ArticlePDF Available

Ginseng, the 'Immunity Boost': The Effects of Panax ginseng on Immune System

Authors:

Abstract and Figures

Thousands of literatures have described the diverse role of ginseng in physiological processes such as cancer, neurodegenerative disorders, insulin resistance, and hypertension. In particular, ginseng has been extensively reported to maintain homeostasis of the immune system and to enhance resistance to illness or microbial attacks through the regulation of immune system. Immune system comprises of different types of cells fulfilling their own specialized functions, and each type of the immune cells is differentially influenced and may be simultaneously controlled by ginseng treatment. This review summarizes the current knowledge on the effects of ginseng on immune system. We discuss how ginseng regulates each type of immune cells including macrophages, natural killer cells, dendritic cells, T cells, and B cells. We also describe how ginseng exhibits beneficial effects on controlling inflammatory diseases and microbial infections.
Content may be subject to copyright.
The Korean Society of Ginseng
354
http://ginsengres.org pISSN: 1226-8453 eISSN: 2093-4947
Review Article
J Ginseng Res Vol. 36, No. 4, 354-368 (2012)
http://dx.doi.org/10.5142/jgr.2012.36.4.354
E-mail: hymin@cau.ac.kr
Tel: +82-2-820-5618, Fax: +82-2-2-816-7338
*
Corresponding author
INTRODUCTION
Ginseng (the root of Panax ginseng Meyer, Family
Araliaceae), one of the most well-known oriental medici-
nal herbs, has been widely used as an herbal remedy for
various disorders [1]. Natural-dried ginseng is known as
white ginseng, and red ginseng is prepared by steaming
fresh ginseng root prior to drying on the purpose of en-
hancing ef cacy, safety, and preservation [2].
Ginseng contains various pharmacological compo-
nents including a series of tetracyclic triterpenoid sapo-
nins (ginsenosides), polyacetylenes, polyphenolic com-
pounds, and acidic polysaccharides [3]. Ginsenosides
can be classi ed into three groups based on the chemical
structure of aglycones: the protopanaxadiol group (e.g.,
Rb1, Rb2, Rb3, Rc, and Rd); the protopanaxatriol group
(e.g., Re, Rf, Rg1, and Rg2); and the oleanane group (e.g.,
Ro) (Fig. 1) [2,4]. Among these components, ginsen-
osides Rh2, Rs4, and Rg3 are found only in red ginseng
Ginseng, the 'Immunity Boost': The E ects of Panax ginseng on Immune
System
Soowon Kang and Hyeyoung Min*
College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
Thousands of literatures have described the diverse role of ginseng in physiological processes such as cancer, neurodegenera tive
disorders, insulin resistance, and hypertension. In particular, ginseng has been extensively reported to maintain homeostasis of the
immune system and to enhance resistance to illness or microbial attacks through the regulation of immune system. Immune system
comprises of different types of cells ful lling their own specialized functions, and each type of the immune cells is differentially
in uenced and may be simultaneously controlled by ginseng treatment. This review summarizes the current knowledge on the
effects of ginseng on immune system. We discuss how ginseng regulates each type of immune cells including macrophages,
natural killer cells, dendritic cells, T cells, and B cells. We also describe how ginseng exhibits bene cial effects on controlling
in ammatory diseases and microbial infections.
Keywords: Panax ginseng, Innate immunity, Acquired immunity, Immunomodulation, Cytokine
This is an Open Access article distributed under the terms of the Cre-
ative Commons Attribution Non-Commercial License (http://creativecom-
mons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial
use, distribution, and reproduction in any medium, provided the original
work is properly cited.
Received 14 Jun. 2012, Revised 19 Jul. 2012, Accepted 19 Jul. 2012
and are derived from the hydrolysis of saponins by heat-
processing [5]. Fermentation of red ginseng by intestinal
microorganisms transforms saponins into readily absorb-
able forms such as compound K (20-O-D-glucopyrano-
syl-20[S]-protopanaxadiol) [6].
Ginseng has been well known as an immune modula-
tor [7-10]. Roots (mostly), stems, leaves of ginseng, and
their extracts have been used for maintaining immune
homeostasis and enhancing resistance to illness or micro-
bial attacks through effects on immune system. Im mune
system is composed of diverse types of cells with their
own specialized functions, and each type of the immune
cells differentially responds to ginseng treatment. This
review summarizes the current knowledge on the effects
of ginseng on immune system. Among the hundreds
of literatures on immune responses, we only embraced
the reports which used mice, rats, or humans as in vivo
355
Kang et al. Ginseng and Immunity
http://ginsengres.org
model organisms, murine or human-derived cell lines as
in vitro experimental models, and P. ginseng as a source
of ginseng.
INNATE IMMUNITY
Innate immunity is the rst barrier of defense against
various infections by foreign organisms or substances
(antigens), and is composed of four principal components.
They comprise 1) physical/chemical barriers such as skin,
mucous membranes, and the acidity of the stomach, 2)
cellular components including phagocytes and natural
killer cells, 3) blood proteins such as complements and
in ammatory mediators, and 4) cytokines that regulating
the cells of innate immune system [11,12]. These com-
ponents of innate immunity recognize foreign microbes,
initiate rapid primary immune responses, and prevent
or eliminate infection of the host [13]. In addition to
defensive function, innate immune system also gener-
ates a signal alarming the presence of infection, and thus
stimulating a subsequent adaptive immune response [12].
Although an innate immunity comprises a variety of com-
ponents mediating diverse functions, we only assess the
effects of ginseng on cellular component in this review.
Macrophages
The mononuclear phagocytes are composed of mono-
cytes circulating in the blood and macrophages in the
tissues [14]. Macrophages exhibit different morphologic
forms in different tissues, and they include microglia of
the central nervous system, Kupffer cells in the liver, al-
veolar macrophages in the lung, and osteoclasts in bone
[14]. Macrophages are principal components of innate
defense against infections, and also play important roles
in adaptive immune responses [12]. When activated by
various stimuli, macrophages enhance phagocytic activ-
ity, increase the ability to kill ingested microbes, and
produce many cytokines and inflammatory mediators
such as nitric oxide and H2O2 [15]. They also express
high levels of class II major histocompatibility complex
(MHC) molecules to present antigens to helper T cells,
and thereby connect the innate and adaptive immune sys-
tems [12].
Through numerous experiments, it was con rmed that
ginseng extract enhanced phagocytic activity of mac-
rophages [16]. Macrophages treated with ginseng poly-
saccharides stimulated the phagocytosis of zymosan, a
preparation of cell wall from Saccharomyces cerevisiae,
and peritoneal macrophages also showed an increase in
Fig. 1. Structures of saponins from Panax ginseng. (A) Protopanaxadiol- and protopanaxatriol-type saponins, (B) ginsenoside Ro, and (C) com-
pound K. Glc, β-D-glucose; Ara(p), arabinose (pyranose form); Ara(f), arabinose (furanose form); Rha, α-L-rhamnose.
http://dx.doi.org/10.5142/jgr.2012.36.4.354
356
J Ginseng Res Vol. 36, No. 4, 354-368 (2012)
phagocytosis following treatment with red ginseng acidic
polysaccharides (RGAPs, ginsan) [16,17]. In a clinical
trial with 20 healthy adults, oral administration of gin-
seng extract improved phagocytic activity [8].
In addition to the effects on phagocytosis, ginseng
can stimulate the generation of in ammatory mediators
such as nitric oxide (NO), a reactive nitrogen intermedi-
ate, through up-regulation of inducible nitric oxide syn-
thase (iNOS) [18]. Macrophages produce NO in order
to destroy phagocytized microbes, and ginseng aqueous
extract and ginsan have shown to up-regulate NO pro-
duction in activated RAW 264.7 cells and primary peri-
toneal macrophages, respectively [16,18,19]. In addition,
treatment of macrophages with a combination of RGAPs
and IFN-γ has shown to potentiate the activation of mac-
rophages, and NO production is greatly improved [18].
Upon activation by microbial products such as lipo-
polysaccharide (LPS) and IFN-γ derived from natural
killer (NK) cells, macrophages secrete IL-1β and TNF-α
to induce in ammatory reactions, and IL-12 to stimulate
NK cells and T cells to produce IFN-γ [17,18]. Isolated
polysaccharides from P. ginseng augmented the secre-
tion of IL-1β and TNF-α from peritoneal macrophages in
vitro [17]. Treatment of macrophages with RGAPs and
IFN-γ signi cantly increased the production of IL-1β and
TNF-α as well as NO as previously described [18]. IL-
12 release was also boosted up by treatment of murine
J774A.1 macrophage cells with ginseng extract and its
isolated components [20].
Dendritic cells
Dendritic cells (DC) play important roles in innate im-
mune responses to infections and in linking innate and
adaptive immune responses [21]. DCs recognize antigens
at the rst sites of defense, migrate into lymphoid organs,
and present antigenic molecules to lymphocytes to initi-
ate lymphocytes activation [21]. In addition, activated
DCs release cytokines such as IL-12, IL-4, IFN-γ, and
TNF-α to stimulate adaptive as well as innate immune
responses [22].
In general, ginseng exhibits immunostimulatory ef-
fects on DCs [23,24]. Takei et al. [24] have reported that
M1 and M4, end products of steroidal ginseng saponins
metabolized in digestive tracts were able to drive the
maturation of DC from human monocytes manifested by
the increased expression of DC maturation markers such
as MHC class II, CD80, CD83, and CD86. These surface
molecules are associated with antigen-presenting capa-
bilities of DCs implying that the increased expression of
MHC class II and co-stimulatory molecules may enhance
the differentiation and proliferation of lymphocytes. In
fact, M4-primed mature DCs displayed enhanced T cells
stimulatory capacity as measured by T cell proliferation,
and the production of IFN-γ and 51Cr release on M4-
primed mature DCs were more augmented than those of
control DCs. Similarly, ginsan also markedly increased
the production of IL-1 and TNF-α as well as the expres-
sion of maturation markers such as MHC class II and
CD86 on DCs [23]. Subsequent experiment demonstrat-
ed that ginsan-treated DCs improved the proliferation of
allogeneic T cells, corroborating the constructive role of
ginseng on DC function.
However, some ginseng saponins have inhibitory effect
on immune maturation of DCs. According to Su et al.
[25], treatment of DCs with total saponins of P. ginseng
followed by oxidized-low density lipoprotein diminished
the secretion of cytokines such as IL-12 and TNF-α, and
the expression of maturation markers including CD40,
CD86, CD1a, and HLA-DR. In other experiments using
steamed ginseng-leaves and flowers, ginsenosides Rg6
and F4 exhibited inhibitory effects on bone marrow-
derived dendritic cells and decrease in the production
of IL-12 p40, a subunit component of IL-12 upon LPS
stimulation [26]. Although it remains unclear why gin-
seng showed such opposite effects, researchers suspect
that ginseng may cause pro-maturation or anti-maturation
of DCs and stimulate different signaling pathways [25].
Natural killer cells
NK cells are a type of cytotoxic lymphocytes of innate
immune system that are involved in the rst line of de-
fense against newly arising malignant cells and infected
cells [27]. Distinguished from cytotoxic T lymphocytes,
they can recognize target cells in the absence of antibod-
ies and MHC, and do not require activation to kill target
cells thereby enabling a fast immune response [27]. In
addition to killing target cells non-speci cally, NK cells
release pro-in ammatory cytokines such as IFN-γ which
activates macrophages to destroy phagocytized microbes
[27].
Many literatures have reported that total ginseng ex-
tract enhances NK cell functions. Oral administration of
aqueous extract of P. ginseng increased natural killing
activity in mice [7], and ginsenoside Rg1-treated mice
also showed an enhanced natural killing activity of the
splenocytes [28]. Interestingly, the stimulatory effects of
ginseng on NK cells were observed regardless of disease
condition of the host. Therefore, the functions of NK
cells stimulated by ginseng were comparable between
immunosuppressed mice and immunocompetent mice
357
Kang et al. Ginseng and Immunity
http://ginsengres.org
[29]. The trial using blood sample of human patients
who had chronic fatigue syndrome and acquired im-
munodeficiency syndrome (AIDS) showed functional
enhancement of NK cells isolated from peripheral blood
mononuclear cells [30]. In vivo test conducted by Sca-
glione et al. [8] also demonstrated that NK cell function
in individuals taking ginseng was promoted compared
to placebo group. Collectively, these results suggest that
ginseng may improve NK cell activity irrespective of
host immune conditions.
ACQUIRED IMMUNITY
Acquired (or adaptive) immunity is an antigen-speci c
immune response which occurs in response to the stimu-
lation by specific antigens [31]. The major players of
acquired immunity are lymphocytes such as T cells and
B cells and their secreted antibodies. B lymphocytes play
a role in the humoral immune response, whereas T lym-
phocytes are primarily involved in cell-mediated immune
response [31]. The key feature of the adaptive immune
response is its speci city and the ability to generate and
maintain immunological memory [32]. Upon antigenic
stimulation, antigen-speci c lymphocytes proliferate and
differentiate into effector cells whose function is to elimi-
nate the antigen, and into memory cells which possess
an enhanced response to subsequent encounters with the
same antigen [31,32].
Humoral immune response
The humoral immune response is an immunity medi-
ated by antibodies [33]. In this response, the immune
system triggers speci c B cells (mostly, plasma cells) to
proliferate and secrete large amounts of speci c antibod-
ies which then ght invading microorganisms or patho-
genic antigens [33]. Antibodies can bind to the antigens
such as toxin and virus, and keep them from contacting
and harming normal cells of the body [33,34]. In addi-
tion, antigens coated with antibodies can be easily rec-
ognized, phagocytized, and destroyed by phagocytes or
macrophages [35]. Antibodies are also able to stimulate
complement system to form membrane attack complex
on the membrane of invading microbes and kill them
[36].
Although numerous studies describe the role of gin-
seng on antibody production, most of them evaluate its
adjuvant potential on the secretion of speci c antibodies.
In general, ginseng extract successfully induced antigen-
speci c IgM, IgG, and IgA antibody responses when ad-
ministered orally or intraperitoneally [37,38]. When mice
were pre-treated with ginsan before oral-immunization
of Salmonella for 2 wk, higher amounts of serum IgG1
and IgG2 as well as secretory IgA against Salmonella
were produced [39]. In addition, subcutaneous injection
of ginsenoside Rg1 developed a high level of specific
antibody responses against Toxoplasma gondii and gin-
senoside Re also significantly enhanced serum specific
IgG, IgG1, IgG2a and IgG2b responses to H3N2 in uenza
virus [40,41]. Collectively, ginseng extract or its puri ed
components appear to elicit improved humoral responses
against microbial invasion irrespective of administration
methods or types of microbes, suggesting the critical
roles of ginseng as an adjuvant in vaccines. More details
on the adjuvant effects of ginseng are well described in
‘GINSENG AS ADJUVANTS’ section in this review.
While many studies have reported the enhanced hu-
moral immune responses by ginseng treatment, ginseng
also reduces antibody production in certain conditions.
For example, long-term (30 consecutive days) oral ad-
ministration of ginseng extract suppressed spleen cell
function and considerably reduced IgG and IgA produc-
tions [38]. In addition, oral administration of ginseng
extract also inhibited an increase in ovalbumin (OVA)-
speci c IgG1 in the serum of OVA-sensitized mice with-
out changing IgG2a, IgA, and IgE levels [42].
Cell-mediated immune response
Cellular immune response is principally mediated by
T cells and NK cells, and is most effective in destroying
virus-infected cells, cells with intracellular bacteria, and
cancer cells presenting tumor antigens [43,44]. It can also
stimulate cells to release various cytokines in response to
antigenic challenge and in uence the function of numer-
ous cells in the immune system [43]. Many reports have
described that ginseng extract significantly enhances
antibody-dependent cellular cytotoxicity and NK cell ac-
tivity derived from human peripheral blood mononuclear
cells (PBMC) or mice [7,29,30]. In addition, ginsenoside
Rc, Rd, Rg1, and ginsan all stimulated T cell prolifera-
tion as well as NK cell activity suggesting a critical im-
munomodulatory effect of ginseng on cellular immune
responses [28,45-47].
In case of autoimmune diseases, immunosuppressive
responses can exert bene cial effects on disease progres-
sion. Hwang et al. [46] has reported that ginsan promotes
the generation of immunosuppressive regulatory T cells
(Tregs) through the activation of transcription factor,
FoxP3, indicating a favorable effect of ginsan on experi-
mental autoimmune encephalomyelitis (EAE), a mouse
model of multiple sclerosis (MS).
http://dx.doi.org/10.5142/jgr.2012.36.4.354
358
J Ginseng Res Vol. 36, No. 4, 354-368 (2012)
CYTOKINE RELEASE
Cytokines are small signaling molecules that are se-
creted by a variety of cells mediating immune responses
[31,48]. Cytokines are synthesized in response to micro-
organisms and other antigenic stimuli, and the binding of
cytokines to their speci c receptors triggers subsequent
cascades of intracellular signaling and in uences the de-
velopment, differentiation, and function of immune cells
[48]. Cytokines are often classi ed, based on their func-
tional properties, into three categories, and they include
modulators of innate immunity, modulators of adaptive
immunity, and regulators of hematopoiesis [48]. How-
ever, this functional classi cation is not always distinct,
and many cytokines play multiple roles in hematopoiesis
and innate and adaptive immunity.
Many literatures have reported that ginseng regulates
immune responses through the modulation of cytokine
secretion. The affected cytokines are mostly proin am-
matory cytokines via the activation or inhibition of Toll-
like receptors (TLRs), but ginseng also changes cytokine
production to control Th1/Th2 lineage differentiation.
Detailed information on the role of ginseng on cytokine
release is described below.
Cytokines of innate immunity
Different cytokines play critical roles in innate im-
munity against microbial invasion, and these mediators
of innate immunity include TNFs, IL-1, IL-6, IL-10, IL-
12, type I interferons, and so on [49]. They are mainly
secreted by macrophages and NK cells while some are
produced by T cells, dendritic cells, and endothelial cells
[49]. In order to detect microbial infection, the innate im-
mune system uses pattern recognition receptors (PRRs),
and TLRs are an important category of PRRs [50,51].
TLR detects the invasion of viruses, bacteria, fungi, and
protozoa, and initiates signaling events which enable
cells to control and clear infections.
Many literatures have shown that ginseng controls
proinflammatory cytokine responses. TNF-α, IL-1β,
IL-6, and IFN-γ are produced by macrophages treated
with ginseng radix extract (GRE) in vitro as well as in
vivo [52]. Spleen cells cultured with ginsan induced an
increase in IL-2, IFN-γ, IL-1α, and granulocyte-macro-
phage colony-stimulating factor production in vitro [53].
However, ginseng can also show opposite effects on
cytokine production. The production of TNF-α, IL-1β,
IL-6, IFN-γ, IL-12, and IL-18, was markedly down-regu-
lated in ginsan-treated mice upon Staphylococcus aureus
challenge, compared with those of control-infected mice
[54]. Ginseng saponin metabolite, compound K inhibited
the expression of inducible nitric-oxide synthase and
proinflammatory cytokines such as TNF-α and IL-1β,
and exerted anti-in ammatory effects in LPS-stimulated
BV2 microglial cells and primary cultured microglia [55].
Similarly, Rg5, a major constituent of steamed ginseng
reduced the expression of IL-1β and TNF-α, as well as
in ammatory enzymes, COX-2 and iNOS in LPS-stim-
ulated alveolar macrophages [56]. In addition, the secre-
tion of in ammatory cytokines such as IFN-γ, IL-1β and
IL-17 were also reduced by ginsan treatment which is
accompanied by a decrease in encephalitogenic response
in EAE, an animal model of MS [46]. TNF-α production
is signi cantly reduced by ginseng and ginsenoside Rb1,
which leads to an anti-arthritic effect on collagen induced
arthritis in mice [57].
The generation of proin ammatory cytokines is main-
ly through the activation of TLRs, and ginseng has been
shown to modulate TLR activation by itself. While the
production of TNF-α and IFN-γ was induced by GRE
in spleen cells and peritoneal macrophages from control
mice, it was impaired in C3H/HeJ mice carrying a defec-
tive TLR-4 gene suggesting that GRE activates TLR-
4 signaling for cytokine release [58]. However, it seems
that GRE does not contain LPS but have a non-LPS
agent that can stimulate TLR-4 according to the endo-
toxin test with the threshold level of 1 ng/mL. In contrast,
some studies have demonstrated that ginseng down-
regulates TLRs and suppresses inflammatory response
[54,59]. Ahn et al. [54] and Ahn et al. [59] reported the
signi cant reduction of TLR-2 and its adaptor molecule
MyD88 expression by ginsan treatment in vitro. In addi-
tion, expression of TLR-2, TLR-4, TLR-9, and MyD88
was considerably reduced in peritoneal macrophages
treated with ginsan, and thereby mice were protected
from S. aureus-induced sepsis [54].
These results indicate that ginseng modulates innate
immunity through TLR expression, and ginseng or gin-
san possesses an antiseptic activity by reducing proin-
ammatory cytokines via TLR signaling and prevents S.
aureus-induced sepsis [54].
Cytokines of acquired immunity
The cytokines of acquired immunity are critical for the
development and activation of effector cells [60]. The ef-
fector cells are further divided into Th1 cells which pro-
duce IL-2, IL-12, IFN-γ, and TNF-β, and Th2 cells which
secrete IL-4, IL-5, IL-10, and IL-13 [60]. The differences
in the cytokines secreted by Th1 and Th2 cells determine
their own differentiation and distinct biological functions
359
Kang et al. Ginseng and Immunity
http://ginsengres.org
[47,60,61].
In addition to the effects on cytokines of innate im-
munity, ginseng also regulates the secretion of cytokines
mediating adaptive immunity. Many literatures have
reported that ginseng itself or various forms of puri-
ed ginsenoside control Th1 and Th2 immune response
differentially [62-66]. The study by Larsen et al. [62]
indicated that ginseng enhanced IL-2 production induc-
ing a stronger Th1 response through the modulation of
stimulated PBMC. Upon challenge with live Candida
albicans, ginsenoside Rg1 helps the infected mice to re-
sist candidiasis by developing Th1-dominant production
of IFN-γ and IL-2 [63]. Similarly, ginseng treatment in
mice inoculated with Pseudomonas aeruginosa stimu-
lated Th1 cytokine response resulting in a faster bacterial
clearance from the lung and significantly milder lung
pathology compared with control group [64]. In addition,
ginsan induced the endogenous production of cytokines
such as IL-1, IL-6, IFN-γ, and IL-12 which are required
for hematopoietic recovery, and was able to enhance Th1
function while interfering with the Th2 response in irra-
diated mice [61]. However, according to the study by Lee
et al. [47], Rg1 enhances Th2 lineage development from
the naïve CD4+T cells both by increasing Th2 specific
cytokine secretion such as IL-4 and by repressing Th1
speci c cytokine production such as IFN-γ. Furthermore,
when ginseng was used as an adjuvant for vaccination,
it seemed to stimulate Th1 and Th2 cytokine responses
equally. For example, when mice were immunized with
vaccines containing ginsenoside Rb1 as adjuvant, aug-
mented productions of IFN-γ, IL-2, IL-4, IL-10, and
TNF-α were reported with no sign of polarization of the
immune response [65]. Similarly, Rd also significantly
promoted the production of the Th1 and Th2 cytokines
in OVA-immunized mice [66]. Such disparities in con-
trolling Th1/Th2 lineage differentiation may have arisen
from differences in dosage, duration of exposure, route
of administration, composition of the extract or the types
of ginsenoside between the studies. Collectively, these
results suggest that ginseng controls Th1/Th2 lineage dif-
ferentiation by changing the balanced production of Th1
and Th2 cytokines. Moreover, the Th1/Th2 fate decision
process is not a unidirectional but bidirectional event
which can change with different experimental conditions.
EFFECTS OF GINSENG ON INFLAMMATORY
DISEASE
Inflammation is a physiological response to various
stimuli such as invading pathogens, irritants, and tissue
injury, and can be classified as acute or chronic [67].
In general, an acute inflammatory response has a rapid
onset with short duration time, and is characterized by
a rapid movement of plasma proteins and leukocytes to
the injured tissues [67]. Although the acute response is
a protective attempt to remove stimuli and to initiate the
healing process, sustained inflammation may lead to a
detrimental, pathologic consequences on host [68]. Such
chronic in ammatory responses may result in signi cant
tissue damages and develop to autoimmune diseases [69].
Ginseng has been shown to reduce the production of
pro-inflammatory cytokines and thus, ameliorate the
symptoms and the progression of in ammatory diseases
[54,70,71]. In an ovalbumin-sensitized murine asthma
model, ginseng effectively suppresses airway hyper-
responsiveness by decreasing inflammatory cell infil-
tration, CD40 engagement of lymphocytes, eosinophil
major basic protein production, and cytokine secretion
[70]. This anti-asthmatic effect of ginseng was con rmed
by Babayigit et al. [72] who have reported the improved
histopathological changes of the lung in asthmatic mice
with ginseng treatment. Additionally, Lim et al. [73]
demonstrated that ginsan administration reduced air-
way hyperresponsiveness, remodeling and eosinophilia,
which was equivalent to the effect of dexamethasone.
Ginseng can also protect host from bacterial septic
responses through the suppression of robust acute in-
ammation. Sepsis is a dangerous and systemic disease
caused by fatal and aberrant inflammatory response to
infections [54]. As previously described in the ‘Cytokines
of innate immunity’ section, ginsan and total polysaccha-
ride have shown to suppress acute in ammation and pro-
tect host mice from sepsis-induced death upon bacterial
challenge [17,54,59]. Interestingly, although the studies
demonstrated the same anti-septic effects upon S. aureus
challenge, the mechanisms behind showed discrepancy
in cytokine production. For example, ginsan down-regu-
lated the secretion of in ammatory cytokines such as IL-
1β, IL-6, IFN-γ, IL-12, and IL-18, but total polysaccha-
ride increased the level of IL-1 and IL-6. Collectively, the
primary anti-septic protective effects given by ginsan or
total polysaccharide seemed to be attributed to enhanced
bacterial clearance through the augmented phagocytic
activity of monocytes and macrophages rather than the
reduced synthesis of sepsis-inducing in ammatory cyto-
kines.
In addition, ginseng exerts beneficial effects on ce-
rebral ischemia by modulating central nervous system
inflammation [55,74]. Ginsenoside Rb1 depressed the
activation of microglia and its conversion to phagocytic
http://dx.doi.org/10.5142/jgr.2012.36.4.354
360
J Ginseng Res Vol. 36, No. 4, 354-368 (2012)
microglia as well as TNF-α and IL-6 production [74].
Compound K inhibited the expressions of inducible
nitric-oxide synthase, proinflammatory cytokines such
as TNF-α and IL-1β, monocyte chemotactic protein-1,
and matrix metalloproteinase-3 and -9 in LPS-stimulated
BV2 microglial cells and primary cultured microglia [55].
These results propose the application of ginseng as a
neuroprotective agent for prevention and/or treatment of
neuroin ammatory diseases such as cerebral ischemia.
In the murine model of collagen-induced arthritis, red
ginseng saponin extract including ginsenosides Rg3,
Rk1 and Rg5, n-butanol extract, and ginsenoside Rb1 all
showed anti-arthritic effects [57,75,76]. They presented
reduced in ammatory cell in ltration, cytokine produc-
tion, and cartilage destruction, and thereby ameliorated
the clinical arthritis score significantly. Taken together,
the results suggest that various forms of ginseng could
be beneficial supplements or alternatives to the current
anti-TNF-α therapeutics for rheumatoid arthritis or other
in ammatory diseases characterized by TNF-α up-regu-
lation.
Lastly, numerous literatures have described the ef-
fects of ginseng on contact and atopic dermatitis [77-82].
Korean red ginseng (KRG) extracts have been shown
to suppress the production of TNF-α and IL-8 in human
keratinocytes [78]. Furthermore, in a mouse model of
atopic dermatitis, topical application of ginseng extract
or ginsenoside Rb1 signi cantly improved the atopic der-
matitis-like skin lesion and scratching behavior as well
as the decrease of serum IgE, IL-4, and IL-10 [5,79,81].
This anti-atopic dermatitis effect was also observed in
mice when KRG was given orally [80], and further con-
rmed in the human study with 30 atopic patients. Lee
and Son [82] reported that 16-week oral ingestion of
KRG decreased serum IgE level and the severity of scor-
ing of the atopic dermatitis index, suggesting a role of
KRG as a functional food for atopic dermatitis.
ANTIMICROBIAL ACTIVITY OF GINSENG
Plants are continuously in contact with different mi-
croorganisms such as viruses, bacteria and fungi. The in-
teractions between plants and microbes may be bene cial
for the plants, but many plant-associated microbes are
pathogens which affect development, reproduction, and
growth of the plants [83]. Therefore, plants produce anti-
microbial compounds as a defense mechanism against
microbial attacks, and these plant-derived compounds
have been reported to prevent bacterial or viral infection
also in humans. Especially, ginseng is one of the best-
known medicinal herbs improving microbial clearance
from the body [8,84,85]. Total ginseng extracts as well
as single or multiple components derived from ginseng
have shown anti-microbial activities, and clinical trials
have also been performed to evaluate the anti-bacterial or
anti-viral activities of ginseng.
Anti-bacterial activity
For successful establishment of bacterial infections, it
is necessary to have adhesion to host cells, colonization
of tissues, and in certain cases, cellular invasion followed
by intracellular multiplication, dissemination to other tis-
sues, or persistence [86]. In general, bacterial adhesion is
mediated by certain interactions between adhesin from
bacteria and carbohydrates on the surface of host cells.
Studies have shown that ginseng polysaccharides could
interact with microbes, interrupt microbial adhesion to
host cells, and block the initiation of infectious disease
[87,88]. For example, PG-F2 and PG-HMW, pectin-
type polysaccharides from P. ginseng, had anti-adhesive
activity against various microbes such as Porphyromo-
nas gingivalis, Actinobacillus actinomycetemcomitans,
Propionibacterium acnes, and S. aureus [87]. These two
polysaccharides did not directly affect microbial growth
but decreased the percentages of attached microbe, sug-
gesting that ginseng polysaccharides have potential roles
in anti-adhesive activity. In addition, polyacetylenes
isolated from P. ginseng hairy root culture exerts direct
bactericidal effects [89]. Four different polyacetylene
compounds were treated to various microorganisms
such as Gram-positive bacteria (methicillin-resistant S.
aureus and Bacillus subtilis) and Gram-negative bacteria
(Escherichia coli and Serratia marcescens), and their
minimum inhibitory concentrations were measured. The
results show that polyacetylene compounds were ef-
fective against bacterial infections and the level of anti-
bacterial activity was dependent on structural features of
the polyacetylene.
Additionally, ginsan had bactericidal activity against
S. aureus [54]. Although ginsan treatment had no direct
effects on bacterial killing in vitro, pretreatment of mice
with ginsan showed enhanced anti-Staphylococcal ac-
tivity by decreasing the numbers of bacteria present in
the spleens, kidney, and blood. The phagocytic activity
against S. aureus was also enhanced considerably in
ginsan-treated mice. In addition to bactericidal activity,
ginseng polysaccharide and ginsan showed anti-septi-
caemic effects against S. aureus infection and protected
mice from septic death during early acute in ammation
as previously discussed [17,54].
361
Kang et al. Ginseng and Immunity
http://ginsengres.org
Microorganisms have their own speci c mode of ac-
tion by which they survive or exert pathogenic effects in
the host. Ginseng can inhibit those survival or pathogenic
mechanisms and thereby exhibit anti-bacterial ability.
For example, acidic saccharides from P. ginseng and red
ginseng extract (RGE) have shown to possess the protec-
tive effects against Helicobacter pylori causing gastric
in ammation, ulceration, and DNA damage [88,90,91].
Through the treatment of acidic saccharides from P. gin-
seng, H. pylori could not attach to gastric cells and initi-
ate in ammatory responses [88,90,91]. Moreover, RGE
was identified to signicantly weaken both H. pylori-
induced DNA damage and gastric cell apoptosis by in-
activating ERK1/2 signaling and attenuating caspase-3
activation and poly ADP ribose polymerase cleavage [91].
Furthermore, literatures have described the anti-
Pseudomonal activity of ginseng. Ginseng treatment
signi cantly decreased the lung pathology and enhanced
bacterial clearance from the lung in a rat model of
chronic P. aeruginosa lung infection mimicking cystic -
brosis [64,84,85,92]. During infectious process by P. ae-
ruginosa, cell-to-cell communication mechanism called
quorum sensing (QS) plays a critical role in the patho-
genesis of the bacteria. The QS of P. aeruginosa uses N-
acetylated homoserine lactone as a signal molecule and
coordinates the formation of bio lms, swarming motil-
ity, exopolysaccharide production, and cell aggregation.
Song et al. [93] have shown that hot water extracts from
ginseng compromises the generation of P. aeruginosa
QS. Although hot water ginseng extracts did not inhibit
the growth of P. aeruginosa directly and enhanced the
production of capsule-like polysaccharides and alginates,
ginseng extracts suppressed the synthesis of virulence
factors such as LasA protease, LasB elastase, and AHL
molecules, and strongly inhibited QS system of P. aeru-
ginosa [93].
Anti-viral activity
In uenza virus, commonly referred to as the u, can
be influenced by RGE [94-97]. After intranasal admin-
istration of ginseng extract with in uenza virus A/PR8,
significant increases in IgA as well as total IgG were
observed in blood, lung, vaginal lavage, and fecal extract
in mice [98]. The increase in total IgG was comparable
to that observed in the aluminium hydroxide or cholera
toxin-adjuvant group. In addition, all serum IgG subtypes
such as IgG1, IgG2a, IgG2b, and IgG3 antibodies were all
augmented [98]. Simultaneously, virus neutralization
activity, cytokine production, body weight changes, and
survival rates were all improved by ginseng treatment
indicating that ginseng extract well-executes immuno-
regulatory function as an adjuvant.
Avian influenza H9N2 is generally related to major
mortality of human endothelial cells and causes serious
in ammation and apoptosis on host cells [95,96]. H9N2
may induce the generation of reactive oxygen species
(ROS), stimulate inflammatory response by produc-
ing IP-10 chemokine, and cause cellular apoptosis and
DNA damages. Two ginsenosides have shown to possess
different protective functions against H9N2 and medi-
ate dual roles in infectious situations [95]. Ginsenoside
protopanaxatriol reduced ROS stress and decreased IP-
10 expression stimulated by ROS, whereas ginsenoside
Re reduced the in uenza-induced DNA damage and cell
death.
Infection of in uenza A (H1N1) virus is also regulated
by red ginseng extracts [97]. RGE treatment inhibited
plaque formation occurred by in uenza A virus in cell-
based system, and oral administration of RGE improved
body weight loss and decreased influenza A titer in
mouse lung.
Human immunodeciency virus type 1 (HIV-1) is a
virus that causes AIDS, a condition in which the immune
system fails due to the destruction of CD4+ helper T
cells (mostly), macrophages, and dendritic cells [99,100].
Owing to the introduction of highly active anti-retroviral
therapy (HAART) and the development of many anti-ret-
roviral drugs, the rates of mortality and morbidity related
to HIV-1 disease have reduced meaningfully [101]. How-
ever, anti-retroviral drug-resistant mutants are incessantly
occurring and limit the availability of effective drugs. In-
terestingly, KRG has been shown to exert positive effects
on HAART by maintaining CD4+ T cell counts and de-
laying the development of resistance mutations in HIV-1
patients treated with HAART [102]. Negative factor (Nef)
is a virulence factor securing T cell activation and the
establishment of a persistent state of infection early in the
HIV life cycle. Nef proteins derived from the long-term
non/slow progressors were found to be defective or far
less capable of enhancing viral replication and infectiv-
ity [103]. Interestingly, KRG increases the frequency of
gross deletion in the nef gene resulting in the delay in dis-
ease progression in HIV-1 patients [104-107]. Similarly,
increased frequency of deletion in the HIV-1 5’ LTR/gag
was found in HIV-1 infected long-term survivors treated
with KRG, suggesting the association of gross deletion
in HIV-1 5’ LTR/gag regions with KRG intake [107-109].
Therefore, combinational treatment or pre-treatment of
KRG with HAART is very valuable for the treatment of
HIV infections.
http://dx.doi.org/10.5142/jgr.2012.36.4.354
362
J Ginseng Res Vol. 36, No. 4, 354-368 (2012)
Ginseng also had inhibitory effects on the growth of
other viruses such as rotavirus, murine norovirus (MNV),
and feline calicivirus (FCV) [110,111]. Pre-treatment but
not post-treatment of mice with RGE exhibited antiviral
effects on MNV and FCV, surrogates for human norovi-
rus. In addition, ginseng polysaccharide fractions showed
anti-rotaviral activity with the inhibitory potency compa-
rable to that of hesperidin, one of the most potent agent
for rotaviral infection [111].
Although the eradication of viral infection mainly
relies on the immune responses of the host, anti-viral
properties of ginseng have been rarely assessed from an
immunological perspective. Instead of examining the ac-
tivities of cytotoxic T cells and NK cells and the produc-
tion of interferon and neutralizing antibody, body weight
loss, survival rate, virus titer, and genetic mutation of
virulence factors have been employed to prove anti-viral
activities of ginseng. Future studies should include the
immunological methods described above to assess anti-
viral activity of ginseng more thoroughly.
GINSENG AS ADJUVANTS
Adjuvants play an important role in vaccine formula-
tions by increasing the immunogenicity of co-adminis-
tered antigens [112,113]. When appropriate adjuvants are
administrated with a supplied antigen, they can enhance
immunization effects while keeping the injected antigen
to a minimum and protect their recipients from deadly
infectious diseases. Adjuvants can act in various ways
to increase antigen presentation and to stimulate the im-
mune system [114]. For example, adjuvants may act as a
depot for the antigen and present the antigen over a long
period of time to maximize the immune responses. They
can also induce mucosal immunity, regulate antibody
avidity, speci city, or isotype, and stimulate cytotoxic T
lymphocytes. Many substances have been developed as
adjuvants [112]. Inorganic aluminium salts such as alu-
minium phosphate and aluminium hydroxide are one of
the most common adjuvants in human vaccine. In addi-
tion, oil emulsions, polymers, carbohydrates, liposomes,
LPSs and other bacterial toxins are also used to augment
immune responses.
Ginseng extract has been applied as an immunologic
adjuvant and ginsenosides are believed to be the key
molecules that have adjuvanticity among the whole gin-
seng extract [115]. There are about 30 ginsenosides pres-
ent in P. ginseng and their ability as an adjuvant is depen-
dent on their characteristic structural features [116,117].
In general, ginseng has no immunogenicity by itself [118].
However, their combinations with antigens or other
adjuvants can promote and enhance immune responses
against immunized antigens.
Antibody production could be regulated by ginseng
extract and speci c ginsenoside. It has been reported that
ginseng extract increases IgM and IgG antibody responses
in immunized mice [7], and ginsenosides Rd, Re and Rg1
also augment specific-antibody responses [41,66,118-
121]. Similarly, a novel adjuvant of ginsenoside-based
nanoparticles (ginsomes) containing ginsenosides Rb2,
Rc, Rb1, and Rd signi cantly increased the levels of spe-
ci c IgG1, IgG2a, IgG2b and IgG3 in mice [119].
Recent studies of ginseng also emphasize the adjuvant
effects of ginseng on the Th1 and Th2 immune responses
as well as antibody responses. In addition to increase in
lymphocyte proliferation, ginseng promotes the produc-
tion of cytokines which stimulate both Th1 and Th2
immune responses. The productions of IFN-γ and IL-5
were increased by administration of ginsenoside Re and
Rg1 in mice [118,119,121]. Ginsenoside Rd and Re also
improved the productions of Th1 and Th2 cytokines
including IFN-γ, IL-2, IL-4, IL-10, IL-12, and TNF-α
[41,66,120], and consequently, up-regulated Th1 and Th2
responses leading to a balanced immunity.
For these reasons, ginseng has been suggested as a po-
tent adjuvant for vaccine. For example, in the vaccine of
T. gondii, ginseng was regarded as a promising vaccine
adjuvant against toxoplasmosis by enhancing antibody
response against T. gondii antigen [41]. In in uenza vi-
rus vaccine, ginsenoside Re is expected to improve the
quality of vaccine that may activate mixed Th1/Th2 im-
mune responses [118]. Overall, these results indicate that
ginseng functions well as an immunologic adjuvant, and
its combination with antigens would bring enhanced im-
mune responses during immunization.
CONCLUDING REMARKS
Here we discussed the current knowledge on the ef-
fects of ginseng on immune system (Table 1). Although
ample studies have examined the immunomodulatory
properties of ginseng in vitro and in animals, most of
them are limited to assess the phenotypic changes at
the cellular level, and only a few studies have looked
at the alterations by ginseng at the molecular level. For
example, cytokine secretion, antibody production, sur-
face marker expression, and cellular functions such as
phagocytosis and cytotoxicity were common criteria to
evaluate immunoregulatory properties, while molecular
components involved in signaling pathway were rarely
363
Kang et al. Ginseng and Immunity
http://ginsengres.org
Table 1. Summary of the ginseng effects on immune system
Effects References
Innate immunity
Macrophages Enhance phagocytosis [8,16,17]
Stimulate the generation of nitric oxide [16,18,19]
Induce in ammatory reactions by IL-1β and TNF-α[17,18]
Stimulate NK cells and T cells by IL-12 [20]
DCs Increase DC maturation markers (MHC class II, CD80, CD83, CD86) [23,24]
Increase the production of IL-1 and TNF-α [23]
Reduce DC maturation markers (CD40, CD86, CD1a, HLA-DR) [25]
Diminish the secretion of TNF-α, IL-12 and IL-12 p40 [25,26]
NK cells Improve natural killing activity [7,8,28-30]
Acquired immunity
Humoral immunity Induce responses and production of IgA, IgM, IgG and IgG subunits [37-41]
Inhibit the OVA-sensitized decrease in IgA level in the small intestine [42]
Cell-mediated immunity Enhance antibody-dependent cellular cytotoxicity [7,29,30]
Stimulate T cell proliferation [28,45-47]
Promote the generation of immunosuppressive regulatory T cells (Tregs) [46]
Cytokine release
Of innate immunity Produce TNF-α, IL-1β, IL-6, and IFN-γ by macrophages [52]
Increase IL-2, IFN-γ, IL-1α, and GM-CSF production [53]
Decrease TNF-α, IL-1β, IL-6, IFN-γ, IL-12, and IL-18 by TLR reduction [54]
Reduce nitric oxide synthesis and production of TNF-α and IL-1β[55-57]
Inhibit secretion of IL-1β, IL-17 and IFN-γ[46]
Of acquired immunity Enhance production of IFN-γ and IL-2 [62,63]
Increase generation of IL-1, IL-6, IFN-γ, and IL-12 [61]
Increase Th2 cytokine (e.g., IL-4) and repress Th1 cytokine (e.g., IFN-γ) [47]
Stimulate Th1 and Th2 cytokine equally (IFN-γ, IL-2, IL-4, IL-10, and TNF-α) [65,66]
In ammatory disease regulation
Mitigate asthma symptoms suppressing airway hyperresponsiveness [70,72,73]
Protect host from bacterial septic responses [17,54,59]
Modulate CNS in ammation and improve cerebral ischemia [55,74]
Ameliorate arthritis by reducing in ammatory cell in ltration, cytokine production, and cartilage destruction [57,75,76]
Alleviate atopic dermatitis by decreasing in ammatory cytokines [77-82]
Anti-microbial activity
Bacteria Interrupt microbial adhesion to host cells [87,88]
Kill Gram positive and Gram nagative bacteria directly [89]
Inhibit survival or pathogenic mechanism of speci c pathogen [88,90,91]
Enhance bacterial clearance and decrease pathology [64,84,85,92]
Interrupt quorum sensing system [93]
In uenza virus Increase antibody production and response with in uenza virus A/PR8 infection [98]
Reduced ROS stress, IP-10, in uenza-induced DNA damage and apoptosis in H9N2 infection [95]
Inhibit proliferation of in uenza virus A (H1N1) [97]
HIV-1 Increase the frequency of gross deletion in the nef gene [104-107]
Increases the frequency of gross deletion in HIV-1 5’ LTR/ gag regions [107-109]
Others Inhibit viral growth of rotavirus, MNV and FCV [110,111]
Adjuvant
Improve antibody production and responsiveness (IgM, IgG, and IgG subunits) [7,41,66,118-121]
Activate mixed Th1 and Th2 immune responses [41,66,118-121]
NK, natural killer; DC, dendritic cells; MHC, major histocompatibility complex; OVA, ovalbumin; GM-CSF, granulocyte-macrophage colony-stimu-
lating factor; CNS, central nervous system; ROS, reactive oxygen species; HIV-1, human immunodeciency virus type 1; MNV, murine norovirus;
FCV, feline calicivirus.
http://dx.doi.org/10.5142/jgr.2012.36.4.354
364
J Ginseng Res Vol. 36, No. 4, 354-368 (2012)
investigated. In order to reveal the underlying molecular
mechanisms for the immunomodulating effects of gin-
seng in more detail, further in-depth studies need to be
provided.
As a well-known herbal immune stimulant, hundreds
of studies have extensively reported the anti-cancer or
chemopreventive effects of ginseng. The anti-cancer ef-
fects of ginseng are mainly through the improvements in
cell-mediated immunity consisting of cytotoxic T cells
and NK cells, while other mechanisms such as oxidative
stress, apoptosis, and angiogenesis are also involved. A
comprehensive review dealing with anti-cancer effects
from the immunological point of view would be impera-
tive in the near future.
ACKNOWLEDGEMENTS
This research was supported by the Public welfare &
Safety research program through the National Research
Foundation of Korea funded by the Ministry of Educa-
tion, Science and Technology (2010-0020842).
REFERENCES
51. Akerele O. WHO guideline for assessment of herbal
medicines. Fitoterapia 1992;63:99-104.
52. Baek SH, Bae ON, Park JH. Recent methodology in gin-
seng analysis. J Ginseng Res 2012;36:119-134.
53. Kim MK, Lee JW, Lee KY, Yang DC. Microbial conver-
sion of major ginsenoside Rb1 to pharmaceutically active
minor ginsenoside Rd. J Microbiol 2005;43:456-462.
54. Cui J, Garle M, Eneroth P, Bjorkhem I. What do commer-
cial ginseng preparations contain? Lancet 1994;344:134.
55. Sohn EH, Jang SA, Lee CH, Jang KH, Kang SC, Park
HJ, Pyo SN. Effects of Korean red ginseng extract for the
treatment of atopic dermatitis-like skin lesions in mice. J
Ginseng Res 2011;35:479-486.
56. Bae EA, Park SY, Kim DH. Constitutive beta-glucosi-
dases hydrolyzing ginsenoside Rb1 and Rb2 from human
intestinal bacteria. Biol Pharm Bull 2000;23:1481-1485.
57. Jie YH, Cammisuli S, Baggiolini M. Immunomodula-
tory effects of Panax ginseng C.A. Meyer in the mouse.
Agents Actions 1984;15:386-391.
58. Scaglione F, Ferrara F, Dugnani S, Falchi M, Santoro G,
Fraschini F. Immunomodulatory effects of two extracts
of Panax ginseng C.A. Meyer. Drugs Exp Clin Res
1990;16:537-542.
59. Choi KT. Botanical characteristics, pharmacological ef-
fects and medicinal components of Korean Panax ginseng
C A Meyer. Acta Pharmacol Sin 2008;29:1109-1118.
10. Kitts D, Hu C. Efficacy and safety of ginseng. Public
Health Nutr 2000;3:473-485.
11. Elias PM. The skin barrier as an innate immune element.
Semin Immunopathol 2007;29:3-14.
12. Medzhitov R, Janeway CA Jr. Innate immunity: impact
on the adaptive immune response. Curr Opin Immunol
1997;9:4-9.
13. Albiger B, Dahlberg S, Henriques-Normark B, Normark S.
Role of the innate immune system in host defence against
bacterial infections: focus on the Toll-like receptors. J In-
tern Med 2007;261:511-528.
14. Gordon S. The macrophage. Bioessays 1995;17:977-986.
15. Guzik TJ, Korbut R, Adamek-Guzik T. Nitric oxide and
superoxide in inflammation and immune regulation. J
Physiol Pharmacol 2003;54:469-487.
16. Shin JY, Song JY, Yun YS, Yang HO, Rhee DK, Pyo S.
Immunostimulating effects of acidic polysaccharides ex-
tract of Panax ginseng on macrophage function. Immuno-
pharmacol Immunotoxicol 2002;24:469-482.
17. Lim DS, Bae KG, Jung IS, Kim CH, Yun YS, Song JY.
Anti-septicaemic effect of polysaccharide from Panax
ginseng by macrophage activation. J Infect 2002;45:32-
38.
18. Choi HS, Kim KH, Sohn E, Park JD, Kim BO, Moon
EY, Rhee DK, Pyo S. Red ginseng acidic polysaccha-
ride (RGAP) in combination with IFN-gamma results
in enhanced macrophage function through activation of
the NF-kappaB pathway. Biosci Biotechnol Biochem
2008;72:1817-1825.
19. Friedl R, Moeslinger T, Kopp B, Spieckermann PG. Stim-
ulation of nitric oxide synthesis by the aqueous extract of
Panax ginseng root in RAW 264.7 cells. Br J Pharmacol
2001;134:1663-1670.
20. Wang H, Actor JK, Indrigo J, Olsen M, Dasgupta A.
Asian and Siberian ginseng as a potential modulator of
immune function: an in vitro cytokine study using mouse
macrophages. Clin Chim Acta 2003;327:123-128.
21. Palucka K, Banchereau J. Dendritic cells: a link be-
tween innate and adaptive immunity. J Clin Immunol
1999;19:12-25.
22. Stockwin LH, McGonagle D, Martin IG, Blair GE. Den-
dritic cells: immunological sentinels with a central role in
health and disease. Immunol Cell Biol 2000;78:91-102.
23. Kim MH, Byon YY, Ko EJ, Song JY, Yun YS, Shin T, Joo
HG. Immunomodulatory activity of ginsan, a polysac-
charide of Panax ginseng, on dendritic cells. Korean J
Physiol Pharmacol 2009;13:169-173.
24. Takei M, Tachikawa E, Hasegawa H, Lee JJ. Dendritic
cells maturation promoted by M1 and M4, end products
of steroidal ginseng saponins metabolized in digestive
365
Kang et al. Ginseng and Immunity
http://ginsengres.org
tracts, drive a potent Th1 polarization. Biochem Pharma-
col 2004;68:441-452.
25. Su W, Sun AJ, Xu DL, Zhang HQ, Yang L, Yuan LY, Jia
JG, Zou YZ, Wu YL, Wang KQ et al. Inhibiting effects of
total saponins of Panax ginseng on immune maturation of
dendritic cells induced by oxidized-low density lipopro-
tein. Cell Immunol 2010;263:99-104.
26. Tung NH, Quang TH, Son JH, Koo JE, Hong HJ, Koh
YS, Song GY, Kim YH. Inhibitory effect of ginsenosides
from steamed ginseng-leaves and flowers on the LPS-
stimulated IL-12 production in bone marrow-derived den-
dritic cells. Arch Pharm Res 2011;34:681-685.
27. Vivier E, Nunes JA, Vely F. Natural killer cell signaling
pathways. Science 2004;306:1517-1519.
28. Kenarova B, Neychev H, Hadjiivanova C, Petkov VD.
Immunomodulating activity of ginsenoside Rg1 from
Panax ginseng. Jpn J Pharmacol 1990;54:447-454.
29. Kim JY, Germolec DR, Luster MI. Panax ginseng as a
potential immunomodulator: studies in mice. Immuno-
pharmacol Immunotoxicol 1990;12:257-276.
30. See DM, Broumand N, Sahl L, Tilles JG. In vitro effects
of echinacea and ginseng on natural killer and antibody-
dependent cell cytotoxicity in healthy subjects and
chronic fatigue syndrome or acquired immunode ciency
syndrome patients. Immunopharmacology 1997;35:229-
235.
31. Hansson GK, Libby P, Schonbeck U, Yan ZQ. Innate and
adaptive immunity in the pathogenesis of atherosclerosis.
Circ Res 2002;91:281-291.
32. Kurtz J. Memory in the innate and adaptive immune sys-
tems. Microbes Infect 2004;6:1410-1417.
33. Slifka MK, Antia R, Whitmire JK, Ahmed R. Humoral
immunity due to long-lived plasma cells. Immunity
1998;8:363-372.
34. Newton SM, Jacob CO, Stocker BA. Immune response to
cholera toxin epitope inserted in Salmonella agellin. Sci-
ence 1989;244:70-72.
35. Vecchiarelli A, Casadevall A. Antibody-mediated effects
against Cryptococcus neoformans: evidence for interde-
pendency and collaboration between humoral and cellular
immunity. Res Immunol 1998;149:321-333.
36. Dunkelberger JR, Song WC. Complement and its role
in innate and adaptive immune responses. Cell Res
2010;20:34-50.
37. Liou CJ, Huang WC, Tseng J. Long-term oral adminis-
tration of ginseng extract modulates humoral immune
response and spleen cell functions. Am J Chin Med
2005;33:651-661.
38. Liou CJ, Li ML, Tseng J. Intraperitoneal injection of gin-
seng extract enhances both immunoglobulin and cytokine
production in mice. Am J Chin Med 2004;32:75-88.
39. Na HS, Lim YJ, Yun YS, Kweon MN, Lee HC. Ginsan
enhances humoral antibody response to orally delivered
antigen. Immune Netw 2010;10:5-14.
40. Yoo DG, Kim MC, Park MK, Park KM, Quan FS, Song
JM, Wee JJ, Wang BZ, Cho YK, Compans RW et al.
Protective effect of ginseng polysaccharides on in uenza
viral infection. PLoS One 2012;7:e33678.
41. Qu DF, Yu HJ, Liu Z, Zhang DF, Zhou QJ, Zhang HL, Du
AF. Ginsenoside Rg1 enhances immune response induced
by recombinant Toxoplasma gondii SAG1 antigen. Vet
Parasitol 2011;179:28-34.
42. Sumiyoshi M, Sakanaka M, Kimura Y. Effects of red gin-
seng extract on allergic reactions to food in Balb/c mice. J
Ethnopharmacol 2010;132:206-212.
43. Behrens G, Li M, Smith CM, Belz GT, Mintern J, Car-
bone FR, Heath WR. Helper T cells, dendritic cells and
CTL Immunity. Immunol Cell Biol 2004;82:84-90.
44. Russell JH, Ley TJ. Lymphocyte-mediated cytotoxicity.
Annu Rev Immunol 2002;20:323-370.
45. Berek L, Szabo D, Petri IB, Shoyama Y, Lin YH, Molnar
J. Effects of naturally occurring glucosides, solasodine
glucosides, ginsenosides and parishin derivatives on mul-
tidrug resistance of lymphoma cells and leukocyte func-
tions. In Vivo 2001;15:151-156.
46. Hwang I, Ahn G, Park E, Ha D, Song JY, Jee Y. An acidic
polysaccharide of Panax ginseng ameliorates experimen-
tal autoimmune encephalomyelitis and induces regulatory
T cells. Immunol Lett 2011;138:169-178.
47. Lee EJ, Ko E, Lee J, Rho S, Ko S, Shin MK, Min BI,
Hong MC, Kim SY, Bae H. Ginsenoside Rg1 enhances
CD4(+) T-cell activities and modulates Th1/Th2 differen-
tiation. Int Immunopharmacol 2004;4:235-244.
48. Seo N, Hayakawa S, Tokura Y. Mechanisms of immune
privilege for tumor cells by regulatory cytokines produced
by innate and acquired immune cells. Semin Cancer Biol
2002;12:291-300.
49. Alfano M, Poli G. Role of cytokines and chemokines in
the regulation of innate immunity and HIV infection. Mol
Immunol 2005;42:161-182.
50. Iwasaki A, Medzhitov R. Toll-like receptor control of the
adaptive immune responses. Nat Immunol 2004;5:987-
995.
51. Medzhitov R. Toll-like receptors and innate immunity.
Nat Rev Immunol 2001;1:135-145.
52. Liou CJ, Huang WC, Tseng J. Short-term oral administra-
tion of ginseng extract induces type-1 cytokine produc-
tion. Immunopharmacol Immunotoxicol 2006;28:227-
240.
53. Kim KH, Lee YS, Jung IS, Park SY, Chung HY, Lee IR,
http://dx.doi.org/10.5142/jgr.2012.36.4.354
366
J Ginseng Res Vol. 36, No. 4, 354-368 (2012)
Yun YS. Acidic polysaccharide from Panax ginseng,
ginsan, induces Th1 cell and macrophage cytokines and
generates LAK cells in synergy with rIL-2. Planta Med
1998;64:110-115.
54. Ahn JY, Song JY, Yun YS, Jeong G, Choi IS. Protection of
Staphylococcus aureus-infected septic mice by suppres-
sion of early acute in ammation and enhanced antimicro-
bial activity by ginsan. FEMS Immunol Med Microbiol
2006;46:187-197.
55. Park JS, Shin JA, Jung JS, Hyun JW, Van Le TK, Kim
DH, Park EM, Kim HS. Anti-in ammatory mechanism of
compound K in activated microglia and its neuroprotec-
tive effect on experimental stroke in mice. J Pharmacol
Exp Ther 2012;341:59-67.
56. Kim TW, Joh EH, Kim B, Kim DH. Ginsenoside Rg5
ameliorates lung in ammation in mice by inhibiting the
binding of LPS to toll-like receptor-4 on macrophages. Int
Immunopharmacol 2012;12:110-116.
57. Kim HA, Kim S, Chang SH, Hwang HJ, Choi YN. Anti-
arthritic effect of ginsenoside Rb1 on collagen induced ar-
thritis in mice. Int Immunopharmacol 2007;7:1286-1291.
58. Nakaya TA, Kita M, Kuriyama H, Iwakura Y, Imanishi
J. Panax ginseng induces production of proin ammatory
cytokines via toll-like receptor. J Interferon Cytokine Res
2004;24:93-100.
59. Ahn JY, Choi IS, Shim JY, Yun EK, Yun YS, Jeong G,
Song JY. The immunomodulator ginsan induces re-
sistance to experimental sepsis by inhibiting Toll-like
receptor-mediated in ammatory signals. Eur J Immunol
2006;36:37-45.
60. Constant SL, Bottomly K. Induction of Th1 and Th2
CD4+ T cell responses: the alternative approaches. Annu
Rev Immunol 1997;15:297-322.
61. Han SK, Song JY, Yun YS, Yi SY. Ginsan improved Th1
immune response inhibited by gamma radiation. Arch
Pharm Res 2005;28:343-350.
62. Larsen MW, Moser C, Hoiby N, Song Z, Kharazmi A.
Ginseng modulates the immune response by induction of
interleukin-12 production. APMIS 2004;112:369-373.
63. Lee JH, Han Y. Ginsenoside Rg1 helps mice resist to
disseminated candidiasis by Th1 type differentiation of
CD4+ T cell. Int Immunopharmacol 2006;6:1424-1430.
64. Song Z, Moser C, Wu H, Faber V, Kharazmi A, Hoiby
N. Cytokine modulating effect of ginseng treatment in a
mouse model of Pseudomonas aeruginosa lung infection.
J Cyst Fibros 2003;2:112-119.
65. Rivera E, Ekholm Pettersson F, Inganas M, Paulie S,
Gronvik KO. The Rb1 fraction of ginseng elicits a bal-
anced Th1 and Th2 immune response. Vaccine 2005;23:
5411-5419.
66. 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.
67. Gabay C, Kushner I. Acute-phase proteins and other
systemic responses to inflammation. N Engl J Med
1999;340:448-454.
68. Nagy G, Clark JM, Buzas EI, Gorman CL, Cope AP. Ni-
tric oxide, chronic in ammation and autoimmunity. Im-
munol Lett 2007;111:1-5.
69. Manzo A, Bombardieri M, Humby F, Pitzalis C. Second-
ary and ectopic lymphoid tissue responses in rheumatoid
arthritis: from in ammation to autoimmunity and tissue
damage/remodeling. Immunol Rev 2010;233:267-285.
70. Kim DY, Yang WM. Panax ginseng ameliorates airway
in ammation in an ovalbumin-sensitized mouse allergic
asthma model. J Ethnopharmacol 2011;136:230-235.
71. Oyagi A, Ogawa K, Kakino M, Hara H. Protective effects
of a gastrointestinal agent containing Korean red ginseng
on gastric ulcer models in mice. BMC Complement Al-
tern Med 2010;10:45.
72. Babayigit A, Olmez D, Karaman O, Bagriyanik HA,
Yilmaz O, Kivcak B, Erbil G, Uzuner N. Ginseng amelio-
rates chronic histopathologic changes in a murine model
of asthma. Allergy Asthma Proc 2008;29:493-498.
73. Lim YJ, Na HS, Yun YS, Choi IS, Oh JS, Rhee JH, Cho
BH, Lee HC. Suppressive effects of ginsan on the devel-
opment of allergic reaction in murine asthmatic model.
Int Arch Allergy Immunol 2009;150:32-42.
74. Zhu J, Jiang Y, Wu L, Lu T, Xu G, Liu X. Suppression
of local in ammation contributes to the neuroprotective
effect of ginsenoside Rb1 in rats with cerebral ischemia.
Neuroscience 2012;202:342-351.
75. Chang SH, Choi Y, Park JA, Jung DS, Shin J, Yang JH,
Ko SY, Kim SW, Kim JK. Anti-in ammatory effects of
BT-201, an n-butanol extract of Panax notoginseng, ob-
served in vitro and in a collagen-induced arthritis model.
Clin Nutr 2007;26:785-791.
76. Kim KR, Chung TY, Shin H, Son SH, Park KK, Choi JH,
Chung WY. Red ginseng saponin extract attenuates mu-
rine collagen-induced arthritis by reducing pro-in amma-
tory responses and matrix metalloproteinase-3 expression.
Biol Pharm Bull 2010;33:604-610.
77. Bae EA, Han MJ, Shin YW, Kim DH. Inhibitory effects
of Korean red ginseng and its genuine constituents gin-
senosides Rg3, Rf, and Rh2 in mouse passive cutaneous
anaphylaxis reaction and contact dermatitis models. Biol
Pharm Bull 2006;29:1862-1867.
78. Hong CE, Lyu SY. Anti-in ammatory and anti-oxidative
effects of Korean red ginseng extract in human keratino-
cytes. Immune Netw 2011;11:42-49.
367
Kang et al. Ginseng and Immunity
http://ginsengres.org
79. Samukawa K, Izumi Y, Shiota M, Nakao T, Osada-Oka
M, Miura K, Iwao H. Red ginseng inhibits scratching be-
havior associated with atopic dermatitis in experimental
animal models. J Pharmacol Sci 2012;118:391-400.
80. Lee JH, Cho SH. Korean red ginseng extract ameliorates
skin lesions in NC/Nga mice: an atopic dermatitis model.
J Ethnopharmacol 2011;133:810-817.
81. Park HJ, Byeon HE, Choi KW, Rhee DK, Lee KR, Pyo
SN. Inhibitory effects of ginsenoside Rb1 on atopic der-
matitis-like skin lesions in mice. J Ginseng Res 2010;34:
363-368.
82. Lee KG, Son SW. Ef cacy of Korean red ginseng in the
treatment of atopic dermatitis. J Ginseng Res 2011;35:
149-154.
83. Gonzalez-Lamothe R, Mitchell G, Gattuso M, Diarra MS,
Malouin F, Bouarab K. Plant antimicrobial agents and
their effects on plant and human pathogens. Int J Mol Sci
2009;10:3400-3419.
84. Song ZJ, Johansen HK, Faber V, Hoiby N. Ginseng treat-
ment enhances bacterial clearance and decreases lung
pathology in athymic rats with chronic P. aeruginosa
pneumonia. APMIS 1997;105:438-444.
85. Song Z, Johansen HK, Faber V, Moser C, Kharazmi A,
Rygaard J, Hoiby N. Ginseng treatment reduces bacterial
load and lung pathology in chronic Pseudomonas aeru-
ginosa pneumonia in rats. Antimicrob Agents Chemother
1997;41:961-964.
86. Pizarro-Cerda J, Cossart P. Bacterial adhesion and entry
into host cells. Cell 2006;124:715-727.
87. Lee JH, Shim JS, Chung MS, Lim ST, Kim KH. Inhibi-
tion of pathogen adhesion to host cells by polysaccha-
rides from Panax ginseng. Biosci Biotechnol Biochem
2009;73:209-212.
88. Lee JH, Shim JS, Lee JS, Kim MK, Chung MS, Kim KH.
Pectin-like acidic polysaccharide from Panax ginseng
with selective antiadhesive activity against pathogenic
bacteria. Carbohydr Res 2006;341:1154-1163.
89. Fukuyama N, Shibuya M, Orihara Y. Antimicrobial poly-
acetylenes from Panax ginseng hairy root culture. Chem
Pharm Bull (Tokyo) 2012;60:377-380.
90. Lee JH, Park EK, Uhm CS, Chung MS, Kim KH. Inhibi-
tion of Helicobacter pylori adhesion to human gastric
adenocarcinoma epithelial cells by acidic polysaccharides
from Artemisia capillaris and Panax ginseng. Planta Med
2004;70:615-619.
91. Park S, Yeo M, Jin JH, Lee KM, Jung JY, Choue R, Cho
SW, Hahm KB. Rescue of Helicobacter pylori-induced
cytotoxicity by red ginseng. Dig Dis Sci 2005;50:1218-
1227.
92. Song Z, Johansen HK, Moser C, Høiby N. Effects of Chi-
nese medicinal herbs on a rat model of chronic Pseudo-
monas aeruginosa lung infection. APMIS 1996;104:350-
354.
93. Song Z, Kong KF, Wu H, Maricic N, Ramalingam B,
Priestap H, Schneper L, Quirke JM, Hoiby N, Mathee
K. Panax ginseng has anti-infective activity against
opportunistic pathogen Pseudomonas aeruginosa by
inhibiting quorum sensing, a bacterial communication
process critical for establishing infection. Phytomedicine
2010;17:1040-1046.
94. Scaglione F, Cattaneo G, Alessandria M, Cogo R. Ef ca-
cy and safety of the standardised ginseng extract G115 for
potentiating vaccination against the influenza syndrome
and protection against the common cold. Drugs Exp Clin
Res 1996;22:65-72.
95. Chan LY, Kwok HH, Chan RW, Peiris MJ, Mak NK,
Wong RN, Chan MC, Yue PY. Dual functions of gin-
senosides in protecting human endothelial cells against
influenza H9N2-induced inflammation and apoptosis. J
Ethnopharmacol 2011;137:1542-1546.
96. Peiris M, Yuen KY, Leung CW, Chan KH, Ip PL, Lai RW,
Orr WK, Shortridge KF. Human infection with in uenza
H9N2. Lancet 1999;354:916-917.
97. Kim JY, Kim HJ, Kim HJ. Effect of oral administration
of Korean red ginseng on in uenza A (H1N1) virus infec-
tion. J Ginseng Res 2011;35:104-110.
98. Quan FS, Compans RW, Cho YK, Kang SM. Ginseng and
Salviae herbs play a role as immune activators and modu-
late immune responses during influenza virus infection.
Vaccine 2007;25:272-282.
99. Douek DC, Roederer M, Koup RA. Emerging concepts
in the immunopathogenesis of AIDS. Annu Rev Med
2009;60:471-484.
100. Weiss RA. How does HIV cause AIDS? Science 1993;
260:1273-1279.
101. Palella FJ Jr, Delaney KM, Moorman AC, Loveless
MO, Fuhrer J, Satten GA, Aschman DJ, Holmberg
SD. Declining morbidity and mortality among patients
with advanced human immunodeficiency virus infec-
tion. HIV Outpatient Study Investigators. N Engl J Med
1998;338:853-860.
102. Sung H, Jung YS, Cho YK. Bene cial effects of a com-
bination of Korean red ginseng and highly active antiret-
roviral therapy in human immunodeficiency virus type
1-infected patients. Clin Vaccine Immunol 2009;16:1127-
1131.
103. Gef n R, Wolf D, Muller R, Hill MD, Stellwag E, Fre-
itag M, Sass G, Scott GB, Baur AS. Functional and struc-
tural defects in HIV type 1 nef genes derived from pedi-
atric long-term survivors. AIDS Res Hum Retroviruses
http://dx.doi.org/10.5142/jgr.2012.36.4.354
368
J Ginseng Res Vol. 36, No. 4, 354-368 (2012)
2000;16:1855-1868.
104. Cho YK, Jung YS, Sung H. Frequent gross deletion in
the HIV type 1 nef gene in hemophiliacs treated with
Korean red ginseng: inhibition of detection by highly ac-
tive antiretroviral therapy. AIDS Res Hum Retroviruses
2009;25:419-424.
105. Cho YK, Lim JY, Jung YS, Oh SK, Lee HJ, Sung H.
High frequency of grossly deleted nef genes in HIV-1
infected long-term slow progressors treated with Korean
red ginseng. Curr HIV Res 2006;4:447-457.
106. Cho YK, Jung YS. Dosage and duration effects of Ko-
rean red ginseng intake on frequency of gross deletions in
the nef gene. J Ginseng Res 2010;34:219-226.
107. Cho YK, Jung YS, Sung H, Sim MK, Kim YK. High
frequency of gross deletions in 5’ LTR/gag and nef genes
in patients infected with CRF02_AG of HIV type 1 who
survived for over 20 years: an association with Korean
red ginseng. AIDS Res Hum Retroviruses 2009;25:535-
541.
108. Cho YK, Jung YJ, Sung HS, Joo CH. Frequent genetic
defects in the HIV-1 5’LTR/gag gene in hemophiliacs
treated with Korean red ginseng: decreased detection of
genetic defects by highly active antiretroviral therapy. J
Ginseng Res 2011;35:413-420.
109. Cho YK, Jung YS. High frequency of gross deletions in
the 5’ LTR and gag regions in HIV type 1-infected long-
term survivors treated with Korean red ginseng. AIDS
Res Hum Retroviruses 2008;24:181-193.
110. Lee MH, Lee BH, Jung JY, Cheon DS, Kim KT, Choi
CS. Antiviral effect of Korean red ginseng extract and
ginsenosides on murine norovirus and feline calicivi-
rus as surrogates for human norovirus. J Ginseng Res
2011;35:429-435.
111. Bae EA, Shin JE, Park SH, Kim DH. Inhibitory effect of
ginseng polysaccharides on rotavirus infection. J Micro-
biol Biotechnol 2004;14:202-204.
112. O’Hagan DT, Valiante NM. Recent advances in the dis-
covery and delivery of vaccine adjuvants. Nat Rev Drug
Discov 2003;2:727-735.
113. McElrath MJ. Selection of potent immunological ad-
juvants for vaccine construction. Semin Cancer Biol
1995;6:375-385.
114. Singh M, O’Hagan DT. Recent advances in veterinary
vaccine adjuvants. Int J Parasitol 2003;33:469-478.
115. Song X, Hu S. Adjuvant activities of saponins from tra-
ditional Chinese medicinal herbs. Vaccine 2009;27:4883-
4890.
116. Liu CX, Xiao PG. Recent advances on ginseng research
in China. J Ethnopharmacol 1992;36:27-38.
117. Sun J, Hu S, Song X. Adjuvant effects of protopanaxa-
diol and protopanaxatriol saponins from ginseng roots
on the immune responses to ovalbumin in mice. Vaccine
2007;25:1114-1120.
118. Song X, Chen J, Sakwiwatkul K, Li R, Hu S. Enhance-
ment of immune responses to in uenza vaccine (H3N2)
by ginsenoside Re. Int Immunopharmacol 2010;10:351-
356.
119. Song X, Zang L, Hu S. Ampli ed immune response by
ginsenoside-based nanoparticles (ginsomes). Vaccine
2009;27:2306-2311.
120. Su F, Yuan L, Zhang L, Hu S. Ginsenosides Rg1 and
Re act as adjuvant via TLR4 signaling pathway. Vaccine
2012;30:4106-4112.
121. 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.
... In one study, glioma-bearing rats were injected through an IV route with EV-like nanoparticles isolated from ginseng (Panax ginseng, Gen-EVs). These formulations contained various bioactive compounds (i.e., miRNAs, proteins, lipids, ginsenosides) that incited an immune response [58]. Here, Kang and Min showed that ptc-miR396f-mediated targeting of genes related to apoptosis in Gen-EVs imparted significant gene silencing on proto-oncogene c-MYC, which resulted in tumor suppression and increased the survival rate of gliomabearing animals. ...
Article
Full-text available
Extracellular vesicles (EVs) have been explored as promising vehicles for drug delivery. One of the most valuable features of EVs is their ability to cross physiological barriers, particularly the blood–brain barrier (BBB). This significantly enhances the development of EV-based drug delivery systems for the treatment of CNS disorders. The present review focuses on the factors and techniques that contribute to the successful delivery of EV-based therapeutics to the brain. Here, we discuss the major methods of brain targeting which includes the utilization of different administration routes, capitalizing on the biological origins of EVs, and the modification of EVs through the addition of specific ligands on to the surface of EVs. Finally, we discuss the current challenges in large-scale EV production and drug loading while highlighting future perspectives regarding the application of EV-based therapeutics for brain delivery.
... Paeoniflorin (Paeonia japonica) and catalpol (Rehmannia glutinosa) regulate immune homeostasis by modulating inflammatory pathways such as MAPK/NF-κB and TLR-4 [49][50][51][52]. Ginsenosides (Panax ginseng) maintain the Th1/Th2 balance [53,54], while astragaloside IV (Astragalus mongholicus) enhances immune responses with anticancer potential [55,56]. Pachymic acid (Poria cocos) and glycyrrhizin (Glycyrrhiza uralensis) play protective roles in inflammation and sepsis by inhibiting NF-κB and caspase-11 activation [57][58][59]. ...
Article
Full-text available
Gagam-palmultang (PMT), a traditional Korean herbal formula, has been used to treat various conditions; however, its immunomodulatory potential remains unclear. Our objective was to assess the immunomodulatory effects of PMT in an immunosuppression mouse model. In vitro, we assessed interleukin (IL)-10 production in concanavalin A (Con A)-stimulated mouse splenocytes using real-time polymerase chain reaction and enzyme-linked immunosorbent assays. In vivo, C57BL/6 mice were intraperitoneally administered cyclophosphamide (CPA) to induce immunosuppression, followed by the oral administration of PMT (100 or 200 mg/kg) once daily for 14 days to evaluate its effects on T lymphocyte activation and immune function restoration. Immune function was evaluated via flow cytometric analysis of CD4⁺ and CD8⁺ T cells, thymus index measurements, thymic histopathology, and hematological analysis of white blood cell, monocyte, lymphocyte, neutrophil, and eosinophil counts. PMT significantly increased IL-10 production in Con A-stimulated splenocytes. In immunosuppressed mice, PMT restored the thymus index, improved thymic histopathology, and enhanced hematological parameters. Flow cytometry showed a significant increase in CD3e⁺CD4⁺ and CD3e⁺CD8⁺ T lymphocytes, and histological evaluation revealed increased CD3⁺ lymphocytes in the thymus. These findings suggest that PMT enhances T lymphocyte activation and restores immune homeostasis under immunosuppressive conditions, demonstrating its potential as a herbal immunomodulatory agent.
... This is further corroborated by the report of [16] who reported the stimulatory effect of Eucalyptus globulus essential oil on cell-mediated immune response after administration in immunosupressed rats. Furthermore, ginseng extracts and their EOs were shown to induce immunostimulatory effects by increasing the levels of tumor necrosis factor (TNFα) and IFNγ or enhancing phagocytic activity [18,[33][34]. ...
Article
Full-text available
Immunosuppression is a prominent critical feature associated with certain genetic disorders, chronic infections, and conventional chemotherapeutics employed in the treatment of diverse pathological conditions. The scrutiny for bioactive principles with immunostimulatory potential from natural sources becomes crucial. This study evaluated the immunomodulatory role of Phoenix dactylifera seed oil (PDSO) in cyclophosphamide-immunosuppressed Wistar rats. Essential oil from the date pit was obtained via steam-distillation technique. Cellular and humoral immunity were measured respectively using delayed type hypersensitivity reaction (DTHR) and hemagglutination reaction (HR), while quantitation of serum immunoglobulin was carried out using enzyme linked immunosorbent assay. The experimental rats were randomly assigned to 7 groups each (n = 4) for DTHR, and (n = 8) for HR assays. Immunosuppression was induced in the animals using cyclophosphamide and PDSO treatment was administered. PDSO significantly (P<0.05) promoted delayed type hypersensitivity response (DTHR), increased antibody titres and immunoglobulin levels in the experimental rats in a dose-related manner. The magnitude of immune stimulation evoked by the essential oil at 250 µl/kg.bw compared favourably with that exhibited by the positive control. Findings from this study showcased P. dactylifera seed oil as a promising immunostimulatory drug candidate with potential application in immunosuppressive conditions.
... T cells and increases IL-2 and IFN-γ production [34]. Similarly, GPs have consistently exhibited immunoregulatory properties in vitro. ...
Article
Full-text available
Panax vietnamensis, indigenous to Vietnam and southern China, is classified into three subspecies: Panax vietnamensis Ha et Grushv. (PVV), Panax vietnamensis var. fuscidiscus (PVF), and Panax vietnamensis var. langbianensis (PVL). A method to distinguish these varieties in their intact form is absent, which poses a possible risk of misclassification. Here, we aimed to devise a plant metabolite-based discrimination algorithm for the three varieties, without causing significant damage to individual plants. A multivariate analysis on mass spectral data of PVV, PVF, and PVL revealed that a peak at m/z 426, which was subsequently identified as an indole alkaloid glycoside, was exclusive to PVF and therefore clearly distinguished PVF from PVV and PVL. Additionally, global metabolic profiling was conducted to elucidate the discrimination markers between PVV and PVL, and lysophospholipids and hydroxy fatty acids were selected as potential discrimination markers. The performance of these markers was validated by cross-validation using machine learning algorithm.
Article
Ginseng, a well-known herbal supplement, is widely recognized for its pharmacological properties, including anti-inflammatory, antioxidant, and immune-modulatory effects. This review explores the potential therapeutic benefits of ginseng, particularly its active compounds, ginsenosides, in promoting nasal mucosa health. The nasal mucosa plays a crucial role in respiratory defense, acting as a barrier to pathogens and particulate matter, while also orchestrating immune responses. Ginseng's bioactive compounds have shown promise in modulating inflammation, reducing oxidative stress, and enhancing immune functions, which could be beneficial in conditions such as allergic rhinitis, chronic rhinosinusitis, and viral infections. Histological studies highlight the impact of ginseng on nasal mucosal cells, particularly in regulating immune responses and promoting tissue resilience. Research demonstrates that ginseng can reduce inflammation in the nasal passages by inhibiting pro-inflammatory cytokines and pathways like NF-κB, while enhancing the activity of immune cells such as natural killer cells and macrophages. Furthermore, ginseng's antioxidant properties help protect nasal tissue from oxidative damage, which is common in chronic nasal conditions. Although promising, the evidence base is still developing, with many studies limited by small sample sizes and variations in ginseng preparations. Further clinical trials are needed to substantiate ginseng's efficacy, optimal dosage, and delivery methods for treating nasal conditions. This review provides insights into the potential of ginseng as a complementary therapeutic approach for enhancing nasal mucosa health and improving respiratory outcomes.
Article
Full-text available
Korean red ginseng (RG), which is a ginseng treated by heating and steaming, has biological activity similar to Panax ginseng. The effect of ginseng on influenza infection has not been studied although it is known to have a broad range of biological activities. The aim of the study is to investigate the effect of RG extract on influenza A (H1N1) virus infection. We investigated the inhibitory effect of RG extract on plaque formation by influenza A virus in a cell-based plaque assay, and the effect of orally administered RG on influenza A virus infection in mice. RG extract, which was applied at a non-cytotoxic concentration, inhibited plaque formation by influenza A virus in the cell-based plaque assay. The orally administered RG extract ameliorated body weight loss and signifi cantly increased survival in mice infected with influenza A virus. Our results suggest that RG extract has components that reduce the severity of infection by influenza A virus and could potentially be used as a complement to treatment of influenza A virus infections.
Article
Full-text available
Allergies are immediate hypersensitive responses to antigens and interleukin (IL)-4 is involved in the initiation and development of allergic responses. has been known to have a variety of biological activities including anti-inflammatory activity, but the effect of on allergic responses is not known yet. The present study was undertaken to examine whether has an inhibitory effect on allergic response in mouse model. In allergic mouse model, our results showed that topical application of on atopic dermatitis (AD)-like skin lesions improved skin condition and inhibited starching behaviors. In addition, application not only suppressed mRNA expression of IL-4 and IL-10, but also prevented the nuclear factor of activated T cells 1 transcription. Moreover, application suppressed IL-4's secretion. Taken together, these results suggest that has a potent inhibitory effect in AD-related T cell cytokine production and may be a candidate for therapeutic agent in allergy.
Article
Full-text available
In the present study, we investigated whether a gross deletion in the nef gene () is induced by Korean red ginseng (KRG) intake. Ten patients were treated with KRG powder for 3 years in the absence of antiretroviral drug therapy. On average, KRG was administered per person over months. There was a mild decrease in CD4 T cell count () over the months (p = 0.059). We obtained 355 nef amplicons using 71 peripheral blood mononuclear cell samples over a 3-year period. All ten patients exhibited gnef (range, 3.2 to 45.9%). At baseline, 3 of 78 amplicons (3.8%) exhibited , whereas 18.8% (52/277) revealed during KRG-intake (p
Article
Full-text available
As much as the popularity of ginseng in herbal prescriptions or remedies, ginseng has become the focus of research in many scientific fields. Analytical methodologies for ginseng, referred to as ginseng analysis hereafter, have been developed for bioactive component discovery, phytochemical profiling, quality control, and pharmacokinetic studies. This review summarizes the most recent advances in ginseng analysis in the past half-decade including emerging techniques and analytical trends. Ginseng analysis includes all of the leading analytical tools and serves as a representative model for the analytical research of herbal medicines.
Article
Full-text available
Atopic dermatitis (AD) is an allergic, inflammatory skin disease characterized by chronic eczema and mechanical injury to the skin, caused by scratching. Korean red ginseng (RG) has diverse biological activities, but the molecular effects of RG on allergic diseases, like AD, are unclear. The present study was designed to investigate whether RG inhibits 1-chloro-2,4-dinitrobenzene (DNCB)-induced AD in a mouse model. DNCB was applied topically on the dorsal surface of Balb/c mice to induce AD-like skin lesions. We observed the scratching behavior and examined the serum IgE level and interleukin (IL)-4 and IL-10 in splenocytes compared with dexamethasone. We also evaluated the DNCB-induced mitogen-activated protein kinases (MAPKs), NF-κB, and Ikaros activities after RG treatment using reverse transcriptase-polymerase chain reaction, Western blotting, and ELISA. Our data showed that the topical application of RG significantly improved the AD-like skin lesions and scratching behavior. RG decreased not only the mRNA expression of IL-4 and IL-10, but also the secretion of IL-4 protein and serum IgE in mice. Additionally, RG treatment decreased the DNCB-induced MAPKs activity and subsequent Ikaros translocation irrespective of NF-κB. We suggest that RG may be useful as a therapeutic nutrition for the treatment of AD.
Article
Polysaccharides and saponins were isolated from the root of Panax ginseng C.A. Meyer (Family Araliaceae), treated at various temperatures, and their inhibitory effects on rotavirus were investigated. As the temperature of processing increased, the molecular weight of the polysaccharides decreased, but the yields of water extracted increased. These polysaccharides inhibited rotavirus infection in MA104 cells, but there were no significant differences in rotavirus infection-inhibitory potency. However, ginseng saponins did not exhibit rotavirus infection-inhibitory activity.
Article
For many years, innate immunity has been considered as a separate entity from the adaptive immune response and has been regarded to be of secondary importance in the hierarchy of immune functions. For the past few years, however, interest in innate immunity has grown enormously, so that now it is studied intensively in many laboratories that seek to integrate these two distinct types of immune function. Our intent in this review is to point out the similarities and differences in these two types of host response to infection, and to indicate our present level of understanding of how these can be integrated into a more complete description of the immune response.