Skip to main content
CNS Neuroscience & Therapeutics logoLink to CNS Neuroscience & Therapeutics
. 2010 Dec 8;17(6):761–768. doi: 10.1111/j.1755-5949.2010.00208.x

Ginsenosides and Their CNS Targets

Khaled Radad 1, Rudolf Moldzio 2, Wolf‐Dieter Rausch 2
PMCID: PMC6493809  PMID: 21143430

SUMMARY

Ginsenosides are a special group of triterpenoid saponins attributed to medical effects of ginseng. Therefore, they have been research targets over the last three decades to explain ginseng actions and a wealth of literature has been presented reporting on ginsenosides’ effects on the human body. Recently, there is increasing evidence on beneficial effects of ginsenosides to the central nervous system (CNS). Using a wide range of in vitro and in vivo models, researchers have attributed these effects to specific pharmacological actions of ginsenosides on cerebral metabolism, oxidative stress and radical formation, neurotransmitter imbalance and membrane stabilizing effects, and even antiapoptotic effects. Modulating these particular mechanisms by ginsenosides has thus been reported to exert either general stimulatory effects on the brain functions or protecting the CNS against various disease conditions. In this review, we try to address the recently reported ginsenosides’ actions on different CNS targets particularly those supporting possible therapeutic efficacies in CNS disorders and neurodegenerative diseases.

Keywords: CNS targets, Ginsenosides, Neurodegenerative diseases, Panax ginseng

Introduction

Ginseng as a name refers to a group of slowly growing plants with fleshy roots belonging to the genus Panax in the family Araliaceae. Among them, Panax ginseng is the most famous and precious herbal medicine consumed worldwide [1]. Ginsenosides are unique triterpenoid saponins found exclusively in Panax species and up to now more than 150 naturally occurring ginsenosides have been isolated from roots, leaves, stems, fruits, and/or flower heads of ginseng [2, 3, 4]. All ginsenosides have a common four‐ring hydrophobic steroid‐like structure but with a different number of sugar moieties [5]. According to the number and position of these sugar moieties, ginsenosides are classified into two major groups: 20(S)‐protopanaxadiol (PD) and 20(S)‐protopanaxatriol (PT) saponins. The diversity of the sugar components between ginsenosides might responsible for the specific action of each ginsenoside [6, 7].

After discovering ginseng as a medical plant in Manchuria (China) over 5000 years ago, its whole roots were first taken as a food and 2000 years later, early Chinese used ginseng as a general tonic and adaptogen helping the body to resist the adverse influence of a wide range of physical, chemical, and biological factors and to restore homeostasis [8, 9]. Since the middle of the last century, scientists in Asian and Western countries have paid great attention to ginseng research, which led to isolation and identification of ginsenosides which have been found to produce a wide range of actions on different body systems. Recently, ginsenosides have been shown to produce a number of beneficial effects in the CNS [10]. These actions of ginsenosides are attributed to specific pharmacological effects on particular CNS targets. Modulation of these targets by ginsenosides found to stimulate the brain functions and induced protection against various disease conditions. This review was designed to address and discuss the recent reports dealt with effects of ginsenosides on different CNS targets particularly those that are associated with therapeutic efficacies in CNS disorders and neurodegenerative diseases.

Ginsenosides‐Modulated CNS Targets Associated With Potentiating of Brain Functions

Ginsenosides Promote Neurogenesis

The dogma that mammalian CNS is an organ unable to regenerate has been challenged about 20 years ago when Reynolds and Weiss [11] published their pioneering work in Science about proliferation and differentiation of cells isolated from the striatum of adult mouse into neurons and astrocytes in vitro. Nowadays, it is fully accepted that adult neurogenesis occurs in the subgranular zone (SGZ) in the hippocampus [12] and the subventricular zone (SVZ) of the lateral ventricle [13]. Compounds that stimulate adult neurogenesis might protect against deterioration of brain functions, most notably memory and learning capabilities, due to aging or adverse conditions. In this respect, it was reported that ginsenoside Rg1increased the proliferating ability of neural progenitors both in vitro and in vivo under both physiological and pathological circumstances. For example, Shen and Zhang [14, 15] found that ginsenoside Rg1 increased the number of proliferating progenitor cell spheres and the number of dividing cells in the hippocampus when incubated with neural proliferating cells and following intraperitoneal administration in adult mice. Cheng et al. [16] and Shen and Zhang [17] reported that ginsenoside Rg1 increased the extent of ischemia‐induced proliferation and differentiation of neural progenitor cells in the dentate gyrus of the hippocampus in ischemic gerbils. The authors related this effect to nitric oxide (NO) and N‐methyl‐d‐aspartate (NMDA) receptors as they found that ginsenoside Rg1 enhanced inducible nitric oxide synthase (iNOS) activity in the hippocampus of ischemic gerbils and systemic injection of MK‐801, NMDA receptor antagonist, completely blocked the proliferating effect of ginsenoside Rg1. Liu et al. [18] found that ginsenoside Rg3 induced neurons in neural stem cells (NSCs). Blocking of such an effect with nifedipine, a Ca2+ channel antagonist, argues for involvement of voltage‐dependent Ca2+ channel in ginsenoside Rg3‐induced neurogenesis.

Ginsenosides Affect Neurotransmission

Neurotransmission is the process of neurotransmitter release from synapses where these compounds act on postsynaptic receptors on other neurons or cells in the CNS. Such processes are essential for propagation of signals from and to CNS via efferent and afferent neurons to further control smooth, skeletal and cardiac muscles, glandular secretions, and organ functions. Ginsenosides Rb1 and Rg1 have been reported to increase the release of the excitatory neurotransmitter glutamate in undifferentiated and differentiated PC12 cells [19]. The authors found that ginsenoside Rb1 promoted glutamate release by increasing phosphorylation of synapsins through cyclic adenosine monophosphate (cAMP)‐dependent protein kinase (PKA). Synapsins are a family of proteins that are implicated to regulate neurotransmitter release at synapses. As a response of phosphorylation of synapsins, cytoskeleton‐bound synaptic vesicles are released and move to the synaptic membrane and release their neurotransmitters into the synaptic cleft. Ginsenosides appear to enhanced central cholinergic function. In this respect, Salim et al. [20] found that ginsenoside Rb1 increased the expression of choline acetyltransferase in the rat's basal forebrain, the enzyme that facilitates the formation of acetylcholine (Ach) via binding acetyl‐CoA to choline. Benishin [21] reported that ginsenoside Rb1 boosted Ach release from hippocampal slices of rats. Moreover, ginseng total saponins (GTS) were found to increase the dopamine in the cerebral cortex, modulate dopaminergic activity at both presynaptic and postsynaptic receptors in mice [22, 23], increase serotonin in rat cortices [24], raise the level of biogenic amines in normal rat brains [25], interact with nicotinic receptor subtypes [26] and regulate transmission of γ‐aminobutyric acid (GABA) in animals [27, 28]. Facilitating neurotransmitter release by ginsenosides may contribute to enhancing brain functions particularly learning, cognition, and memory.

Ginsenosides Support Memory and Learning

Cognitive deficits involving impairment of memory and learning are considered one of the most important consequences of aging [29]. Thus, searching for drugs that can prevent or even retard age‐related cognitive deficits would be important for healthy aging. Ginsenosides were reported to improve cognitive function in aged animals and animal models of memory impairment. For instance, Wang et al. [30] reported that GTS facilitated learning and memory in normal Wistar rats. Wang et al. [30] and Yang et al. [31] found that ginsenoside Rh1 and Rh2 significantly ameliorated scopolamine‐induced memory impairment and learning deficit in mice, respectively. They concluded that ginsenosides enhanced cognitive functions including memory and learning through targeting synapses. In this context, Wang and Zhang [32] reported that ginsenoside Rg1 enhanced the basic synaptic transmission and the magnitude of long‐term potentiation (LTP) in the dentate gyrus of rats. Mook‐Jung et al. [33] found that ginsenosides Rb1 and Rg1 increased hippocampal synaptic density as treated mice with ginsenosides contained higher density of a synaptic marker protein, synaptophysin, compared to control mice. Also, Zhao et al. [34] observed that a number of ginsenosides including Ra, Rb1, Rb2, Rc, Rd, Re, Rf, Rg1, Rg2, Rg3, and Ro were effective to prevent memory impairment by upregulating plasticity‐related proteins in the hippocampus of 12 months old C57BL16 mice. Modulation of synaptic plasticity is considered one of the important molecular mechanisms of learning and memory.

Ginsenosides Affects and Regulate Ion Channels in Neurons

Neuronal ion channels are gated pores on cellular membranes. They are often selectively permeable to ions such as calcium (Ca2+), potassium (K+), or sodium (Na+). The opening and closing of these channels are either voltage‐ or ligand‐dependent.

Voltage‐Dependent Ion Channels

Voltage‐Dependent Ca2+ Channels

There are five different voltage‐dependent Ca2+ channel subtypes such as L‐, N‐, P/Q‐ and T‐type [35]. Excessive stimulation of voltage‐dependent Ca2+ channels results in intracellular Ca2+ overload that leads to production of oxygen radicals and triggers the activation of various enzymes which are harmful to cells [36]. There is some evidence showing that ginsenosides can inhibit Ca2+ channels in neuronal cells. For example, Kim et al. [37] observed that GTS reduced KCl‐induced neuronal loss in primary cortical neurons through inhibition of L‐type Ca2+ channels. Similarly, Zhang et al. [38] found that ginsenoside Rg1 inhibited Ca2+ influx through L‐type Ca2+ channels in primary hippocampal neurons.

Voltage‐Dependent K+ Channels

Neurons like other cells have different types of K(+)‐selective ion channels including voltage‐dependent, Ca2+‐activated, ATP‐sensitive, and G‐protein‐coupled inwardly rectifying (GIRK) channels. They are critical determinants of membrane excitability in neuronal cells as they set the resting membrane potential, oppose depolarization, and repolarize action potential [39]. Some ginsenosides were reported to regulate the electrical state of excitable neurons by activation of K+ channels. For example, ginsenoside Rf activated GIRK channels through an unidentified G protein‐coupled receptor in rat's brain [40].

Voltage‐Dependent Na+ Channels

Voltage‐dependent Na+ channels play a primary role in neuronal excitability and are critical target for neuromodulation [41]. There is some evidence on the regulation of neuronal Na+ channels by ginsenosides. Kim et al. [42] reported that ginsenoside Rg3's carbohydrate component inhibited the inward Na+ peak current in Xenopus oocytes. Lee et al. [43] observed that ginsenoside Rg3 inhibited Na+ in Xenopus oocytes through acting on the resting and open states of Na+ channel via interaction with the S4 voltage‐sensor segment of domain II. Duan and Nicholson [44] reported that ginsenoside Rh2 inhibited veratridine‐dependent depolarization (inhibition of Na+ channel function) of mouse synaptoneurosomes with subsequent inhibition of release of l‐glutamate and GABA. Liu et al. [45] reported that American ginseng (Panax quinquefolius) and ginsenoside Rb1 blocked ischemia‐induced Na+ fluxes in human embryonic kidney tsA201 cells through interaction with the inactive state of the channel.

Ligand‐Dependent Ion Channels

The common ligand‐dependent ion channels (ionotropoic receptors) include NMDA‐, nicotine acetylcholine‐, and serotonin‐gated ion channels.

NMDA‐Gated Ion Channels

Activation of NMDA receptors by glutamate, the main excitatory neurotransmitter released extensively in neurological diseases and disorders such as Alzheimer disease (AD), ischemia, seizures, and trauma of head and neck, results in massive influx of Ca2+ into neurons. Intracellular Ca2+ overload is a key component of glutamate‐mediated neurotoxicity [46]. Modulation of NMDA receptors by ginsenosides has been reported in some CNS disorders. For example, ginsenosides Rb1 and Rg3 have been reported to protect rat cortical and hippocampal neurons against glutamate‐ and NMDA‐induced neurotoxicity, respectively, by deactivation of NMDA receptors [47, 48]. Precisely, it was reported that ginsenosides inhibited NMDA receptors via interaction with different regulatory sites such as NMDA‐, co‐agonist glycine‐, polyamine‐binding sites, and sites within the lumen of the channel. In this context, Kim et al. [49] and Lee et al. [50] found that ginsenosides Rg3 and Rh2 selectively targeted NMDA receptor in primary hippocampal neurons via interaction with glycine‐ and polyamine‐binding sites, respectively.

Nicotine Acetylcholine Ligand‐Gated Ion Channels

Neuronal nicotinic receptors consist of α (α2 –α9) and β (β2 –β4) subunits. Activation of these receptors by ACh allows the influx of Na+ into the cells. Various reports showed the ability of ginsenosides to regulate nicotine ACh receptor channels. Tachikawa et al. [51] reported that ginsenosides Rb1, Rb2, Rb3, Rc, Rd, Re, Rf, Rg1, Rh1, Ro, and Rs1 reduced ACh‐evoked secretion of catecholamines from bovine adrenal chromaffin cells which contain mainly α3β4 subunits of nicotinic receptors. Moreover, Choi et al. [52] found that ginsenosides Rg2, Rf, Re, Rg1, Rc, Rb2, and Rb1 inhibited acetylcholine‐induced inward currents in Xenopus oocytes expressed with nicotinic receptors containing α1β1δɛ or α3β4 subunits.

Serotonin‐Gated Ion Channels

Among serotonin (5‐hydroxytryptamine, 5‐HT) receptors, activation of 5‐HT3 receptor renders it permeable to Na+ and K+ ions. Ginsenoside Rg2 and ginsenoside metabolites such as compound K and M4 were reported to inhibit the 5‐HT3 receptor‐gated ion currents in Xenopus oocytes expressing 5‐HT3A receptors [53, 54]. Lee et al. [55] connected the effect of ginsenosides on 5‐HT receptors to the interaction with residues V291, F292, and I295 in the channel gating region of transmembrane domain 2 (TM2). The authors found that mutations of V291, F292, and I295 in the channel gating region of TM2 greatly attenuated or abolished ginsenoside Rg3‐induced inhibition of peak I 5‐HT.

Ginsenosides‐Modulated CNS Targets Associated with neuroprotection

Parkinson Disease

Parkinson disease (PD) is a progressive neurodegenerative disease affecting 0.3% of the general populations worldwide. The main symptoms of PD are caused as the result of the progressive degeneration of the nigrostriatal dopaminergic pathway. Most of current therapies only provide symptomatic treatment and up till now, no drug has yet been found to prevent the progressive loss of dopaminergic neurons in PD patients [56]. Recently, it has been shown that ginseng and its active ingredients, ginsenosides, have beneficial effects on both in vitro and in vivo PD models. Ginsenosides, through involving antiapoptotic, antioxidant, and anti‐inflammatory mechanisms, have been reported to protect nigrostriatal system in animal models. For example, Xu et al. [57] reported that ginsenoside Re rescued dopaminergic neurons in 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine (MPTP)‐treated mice through upregulating the expression of Bcl2 protein and downregulating the expression of Bax and iNOS proteins and inhibiting activation of caspase‐3. Chen et al. [58] found that attenuation of glutathione (GSH) reduction and the phosphorylation of JNK and C‐Jun by ginsenoside Rg1 protected against MPTP‐induced dopaminergic cell loss in C57‐BL mice. Wang et al. [59] found that ginsenoside Rg1 rescued dopaminergic neurons in MPTP‐treated mice through P38 signaling pathway that plays an important role in modulating cyclooxygenase‐2 (COX‐2) expression.

Moreover, Wang et al. [60] reported that ginsenoside Rg1 increased dopamine and its metabolites in the striatum and tyrosine hydroxylase (TH) expression in MPTP‐treated mice by upregulating certain iron transporter proteins such as iron import protein, divalent metal transporter (DMT1) and iron export protein, ferroportin 1(FP1). Elevated iron levels in substantia nigra (SN) may play a key role in the development of PD by enhancing the generation of oxidative stress [61, 62]. Xu et al. [63] reported that activation of the insulin‐like growth factor‐1 (IGF‐1) mediated the neuroprotective effects of ginsenoside Rg1 in 6‐hydroxydopamine (6‐OHDA)‐treated rats. This neuroprotective effect of ginsenoside Rg1 was blocked by IGF‐1 receptor antagonist, JB‐1.

Similar to their effects on animal models, ginsenosides have been also found to protect in vitro cellular PD models through antiapoptotic, antioxidant, and anti‐inflammatory mechanisms. For instance, it was reported that ginsenosides Rg1 attenuated rotenone‐induced apoptosis in primary cultured rat nigral neurons by inhibiting the mitochondrial apoptotic pathway [64]. This effect of ginsenoside Rg1 was mediated by prevention of cytochrome c release from mitochondrial membrane and an increase in the inhibition of phosphorylation of the proapoptotic protein Bad through activation of the PI3K/Akt pathway. Also, the protective effect of ginsenoside Rg1 was blocked by glucocorticoid receptor (GR) antagonist RU486, indicating that the action of Rg1 is mediated through GR activation [64]. Regarding antioxidant activity, Ye et al. [65] concluded that ginsenoside Rd played a potent antioxidant role as it reduced intracellular reactive oxygen species (ROS) levels, decreased malondialdehyde (MDA) production, the common index of lipid peroxidation, and enhanced the antioxidant enzymatic activities of superoxide dismutase (SOD) and glutathione peroxidase (GPx) in hydrogen peroxide (H2O2)‐treated PC12 cells. Against lipopolysaccharides (LPS)‐induced neuroinflammation in primary dopaminergic cell culture, ginsenoside Rd was found to be effective through reduction of NO formation and PGE2 synthesis [66].

Alzheimer's Disease

Alzheimer's disease (AD), the most common cause of dementia in elderly people, has no cure so far. According to the current research, promising therapeutic strategies for AD include reduction of generation or aggregation of β amyloid peptide (Aβ), enhancement removal of Aβ from the cells, interruption of τ hyperphosphorylation and the use of more efficacious antioxidant and anti‐inflammatory drugs. Ginsenosides have been recently reported to produce protective and trophic effects against AD. Yang et al. [67] reported that ginsenoside Rg3 significantly reduced the levels of Aβ 40 and 42 in SK‐N‐SH cells transfected with Swedish mutant SweApp through enhancing neprilysin (NEP) gene expression, the rate‐limiting enzyme in the Aβ degradation in the brain. Moreover, ginsenoside Rg3 was found to promote Aβ uptake, internalization and digestion by microglia in rat primary culture and this effect may be related to type A macrophage scavenging receptor (MSRA) expression [68].

There is large evidence indicating that phosphorylated τ plays a prominent role in the pathogenesis of AD [69]. Xie et al. [70] reported that pretreatment of hippocampal neurons with ginsenoside Rb1 reduced hyperphosphorylation of τ protein induced by Aβ25–35. Chen et al. [71] found that ginsenoside Rb1 decreased τ phosphorylation in cortical neurons through inhibiting p35 expression, the cyclin‐related activator molecule that regulates cyclin‐dependent kinase‐5 (CDK5) which, upon activation, results in τ phosphorylation.

Moreover, Shieh et al. [72] found that ginsenoside Rh2 could attenuate Aβ‐induced apoptosis in type I rat brain astrocytes through increasing expression of the neurotrphic factor, pituitary adenylate cyclase activating polypeptide (PACAP). Chen et al. [73] reported that ginsenoside Rg1 attenuated Aβ1–40‐induced apoptosis in rat cortical neurons via inhibiting the activity of cyclin‐dependent kinase‐4 (CDK4), decreasing the phosphorylation of pRB and downregulation the expression of E2F1 mRNA. Wei et al. [74] found that ginsenoside Rg1 prevented Chinese hamster ovarian (CHO) tumor cells transfected with mutant PSIM146L gene from apoptosis through inhibiting the production of Aβ 42 and decreasing the expression of the active caspase‐3. Furthermore, it was found that ginsenosides did not only reduced Aβ formation and Aβ‐induced hyperphosphorylation of τ and neuronal apoptosis but they also found to affect the activity of choline acetyltransferase and acetylcholinestrase. In parallel, Wang et al. [75] reported that ginsenoside Rg1 significantly ameliorated the learning and memory impairment induced by β‐AP(25–35). The authors returned this effect of ginsenoside Rg1 to the prevention of cortical and hippocampal choline acetyltransferase activity decline and inhibition of the activity of acetylcholinestarse. It is of importance to know that central acting cholinesterase inhibitors are the first primary pharmacological treatment approved for AD.

Amyotrophic Lateral Sclerosis

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by degeneration of motoneurons in the spinal anterior horn [76]. Using mutant SOD1 transgenic mice (B6SJL‐TgN(SOD‐1G93A)1Gur), the relevant animal model for ALS, it was shown that crude ginseng powder significantly delayed the onset of signs of motor impairment and prolonged the survival of mice [77]. In this context, Kim et al. [78] reported that ginsenoside Rb2 significantly activated SOD1 gene through transcription factor AP2‐binding site in human HepG2 hepatoma cells.

Huntington's Disease

Huntington's disease (HD) is an inherited progressive neurodegenerative disorder originated by the mutation of the gene encoding the huntingtin‐protein (htt). The disease is characterized clinically by involuntary abnormal movements, cognitive deficits, and psychiatric disturbance and pathologically by degeneration of the GABAergic medium size spiny neurons [79]. In primary medium spiny striatal neuronal culture (MSN) from the YAC128 HD mouse, an in vitro model for HD, ginsenosides Rb1, Rc, and Rg5 protected cultured cells from glutamate‐induced apoptosis through inhibiting glutamate‐induced Ca2+ responses [80]. HD mutation affected Ca2+ signaling in MSN cultures by stimulating NR1/NR2B subunit of NMDA receptors [81]. Moreover, post‐mortem tissue from HD patients showed reduced binding to glutamate receptors in the striatum [82, 83] and decreased mRNA levels of the NR1 and NR2 subunits suggesting the loss of neurons containing NMDA receptors [84].

Brain Ischemia

Brain ischemia, the most important cause of chronic disability, is caused by low oxygen and glucose supply and a decrease in adenosine triphosphate (ATP) in the brain. Most of the recent studies support the role of oxidative mechanisms and energy depriving injuries in cerebral ischemia as the brain has low concentrations of oxygen radical scavenging enzymes and low fuel reserves, respectively [85]. Some ginsenosides have been reported to target different injurious factors in the ischemic cascade. For example, ginsenoside Rb1 was reported to prevent ischemic neuronal death in rats and protect ischemic CA1 neurons in gerbils through scavenging free radicals and increasing expression of antiapoptotic genes and modulating the expression of glial‐derived neurotrophic factor (GDNF), respectively [86, 87]. Moreover, ginsenosides Rb and RO protected against cerebral ischemia/reperfusion injury in rats by facilitating synthesis and release of prostacyclin in the brain and reducing formation of thromboxane A2 [88]. Also, ginsenosides Re and Rg3 were found to show neuroprotection against cerebral ischemia/reperfusion injury in rats by stabilizing mitochondria and enhancing the activities of SOD and GPx [89, 90, 91]. Similar to animal models, ginsenosides Rg1 and Rd reduced neuronal death of primary rat hippocampal neurons induced by oxygen glucose deprivation by activation of GR and inhibition of calcium influx through NMDA receptors and L‐type voltage‐dependent Ca2+ channels and by enhancing the antioxidant activities of catalase, SOD, and GPx, respectively [38, 92].

Neuroinflammation

Following neural injury, microglia the resident immune cells of the CNS is activated and releases various neurotrophic factors such as BDNF, NT‐3, and NGF. Upon over‐activation, microglia releases neurotoxic factors, most notably NO and proinflammatory cytokines, which play an important role in the progression of neurodegenerative diseases such as AD and PD [93, 94]. Ginseng extract and its saponins have been reported to protect neuronal cells against microglia‐induced inflammatory processes. In this context, it has been reported that ginsenosides Rd, Rg1, Re, Rg3, and Rh2 reduced cytokines production by LPS‐ and Aβ42‐stimulated microglial cells [66, 95, 96, 97]. Using immortalized murine BV2 microglial cell line, Park et al. [98] found that inhibition of NF‐κB and MAP kinase, the upstream signaling inflammatory molecules, may be correlated with the anti‐inflammatory effects of ginseng. Moreover, He and Zhu [99] reported that Panax notoginseng saponins (PNS) attenuated the ischemic brain inflammation through decreasing expression of ICAM‐1 and subsequent inhibition of neutrophil infiltration.

Brain Tumors

Among ginsenosides, ginsenoside Rh2 and compound K were reported to inhibit the in vitro invasiveness of astroglioma cells. Such effect was found to be mediated by inhibition of matrix metalloproteinase‐9 (MMP‐9), the enzyme that degrades extracellular matrix and consequently plays an important role in facilitating infiltration, migration, and invasion of gliomas [100, 101]. Using PMA‐treated astroglioma cells, the authors found that ginsenoside Rh2 and compound K, 20‐O‐β‐(d‐glucopyranosyl)‐20(S)‐protopanaxadiol derived from protopanaxadiol ginsenosides by intestinal bacteria, repressed the expression of MMP‐9 through the inhibition of NF‐κB, AP‐1 and P38MAPK, ERK, and JNK. Moreover, it was reported that ginsenoside Rh2 inhibited cell growth in MCF‐7 human breast cancer cells and SK‐HEP‐1 hepatoma cells [102, 103] and induced apoptosis in various cell lines including C6 rat glioma, SK‐N‐BE(2) C human neuroblastoma, and A375‐S2 human malignant melanoma cells [104, 105, 106]. Also, Surh et al. [107] and Park et al. [108] observed that ginsenoside Rg3 inhibited the formation of PMA‐induced mouse skin tumors.

The most important effects of ginsenosides Rg1, Rg3, Rg2, Rb1, Rb2, Rf, Rh2, Re and Rd on different CNS targets are summarized in Table 1.

Table 1.

Important ginsenosides’ effects following activation of some CNS targets

Ginsenosides CNS targets Effects
Rg1 iNOS and NMDA receptor Proliferation and differentiation of neuronal progenitor cells in dentate gyrus of the hippocampus in ischemic gerbils [16, 17].
Synapses Increasing cognitive functions such as memory and learning through enhancing the basic synaptic transmission and the magnitude of LTP in the dentate gyrus of rats [32].
L‐type Ca2+ channels Inhibition of Ca2+ influx into primary cultured hippocampal neurons [38].
P38 Modulation of COX‐2 expression with subsequent protection of dopaminergic neurons in MPTP‐treated mice [59].
DMT1 and FPI Increasing dopamine and its metabolites in the striatum and TH expression in MPTP‐treated mice [60].
IGF‐1 Protection of dopaminergic neurons in 6‐OHDA‐treated rats [63].
GR Restoring of ΔΨm, prevention the release of cytochrome c from mitochondrial membrane and increasing the phosphorylation inhibition of the pro‐apoptotic protein Bad through activation of P13K/Akt pathways in rotenone‐treated primary nigral cell culture [64].
CDK4, pRB, and E2F1 Attenuation of Aβ‐40‐induced apoptosis in rat cortical neurons [73].
Rg3 Voltage‐dependent Ca2+ channels Neuronal induction in neural stem cells (NSCs) [18].
Glycine binding site of NMDA receptor Protection of rat hippocampal neurons against NMDA‐induced neurotoxicity [49].
Neprilysin gene Reduction the levels of Aβ 40 and 42 in SK‐N‐SH cells transfected with Swedish mutant SweApp [67].
MSRA Promotion of uptake, internalization and digestion of Aβ by microglia in rat primary culture [68].
Rg2 α1β1δɛ or α3β4 subunits of NMDA receptor Inhibition of acetylcholine‐induced inward currents in Xenopus oocytes [52].
V291, F292, and I295 residues of 5‐HT(3A) receptor channels Inhibition of 5‐HT3 receptor‐gated ion currents in Xenopus oocytes [55].
Rb1 (cAMP)‐dependent PKA Phosphorylation of synapsins with subsequent release of glutamate in PC12 cells [19].
Choline acetyltransferase Formation of Ach in rat's basal forebrain [20].
NMDA receptor Protection of rat cortical neurons against glutamate neurotoxicity [47].
α3β4 subunits of nicotinic receptors Reduction of Ach‐evoked secretion of catecholamines from adrenal chromaffin cells [51].
P35 Decreasing hyperphosphorylation of τ protein induced by Aβ 25–35 in cortical neurons [71].
Rb2 Transcription factor AP2 binding site Activation of SOD1 gene in human HepG2 hepatoma cells [78].
Rf Unidentified G protein coupled receptor Activation of GIRK channels in rat's brain [40].
Rh2 Na+ channels Inhibition of L‐glutamate and GABA release in mouse synaptoneurosomes [44].
Polyamine binding site of NMDA receptor Protection of hippocampal neurons against glutamate and NMDA neurotoxicity [50].
PACAP Attenuation of Aβ‐induced apoptosis in type I rat brain astrocytes [72].
NF‐κB and MAP kinase Inhibition of microglial activation in immortalized murine BV2 microglial cell line [98].
Suppression of MMP‐9 in PHA‐treated astroglioma cells [100, 101].
Re Bcl2, Bax, iNOS. and caspase‐3 Rescuing dopaminergic neurons in MPTP‐treated mice [57].
Rd iNOS and COX‐2 Reduction of NO formation and PGE2 synthesis in LPS‐treated dopaminergic neurons [66].
GR Reduction of neuronal death of primary rat hippocampal neurons induced by oxygen glucose deprivation [92].
PNS ICAM‐1 Attenuation of ischemic brain inflammation and inhibition of neutrophil infiltration [99].

Concluding Remarks

Ginseng's history can be divided into three eras. The first era was when the whole ginseng's roots were primarily used as a food by earlier Chinese. The second era was 2000 years later when ginseng was discovered to act as a tonic and adaptogen. The third one has begun since identification and isolation of ginsenosides in the middle of the last century. From that time, ginseng has been attracted much attention by researchers worldwide and hundreds of reports that described the beneficial effects of ginseng have been published worldwide. Most of these publications have been precisely described the mechanistic actions of different ginsenosides on various body systems using cellular and animal models. Obtained data are still not validated for patients against a particular disease due to shortage in clinical testing and validation. This review addressed the effect of the most common ginsenosides effects on different CNS targets and to try to prompt researchers in the future to analyze ginsenosides’ actions using modern analytical tools particularly in clinical trials.

Conflicts of Interest

The authors declare no conflict of interests.

References

  • 1. Tyler VE. Herbal remedies. J Pharm Technol 1995;11:214–220. [Google Scholar]
  • 2. Liu CX, Xiao PG. Recent advances on ginseng research in China. J Ethnopharmacol 1992;36:27–38. [DOI] [PubMed] [Google Scholar]
  • 3. Baek NI, Kim DS, Lee YH, Park JD, Lee CB, Kim SI. Ginsenoside Rh4, a genuine dammarane glycoside from Korean red ginseng. Planta Med 1996;62:86–87. [DOI] [PubMed] [Google Scholar]
  • 4. Christensen LP. Ginsenosides chemistry, biosynthesis, analysis and potential health effects. Adv Food Nutr Res 2009;55:1–99. [DOI] [PubMed] [Google Scholar]
  • 5. Chen RJY, Chung T, Li F, Lin N, Tzen JTC. Effect of sugar positions in ginsenosides and their inhibitory potency on Na+/K+‐ATPase activity. Acta Pharmacol Sin 2009;30:61–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Nah SY, Park HJ, McCleskey EW. A trace component of ginseng that inhibit Ca2+ channels through a pertussis toxin‐sensitive G protein. Proc Natl Acad Sci USA 1995;92:8739–8743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Choi S, Kim HJ, Ko YS, et al G alpha(q/11) coupled tomammalian phospholipase C beta 3‐like enzyme mediates the ginsenoside effect on Ca2+‐activated Cl−current in the Xenopus oocyte. J Biol Chem 2001;276:48797–48802. [DOI] [PubMed] [Google Scholar]
  • 8. Brekhman I, Dardymov I. New substances of plant origin which increase non specific resistance. Ann Rev Pharmacol 1969;9:419–430. [DOI] [PubMed] [Google Scholar]
  • 9. Nocerino E, Amato M, Izzo AA. The aphrodisiac and adaptogenic properties of ginseng. Fitoterapia 2000;71:1–5. [DOI] [PubMed] [Google Scholar]
  • 10. Van Kampen J, Robertson H, Hagg T, Drobitch R. Neuroprotective actions of the ginseng extract G115 in two rodent models of Parkinson's disease. Exp Neurol 2003;184:21–29. [DOI] [PubMed] [Google Scholar]
  • 11. Reynolds BA, Weiss S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science 1992;5052:1707–1710. [DOI] [PubMed] [Google Scholar]
  • 12. Eriksson PS, Perfilieva E, Bjork‐Eriksson T, Alborn A, Nordborg C, Peterson DA, Gage FH. Neurogenesis in the adult human hippocampus. Nat Med 1998;4:1313–1317. [DOI] [PubMed] [Google Scholar]
  • 13. Johansson CB, Momma S, Clark DL, Risling M, Lendahl U, Frisen J. Identification of a neural stem cell in the adult mammalian brain. Cell 1999;96:25–34. [DOI] [PubMed] [Google Scholar]
  • 14. Shen LH, Zhang JT. Effects of ginsenoside Rg1 on neural progenitor's proliferation in vitro and in vivo In: Baek NI, editors. Advances in ginseng research. Seoul: The Korea Society of Ginseng, 2002;522–530. [Google Scholar]
  • 15. Shen LH, Zhang JT. Ginsenoside Rg1 promotes proliferation of hippocampal progenitor cells. Neurol Res 2004;26:422–428. [DOI] [PubMed] [Google Scholar]
  • 16. Cheng Y, Shen LH, Zhang JT. Anti‐amnestic and anti‐aging effects of ginsenoside Rg1 and Rb1 and its mechanism of action. Acta Pharmacol Sin 2005;26:143–149. [DOI] [PubMed] [Google Scholar]
  • 17. Shen L, Zhang J. NMDA receptor and iNOS are involved in the effects of ginsenoside Rg1 on hippocampal neurogenesis in ischemic gerbils. Neurol Res 2007;29:270–273. [DOI] [PubMed] [Google Scholar]
  • 18. Liu JW, Tian SJ, de Barry J, Luu B. Panaxadiol glycosides that induce neuronal differentiation in neurosphere stem cells. J Nat Prod 2007;70:1329–1334. [DOI] [PubMed] [Google Scholar]
  • 19. Xue JF, Liu ZJ, Hu JF, Chen H, Zhang JT, Chen NH. Ginsenoside Rb1 promotes neurotransmitter release by modulating phosphorylation of synapsins through a cAMP‐dependent protein kinase pathway. Brain Res 2006;1106:91–98. [DOI] [PubMed] [Google Scholar]
  • 20. Salim KN, McEwen BS, Chao HM. Ginsenoside Rb1 regulates ChAT, NGF and trkA mRNA expression in the rat brain. Brain Res Mol Brain Res 1997;47:177–182. [DOI] [PubMed] [Google Scholar]
  • 21. Benishin CG. Actions of ginsenoside Rb1 on choline uptake in central cholinergic nerve endings. Neurochem Int 1992;21:1–5. [DOI] [PubMed] [Google Scholar]
  • 22. Itoh T, Zang YF, Murai S, Saito H. Effects of Panax ginseng root on the vertical and horizontal motor activities and on brain monoamine‐related substances in mice. Planta Med 1989;55:429–433. [DOI] [PubMed] [Google Scholar]
  • 23. Kim HS, Kang JG, Seong YH, Nam KY, Oh KW. Blockade by ginseng total saponins of the development of cocaine induced reverse tolerance and dopamine receptor supersensitivity in mice. Pharmacol Biochem Behav 1995;50:23–27. [DOI] [PubMed] [Google Scholar]
  • 24. Petkov V. Effect of ginseng on the brain biogenic monoamines and 3’,5’‐AMP system. Experiments on rats. Arzneimittelforschung 1978;28:388–393. [PubMed] [Google Scholar]
  • 25. Kimura T, Saunders PA, Kim HS, Rheu HM, Oh KW, Ho IK. Interactions of ginsenosides with ligand‐bindings of GABA(A) and GABA(B) receptors. Gen Pharmacol 1994;25:193–199. [DOI] [PubMed] [Google Scholar]
  • 26. Wang A, Cao Y, Wang Y, Zhao R, Liu C. Effects of Chinese ginseng root and stem‐leaf saponins on learning, memory and biogenic monoamines of brain in rats. Zhongguo Zhong Yao Za Zhi 1995;20:493–495. [PubMed] [Google Scholar]
  • 27. Sala F, Mulet J, Choi S, et al Effects of ginsenoside Rg2 on human neuronal nicotinic acetylcholine receptors. J Pharmacol Exp Ther 2002;301:1052–1059. [DOI] [PubMed] [Google Scholar]
  • 28. Choi SE, Choi S, Lee JH, Whiting PJ, Lee SM, Nah SY. Effects of ginsenosides on GABA(A) receptor channels expressed in Xenopus oocytes. Arch Pharm Res 2003;26:28–33. [DOI] [PubMed] [Google Scholar]
  • 29. Erickson CA, Barnes CA. The neurobiology of memory changes in normal aging. Exp Gerontol 2003;38:61–69. [DOI] [PubMed] [Google Scholar]
  • 30. Wang YZ, Chen J, Chu SF, Wang YS, Wang XY, Chen NH, Zhang JT. Improvement of memory in mice and increase of hippocampal excitability in rats by ginsenoside Rg1's metabolites ginsenoside Rh1 and protopanaxatriol. J Pharmacol Sci 2009;109:504–510. [DOI] [PubMed] [Google Scholar]
  • 31. Yang JH, Han SJ, Ryu JH, Jang IS, Kim DH. Ginsenoside Rh2 ameliorates scopolamine‐induced learning deficit in mice. Biol Pharm Bull 2009;32:1710–1715. [DOI] [PubMed] [Google Scholar]
  • 32. Wang XY, Zhang JT. Effects of ginsenoside Rg1 on synaptic plasticity of freely moving rats and its mechanism of action. Acta Pharmacol Sin 2001;22:657–662. [PubMed] [Google Scholar]
  • 33. Mook‐Jung I, Hong HS, Boo JH, et al Ginsenoside Rb1 and Rg1 improve spatial learning and increase hippocampal synaptophysin level in mice. J Neurosci Res 2001;63:509–515. [DOI] [PubMed] [Google Scholar]
  • 34. Zhao H, Li Q, Pei X, Zhang Z, Yang R, Wang J, Li Y. Long‐term ginsenoside administration prevents memory impairment in aged C57BL/6J mice by up‐regulating the synaptic plasticity‐related proteins in hippocampus. Behav Brain Res 2009;201:311–317. [DOI] [PubMed] [Google Scholar]
  • 35. Miller RJ. Rocking and rolling with Ca2+ channels. Trends Neurosci 2001;24:445–449. [DOI] [PubMed] [Google Scholar]
  • 36. Berridge MJ, Bootman MD, Lipp P. Calcium–a life and death signal. Nature 1998;395:645–648. [DOI] [PubMed] [Google Scholar]
  • 37. Kim S, Nah SY, Rhim H. Neuroprotective effects of ginseng saponins against L‐type Ca2+ channel‐mediated cell death in rat cortical neurons. Biochem Biophys Res Commun 2008;365:399–405. [DOI] [PubMed] [Google Scholar]
  • 38. Zhang YF, Fan XJ, Li X, Peng LL, Wang GH, Ke KF, Jiang ZL. Ginsenoside Rg1 protects neurons from hypoxic‐ischemic injury possibly by inhibiting Ca2+ influx through NMDA receptors and L‐type voltage‐dependent Ca2+ channels. Eur J Pharmacol 2008;586:90–99. [DOI] [PubMed] [Google Scholar]
  • 39. Kim J, Hoffman DA. Potassium channels: Newly found players in synaptic plasticity Neuroscientist 2008;14:276–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Choi S, Jung SY, Ko YS, Koh SR, Rhim H, Nah SY. Functional expression of a novel ginsenoside Rf binding protein from rat brain mRNA in Xenopus laevis oocytes. Mol Pharmacol 2002;61:928–935. [DOI] [PubMed] [Google Scholar]
  • 41. Cantrell AR, Catterall WA. Neuromodulation of Na+ channels: An unexpected form of cellular plasticity. Nat Rev Neurosci 2001;2:397–407. [DOI] [PubMed] [Google Scholar]
  • 42. Kim JH, Hong YH, Lee JH, et al A role for the carbohydrate portion of ginsenoside Rg3 in Na+ channel inhibition. Mol Cells 2005;19:137–142. [PubMed] [Google Scholar]
  • 43. Lee JH, Jeong SM, Kim JH, et al Characteristics of ginsenoside Rg3‐mediated brain Na+ current inhibition. Mol Pharmacol 2005;68:1114–1126. [DOI] [PubMed] [Google Scholar]
  • 44. Duan Y, Nicholson RA. 20(S)‐protopanaxadiol and the ginsenoside Rh2 inhibit Na+ channel‐activated depolarization and Na+ channel‐dependent amino acid neurotransmitter release in synaptic fractions isolated from mammalian brain. Comp Biochem Physiol C Toxicol Pharmacol 2008;147:351–356. [DOI] [PubMed] [Google Scholar]
  • 45. Liu D, Li B, Liu Y, Attele AS, Kyle JW, Yuan CS. Voltage‐dependent inhibition of brain Na(+) channels by American ginseng. Eur J Pharmacol 2001;413:47–54. [DOI] [PubMed] [Google Scholar]
  • 46. Sattler R, Tymianski M. Molecular mechanisms of calcium‐dependent excitotoxicity. J Mol Med 2000;78:3–13. [DOI] [PubMed] [Google Scholar]
  • 47. Kim YC, Kim SR, Markelonis GJ, Oh TH. Ginsenosides Rb1 and Rg3 protect cultured rat cortical cells from glutamate‐induced neurodegeneration. J Neurosci Res 1998;53:426–432. [DOI] [PubMed] [Google Scholar]
  • 48. Kim S, Ahn K, Oh TH, Nah SY, Rhim H. Inhibitory effect of ginsenosides on NMDA receptor‐mediated signals in rat hippocampal neurons. Biochem Biophys Res Commun 2002;296:247–254. [DOI] [PubMed] [Google Scholar]
  • 49. Kim S, Kim T, Ahn K, Park WK, Nah SY, Rhim H. Ginsenoside Rg3 antagonizes NMDA receptors through a glycine modulatory site in rat cultured hippocampal neurons. Biochem Biophys Res Commun 2004;323:416–424. [DOI] [PubMed] [Google Scholar]
  • 50. Lee E, Kim S, Chung KC, Choo MK, Kim DH, Nam G, Rhim H. 20(S)‐ginsenoside Rh2, a newly identified active ingredient of ginseng, inhibits NMDA receptors in cultured rat hippocampal neurons. Eur J Pharmacol 2006;536:69–77. [DOI] [PubMed] [Google Scholar]
  • 51. Tachikawa E, Kudo K, Kashimoto T, Takahashi E. Ginseng saponins reduce acetylcholine‐evoked Na+ influx and catecholamine secretion in bovine adrenal chromaffin cells. J Pharmacol Exp Ther 1995;273:629–636. [PubMed] [Google Scholar]
  • 52. Choi S, Jung SY, Lee JH, et al Effects of ginsenosides, active components of ginseng, on nicotinic acetylcholine receptors expressed in Xenopus oocytes. Eur J Pharmacol 2002;442:37–45. [DOI] [PubMed] [Google Scholar]
  • 53. Choi S, Lee JH, Oh S, Rhim H, Lee SM, Nah SY. Effects of ginsenoside Rg2 on the 5‐HT3A receptormediated ion current in Xenopus oocytes. Mol Cells 2003;15:108–113. [PubMed] [Google Scholar]
  • 54. Lee BH, Jeong SM, Lee JH, et al Differential effect of ginsenoside metabolites on the 5‐HT3A receptor‐mediated ion current in Xenopus oocytes. Mol Cells 2004;17:51–56. [PubMed] [Google Scholar]
  • 55. Lee BH, Lee JH, Yoon IS, et al Mutations of arginine 222 in pre‐transmembrane domain I of mouse 5‐HT(3A) receptor abolish 20(R)‐ but not 20(S)‐ginsenoside Rg(3) inhibition of 5‐HT‐mediated ion currents. Biol Pharm Bull 2007;30:1721–1726. [DOI] [PubMed] [Google Scholar]
  • 56. Schapira AH. Molecular and clinical pathways to neuroprotection of dopaminergic drugs in Parkinson disease. Neurology 2009;72:44–50. [DOI] [PubMed] [Google Scholar]
  • 57. Xu BB, Liu CQ, Gao X, Zhang WQ, Wang SW, Cao YL. Possible mechanisms of the protection of ginsenoside Re against MPTP‐induced apoptosis in substantia nigra neurons of Parkinson's disease mouse model. J Asian Nat Prod Res 2005;7:215–224. [DOI] [PubMed] [Google Scholar]
  • 58. Chen XC, Zhou YC, Chen Y, Zhu YG, Fang F, Chen LM. Ginsenoside Rg1 reduces MPTP‐induced substantia nigra neuron loss by suppressing oxidative stress. Acta Pharmacol Sin 2005;26:56–62. [DOI] [PubMed] [Google Scholar]
  • 59. Wang Q, Zheng H, Zhang ZF, Zhang YX. Ginsenoside Rg1 modulates COX‐2 expression in the substantia nigra of mice with MPTP‐induced Parkinson disease through the P38 signaling pathway. Nan Fang Yi Ke Da Xue Xue Bao 2008;28:1594–1598. [PubMed] [Google Scholar]
  • 60. Wang J, Xu HM, Yang HD, Du XX, Jiang H, Xie JX. Rg1 reduces nigral iron levels of MPTP‐treated C57BL6 mice by regulating certain iron transport proteins. Neurochem Int 2009b;54:43–48. [DOI] [PubMed] [Google Scholar]
  • 61. Rouault TA. Iron on the brain. Nat Genet 2001;28:299–300. [DOI] [PubMed] [Google Scholar]
  • 62. Ke Y, Qian ZM. Iron misregulation in the brain: A primary cause of neurodegenerative disorders. Lancet Neurol 2003;2:246–253. [DOI] [PubMed] [Google Scholar]
  • 63. Xu L, Chen WF, Wong MS. Ginsenoside Rg1 protects dopaminergic neurons in a rat model of Parkinson's disease through the IGF‐I receptor signalling pathway. Br J Pharmacol 2009;158:738–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Leung KW, Yung KK, Mak NK, Chan YS, Fan TP, Wong RN. Neuroprotective effects of ginsenoside‐Rg1 in primary nigral neurons against rotenone toxicity. Neuropharmacology 2007;52:827–835. [DOI] [PubMed] [Google Scholar]
  • 65. Ye R, Han J, Kong X, Zhao L, Cao R, Rao Z, Zhao G. Protective effects of ginsenoside Rd on PC12 cells against hydrogen peroxide. Biol Pharm Bull 2008;31:1923–1927. [DOI] [PubMed] [Google Scholar]
  • 66. Lin WM, Zhang YM, Moldzio R, Rausch WD. Ginsenoside Rd attenuates neuroinflammation of dopaminergic cells in culture. J Neural Transm Suppl 2007;72:105–112. [DOI] [PubMed] [Google Scholar]
  • 67. Yang L, Hao J, Zhang J, et al Ginsenoside Rg3 promotes beta‐amyloid peptide degradation by enhancing gene expression of neprilysin. J Pharm Pharmacol 2009;61:375–380. [DOI] [PubMed] [Google Scholar]
  • 68. Joo SS, Lee DI. Potential effects of microglial activation induced by ginsenoside Rg3 in rat primary culture: Enhancement of type A Macrophage Scavenger Receptor expression. Arch Pharm Res 2005;28:1164–1169. [DOI] [PubMed] [Google Scholar]
  • 69. Selkoe DJ. Alzheimer's disease: Genes, proteins, and therapy. Physiol Rev 2001;81:741–766. [DOI] [PubMed] [Google Scholar]
  • 70. Xie YH, Chen XC, Zhang J, et al Ginsenoside Rb1 attenuates beta‐amyloid peptide(25–35)‐induced hyperphosphorylation of tau protein through CDK5 signal pathway. Yao Xue Xue Bao 2007;42:828–832. [PubMed] [Google Scholar]
  • 71. Chen X, Huang T, Zhang J, Song J, Chen L, Zhu Y. Involvement of calpain and p25 of CDK5 pathway in ginsenoside Rb1's attenuation of beta‐amyloid peptide25–35‐induced tau hyperphosphorylation in cortical neurons. Brain Res 2008;1200:99–106. [DOI] [PubMed] [Google Scholar]
  • 72. Shieh PC, Tsao CW, Li JS, Wu HT, Wen YJ, Kou DH, Cheng JT. Role of pituitary adenylate cyclase‐activating polypeptide (PACAP) in the action of ginsenoside Rh2 against beta‐amyloid‐induced inhibition of rat brain astrocytes. Neurosci Lett 2008;434:1–5. [DOI] [PubMed] [Google Scholar]
  • 73. Chen XC, Chen LM, Zhu YG, Fang F, Zhou YC, Zhao CH. Involvement of CDK4, pRB, and E2F1 in ginsenoside Rg1 protecting rat cortical neurons from beta‐amyloid‐induced apoptosis. Acta Pharmacol Sin 2003;24:1259–1264. [PubMed] [Google Scholar]
  • 74. Wei CB, Jia JP, Liang P, Guan YQ. Ginsenoside‐Rg1 inhibits cell apoptosis induced by beta amyloid. Zhonghua Yi Xue Za Zhi 2008;88:1763–1766. [PubMed] [Google Scholar]
  • 75. Wang XY, Chen J, Zhang JT. Effect of ginsenoside Rg1 on learning and memory impairment induced by beta‐amzloid peptide(25–35) and its mechanism of action. Yao Xue Xue Bao 2001;36:1–4. [PubMed] [Google Scholar]
  • 76. Kanekura K, Suzuki H, Aiso S, Matsuoka M. ER stress and unfolded protein response in amyotrophic lateral sclerosis. Mol Neurobiol 2009;39:81–89. [DOI] [PubMed] [Google Scholar]
  • 77. Jiang F, DeSilva S, Turnbull J. Beneficial effect of ginseng root in SOD‐1 (G93A) transgenic mice. J Neurol Sci 2000;180:52–54. [DOI] [PubMed] [Google Scholar]
  • 78. Kim YH, Park KH, Rho HM. Transcriptional activation of the Cu,Zn‐superoxide dismutase gene through the AP2 site by ginsenoside Rb2 extracted from a medicinal plant, Panax ginseng. J Biol Chem 1996;40:24539–24543. [DOI] [PubMed] [Google Scholar]
  • 79. Damiano M, Galvan L, Déglon N, Brouillet E. Mitochondria in Huntington's disease. Biochim Biophys Acta 2010;1802:52–61. [DOI] [PubMed] [Google Scholar]
  • 80. Wu J, Jeong HK, Bulin SE, Kwon SW, Park JH, Bezprozvanny I. Ginsenosides protect striatal neurons in a cellular model of Huntington's disease. J Neurosci Res 2009;87:1904–1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Zhang H, Li Q, Graham RK, Slow E, Hayden MR, Bezprozvanny I. Full length mutant huntingtin is required for altered Ca2+ signaling and apoptosis of striatal neurons in the YAC mouse model of Huntington's disease. Neurobiol Dis 2008;31:80–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Young AB, Greenamyre JT, Hollingsworth Z, Albin R, D’Amato C, Shoulson I, Penney JB. NMDA receptor losses in putamen from patients with Huntington's disease. Science 1988;241:981–983. [DOI] [PubMed] [Google Scholar]
  • 83. Albin RL, Young AB, Penney JB, et al Abnormalities of striatal projection neurons and N‐methyl‐D‐aspartate receptors in presymptomatic Huntington's disease. N Engl J Med 1990;322:1293–1298. [DOI] [PubMed] [Google Scholar]
  • 84. Arzberger T, Krampfl K, Leimgruber S, Weindl A. Changes of NMDA receptor subunit (NR1, NR2B) and glutamate transporter (GLT1) mRNA expression in Huntington's disease — an in situ hybridization study. J Neuropathol Exp Neurol 1997;56:440–454. [DOI] [PubMed] [Google Scholar]
  • 85. Chen YH, Du GH, Zhang JT. Salvianolic acid B protects brain against injuries caused by ischemia‐reperfusion in rats. Acta Pharmacol Sin 2000;21:463–466. [PubMed] [Google Scholar]
  • 86. Lim JH, Wen TC, Matsuda S, et al Protection of ischemic hippocampal neurons by ginsenoside Rb1, a main ingredient of ginseng root. Neurosci Res 1997;28:191–200. [DOI] [PubMed] [Google Scholar]
  • 87. Yuan QL, Yang CX, Xu P, et al Neuroprotective effects of ginsenoside Rb1 on transient cerebral ischemia in rats. Brain Res 2007;1167:1–12. [DOI] [PubMed] [Google Scholar]
  • 88. Chu GX, Chen X. Anti‐lipid peroxidation and protection of ginsenosides against cerebral ischemia‐reperfusion injuries in rats. Zhongguo Yao Li Xue Bao 1990;11:119–123. [PubMed] [Google Scholar]
  • 89. Tian J, Fu F, Geng M, et al Neuroprotective effect of 20(S)‐ginsenoside Rg3 on cerebral ischemia in rats. Neurosci Lett 2005;37:92–97. [DOI] [PubMed] [Google Scholar]
  • 90. Zhou XM, Cao YL, Dou DQ. Protective effect of ginsenoside‐Re against cerebral ischemia/reperfusion damage in rats. Biol Pharm Bull 2006;29:2502–2505. [DOI] [PubMed] [Google Scholar]
  • 91. Chen LM, Zhou XM, Cao YL, Hu WX. Neuroprotection of ginsenoside Re in cerebral ischemia‐reperfusion injury in rats. J Asian Nat Prod Res 2008;10:439–445. [DOI] [PubMed] [Google Scholar]
  • 92. Ye R, Li N, Han J, Kong X, Cao R, Rao Z, Zhao G. Neuroprotective effects of ginsenoside Rd against oxygen‐glucose deprivation in cultured hippocampal neurons. Neurosci Res 2009;64:306–310. [DOI] [PubMed] [Google Scholar]
  • 93. Kim, SU , de Vellis J. 2005. Microglia in health and disease. J Neurosci Res 2005;81:302–313. [DOI] [PubMed] [Google Scholar]
  • 94. Wyss‐Coray T, Mucke L. Inflammation in neurodegenerative disease: A double edged sword. Neuron 2002;35:419–432. [DOI] [PubMed] [Google Scholar]
  • 95. Bae EA, Kim EJ, Park JS, Kim HS, Ryu JH, Kim DH. Ginsenoside Rg3 and Rh2 inhibit the activation of AP‐1 and protein kinase A pathway in lipopolysaccharide/interferon‐γ‐stimulated BV‐2 microglial cells. Planta Med 2006;72:627–633. [DOI] [PubMed] [Google Scholar]
  • 96. Wu CF, Bi XL, Yang JY, et al Differential effects of ginsenosides on NO and TNF‐α production by LPSactivated N9 microglia. Int Immunopharmacol 2007;7:313–320. [DOI] [PubMed] [Google Scholar]
  • 97. Joo SS, Yoo YM, Ahn BW, Nam SY, Kim YB, Hwang KW, Lee do I. Prevention of inflammation‐mediated neurotoxicity by Rg3 and its role in microglial activation. Biol Pharm Bull 2008;31:1392–1396. [DOI] [PubMed] [Google Scholar]
  • 98. Park JS, Park EM, Kim DH, et al Anti‐inflammatory mechanism of ginseng saponins in activated microglia. J Neuroimmunol 2009;209:40–49. [DOI] [PubMed] [Google Scholar]
  • 99. He W, Zhu Z. Effect of Panax notoginseng saponins on intercellular adhesion molecule‐1 expression and neutrophil infiltration in cerebral infarction. Zhong Yao Cai 2005;28:403–405. [PubMed] [Google Scholar]
  • 100. Jung SH, Woo MS, Kim SY, et al Ginseng saponin metabolite suppresses phorbol ester‐induced matrix metalloproteinase‐9 expression through inhibition of activator protein‐1 and mitogen‐activated protein kinase signaling pathways in human astroglioma cells. Int J Cancer 2006;118:490–497. [DOI] [PubMed] [Google Scholar]
  • 101. Kim SY, Kim DH, Han SJ, Hyun JW, Kim HS. Repression of matrix metalloproteinase gene expression by ginsenoside Rh2 in human astroglioma cells. Biochem Pharmacol 2007;74:1642–1651. [DOI] [PubMed] [Google Scholar]
  • 102. Oh M, Choi YH, Choi S, et al Antiproliferating effects of ginsenoside Rh2 on MCF‐7 human breast cancer cells. Int J Oncol 1999;14:869–875. [DOI] [PubMed] [Google Scholar]
  • 103. Oh JI, Chun KH, Joo SH, Oh YT, Lee SK. Caspase‐3‐ dependent protein kinase C delta activity is required for the progression of Ginsenoside‐Rh2‐induced apoptosis in SKHEP‐1 cells. Cancer Lett 2005;230:228–238. [DOI] [PubMed] [Google Scholar]
  • 104. Kim HE, Oh JH, Lee SK, Oh YJ. Ginsenoside RH‐2 induces apoptotic cell death in rat C6 glioma via a reactive oxygenand caspase‐dependent but Bcl‐X(L)‐independent pathway. Life Sci 1999;65:33–40. [DOI] [PubMed] [Google Scholar]
  • 105. Fei XF, Wang BX, Tashiro S, Li TJ, Ma JS, Ikejima T. Apoptotic effects of ginsenoside Rh2 on human malignant melanoma A375‐S2 cells. Acta Pharmacol Sin 2002;23:315–322. [PubMed] [Google Scholar]
  • 106. Kim YS, Jin SH. Ginsenoside Rh2 induces apoptosis via activation of caspase‐1 and ‐3 and up‐regulation of Bax in human neuroblastoma. Arch Pharm Res 2004;27:834–839. [DOI] [PubMed] [Google Scholar]
  • 107. Surh YJ, Lee JY, Choi KJ, Ko SR. Effects of selected ginsenosides on phorbol ester‐induced expression of cyclooxygenase‐2 and activation of NF‐kappaB and ERK1/2 in mouse skin. Ann NY Acad Sci 2002;973:396–401. [DOI] [PubMed] [Google Scholar]
  • 108. Park SA, Kim EH, Na HK, Surh YJ. KG‐135 inhibits COX‐2 expression by blocking the activation of JNK and AP‐1 in phorbol ester‐stimulated human breast epithelial cells. Ann NY Acad Sci 2007;1095:545–553. [DOI] [PubMed] [Google Scholar]

Articles from CNS Neuroscience & Therapeutics are provided here courtesy of Wiley

RESOURCES