M.K., Charpentier P.A. Isolation and immunosuppressive effects of 6″- O-acetylginsenoside Rb1 extracted from north american ginseng. Planta Med. 2014;80:509–516. doi: 10.1055/s-0034-1368319. [DOI] [PubMed] [Google Scholar]
  • 83.Infante M., Allavena P., Garlanda C., Nebuloni M., Morenghi E., Rahal D., et al. Prognostic and diagnostic potential of local and circulating levels of pentraxin 3 in lung cancer patients. Int J Cancer. 2016;138:983–991. doi: 10.1002/ijc.29822. [DOI] [PubMed] [Google Scholar]
  • 84.Ahmmed B., Kampo S., Khan M., Faqeer A., kumar S.P., Yulin L., et al. Rg3 inhibits gemcitabine-induced lung cancer cell invasiveness through ROS-dependent, NF-κB- and HIF-1α-mediated downregulation of PTX3. J Cell Physiol. 2019;234:10680–10697. doi: 10.1002/jcp.27731. [DOI] [PubMed] [Google Scholar]
  • 85.Zhang Y., Wang S., Song S., Yang X., Jin G. Ginsenoside Rg3 alleviates complete Freund's adjuvant-induced rheumatoid arthritis in mce by regulating CD4+CD25+Foxp3+Treg cells. J Agric Food Chem. 2020;68:4893–4902. doi: 10.1021/acs.jafc.0c01473. [DOI] [PubMed] [Google Scholar]
  • 86.Wang Y., Qin Q., Chen J., Kuang X., Xia J., Xie B., et al. Synergistic effects of Isatis tinctoria L. and tacrolimus in the prevention of acute heart rejection in mice. Transpl Immunol. 2009;22:5–11. doi: 10.1016/j.trim.2009.09.004. [DOI] [PubMed] [Google Scholar]
  • 87.Ma P.F., Gao C., Cheng P.P., Li J.L., Huang X., Lin Y.Y., et al. Immunosuppressive effect of compound K on islet transplantation in an STZ-induced diabetic mouse model. Diabetes. 2014;63:3458–3469. doi: 10.2337/db14-0012. [DOI] [PubMed] [Google Scholar]
  • 88.Wang L., Zhang Y., Chen J., Li S., Wang Y., Hu L., et al. Immunosuppressive effects of ginsenoside-Rd on skin allograft rejection in rats. J Surg Res. 2012;176:267–274. doi: 10.1016/j.jss.2011.06.038. [DOI] [PubMed] [Google Scholar]
  • 89.Yamamura H., Matsuura H., Nakagawa J., Fukuoka H., Domi H., Chujoh S. Effect of favipiravir and an anti-inflammatory strategy for COVID-19. Crit Care. 2020;24:413. doi: 10.1186/s13054-020-03137-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Xia Q.-D., Xun Y., Lu J.-L., Lu Y.-C., Yang Y.-Y., Zhou P., et al. Network pharmacology and molecular docking analyses on Lianhua Qingwen capsule indicate Akt1 is a potential target to treat and prevent COVID-19. Cell Prolif. 2020;53 doi: 10.1111/cpr.12949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Zhang J.L., Li W.X., Li Y., Wong M.S., Wang Y.J., Zhang Y. Therapeutic options of TCM for organ injuries associated with COVID-19 and the underlying mechanism. Phytomedicine. 2021;85:153297. doi: 10.1016/j.phymed.2020.153297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Lee W.S., Rhee D.K. Corona-Cov-2 (COVID-19) and ginseng: comparison of possible use in COVID-19 and influenza. J Ginseng Res. 2021;45:535–537. doi: 10.1016/j.jgr.2020.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Zhang D., Hamdoun S., Chen R., Yang L., Ip C.K., Qu Y., et al. Identification of natural compounds as SARS-CoV-2 entry inhibitors by molecular docking-based virtual screening with bio-layer interferometry. Pharmacol Res. 2021:105820. doi: 10.1016/j.phrs.2021.105820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Xu W., Larbi A. Immunity and inflammation: from jekyll to hyde. Exp Gerontol. 2018;107:98–101. doi: 10.1016/j.exger.2017.11.018. [DOI] [PubMed] [Google Scholar]
  • 95.Xiao T.S. Innate immunity and inflammation. Cell Mol Immunol. 2017;14:1–3. doi: 10.1038/cmi.2016.45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Akaza H., Kotake T., Machida T. Bropirimine, an orally active anticacner agent for superficial bladder cancer. Eur Urol. 1998;34:107–110. doi: 10.1159/000019693. [DOI] [PubMed] [Google Scholar]
  • 97.Trevillian P. Immunosuppressants - clinical applications. Aust Prescr. 2006;29:102–108. [Google Scholar]
  • 98.Baeg I.H., So S.H. The world ginseng market and the ginseng (Korea) J Ginseng Res. 2013;37:1–7. doi: 10.5142/jgr.2013.37.1. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Datasets related to this article can be found at https://www.ncbi.nlm.nih.gov/pubmed, hosted at the U.S. National Institutes of Health's National Library of Medicine (NIH/NLM).


Articles from Journal of Ginseng Research are provided here courtesy of Elsevier

RESOURCES