Skip to main content
Acta Pharmacologica Sinica logoLink to Acta Pharmacologica Sinica
. 2013 Oct 14;34(11):1367–1373. doi: 10.1038/aps.2013.100

Yin and Yang of ginseng pharmacology: ginsenosides vs gintonin

Dong-soon Im 1,*, Seung-yeol Nah 2,*
PMCID: PMC3818586  PMID: 24122014

Abstract

Ginseng, the root of Panax ginseng, has been used in traditional Chinese medicine as a tonic herb that provides many beneficial effects. Pharmacologic studies in the last decades have shown that ginsenosides (ginseng saponins) are primarily responsible for the actions of ginseng. However, the effects of ginseng are not fully explained by ginsenosides. Recently, another class of active ingredients called gintonin was identified. Gintonin is a complex of glycosylated ginseng proteins containing lysophosphatidic acids (LPAs) that are the intracellular lipid mitogenic mediator. Gintonin specifically and potently activates the G protein-coupled receptors (GPCRs) for LPA. Thus, the actions of ginseng are now also linked to LPA and its GPCRs. This linkage opens new dimensions for ginseng pharmacology and LPA therapeutics. In the present review, we evaluate the pharmacology of ginseng with the traditional viewpoint of Yin and Yang components. Furthermore, we will compare ginsenoside and gintonin based on the modern view of molecular pharmacology in terms of ion channels and GPCRs.

Keywords: ginseng, ginsenoside, gintonin, lysophosphatidic acid, ion channels, G protein-coupled receptors, Ca2+, traditional Chinese medicine

Introduction

Since ginsenosides (ginseng saponins) were first identified as active ingredients in ginseng, many studies have shown that ginsenosides negatively regulate ion channels1. For example, ginsenoside Rg3 inhibits not only voltage-gated Ca2+ and Na+ channels but also ligand-gated ion channels such as 5-HT3 receptors2,3,4. Furthermore, ginsenosides stimulate anion-gated GABAA and glycine receptors5,6. Therefore, ginsenosides decrease the excitability of excitable cells by inhibition of cation influx and stimulation of anion influx across the plasma membrane.

Crude ginseng saponin fractions elevate the intracellular Ca2+ levels in mammalian cells and activate Ca2+-activated Cl channels in Xenopus oocytes7. However, this effect was not explained by ginsenosides. Recently, the active ingredients responsible for the Ca2+ rise were elucidated as novel glycolipoproteins and named gintonins8. Gintonin is a complex of glycosylated ginseng proteins containing lysophosphatidic acids (LPAs). LPA is a representative mitogenic lipid mediator9. Gintonin activates G protein-coupled receptors (GPCRs) for LPA8. Unexpectedly, the action of ginseng is now linked to LPA and its GPCRs and vice versa. This linkage opens new dimensions for ginseng pharmacology and LPA therapeutics.

Ginsenosides tend to attenuate cell excitations by blocking cation Ca2+ and Na+ influxes or by enhancing anion Cl influx. However, gintonin induces transient Ca2+ elevation and activation of MAPK, PI3K, PKC, and Rho kinase via LPA receptors to evoke stimulatory cellular responses4,8,10,11. Thus, ginsenoside acts as a negative regulator (Yin), and gintonin acts as a positive regulator (Yang) for ginseng pharmacology. The pharmacological actions of ginseng are now explainable and complementary with the opposing actions of gintonin and ginsenoside. Therapeutic application of LPA might be expanded by traditional usages of ginseng and vice versa. In this article, both traditional and modern ginseng pharmacology will be discussed from the viewpoint of Yin and Yang.

Ginseng and ginsenosides

Ginseng, the root of Panax ginseng CA Meyer, has been used for thousands of years in Asian countries such as Korea, China, and Japan. Panax means 'all heal' in Greek, and the Chinese characters of ginseng originated from the human-like shape of the ginseng root12. The traditional beneficial effects of ginseng are replenishment of vital energy, mood elevation, and longevity. Therefore, in ancient times, ginseng was considered as a panacea that provided eternal youth1,12. In addition, modern pharmacological studies have revealed ginseng's adaptogenic activities against stress, fatigue, cardiovascular dysfunction, and various diseases, including cancer and neurodegenerative disorders. Its active components also have been intensively studied over the past decades.

The representative active ingredients of ginseng are ginsenosides (ginseng saponin), which are derivatives of the triterpenoid dammarane1. More than 100 different types of ginsenosides have been isolated and identified from the roots of Korean and American ginseng13. Each ginsenoside is composed of three parts, including a hydrophobic four-ring backbone structure, an attached carbohydrate portion, and an aliphatic side chain (Figure 1).

Figure 1.

Figure 1

Virtual docking model of ginsenoside Rg3 to chick α7 nicotinic acetylcholine receptor (nAChR mutant L247T) and hydrogen bonds. Virtual dockings of ginsenoside Rg3 to chick α7 nicotinic acetylcholine receptor (AChR, L247T mutant) channel homology models. (A) Top view of the highest-ranked docking model of ginsenoside Rg3 to chick α7 nicotinic acetylcholine receptor (nAChR mutant, L247T) channel. The channel is shown as a cartoon diagram, and ginsenoside Rg3 is represented by a ball and chain model. Subunits are shown in different colors. (B) Side view of the docking model of ginsenoside Rg3 to nAChR receptor. One of the subunits is omitted in side view for clarity. (C) Poseview analysis of protein-ligand interactions. Hydrogen bonds are denoted by dotted lines. Spline sections indicate hydrophobic contacts, highlight the hydrophobic regions of ginsenoside Rg3, and provide the identities of contacting amino acids. The roman numerals in parenthesis indicate subunits of the pentamer. Adapted from Lee et al18.

Ginsenosides and the modulation of ion channels

Because ginsenosides were first identified as the active ingredients in ginseng, many studies have demonstrated negative regulation of ion channels by ginsenosides1. Their actions are stereoselective, but they lack the specificity and selectivity of other channel inhibitors. The EC50 values of ginsenosides are in the μmol/L range, and many ion channels are affected. For example, the EC50 values for the ginsenoside Rg3 are approximately 0.41–97.3 μmol/L4,14,15,16,17,18,19,20. The ginsenoside Rg3 not only inhibits voltage-gated Ca2+, K+, and Na+ channels, ligand-gated 5-HT3, α7 nicotinic acetylcholine, and NMDA receptors but also activates K+ channels such as KCNQ K+, BKCa, and hERG K+4,14,15,16,17,18,19,20,21. Other ginsenosides enhance the activation of anion GABAA and glycine receptors22,23 (Table 1). Therefore, ginsenosides decrease the cellular excitability of excitable cells by inhibiting cation influx and by stimulating anion influx across plasma membranes.

Table 1. Summary of the EC50 and IC50 values of ginsenoside-induced inhibitions or stimulations of the activities of various voltage-gated ion or ligand-gated ion channels.

Voltage-gated ion channels Ginsenoside EC50 or IC50 (μmol/L) Interacting amino acids
Ca2+
L Rg3, Rb1 39.9±9.51,2 L417, N428, L431
N Rg3 64.4±13.61  
P/Q Rg3 29.6±11.31  
R Rg3 57.5±12.51  
T Rg3 97.3±12.41  
K+
Kv1.4 Rg3 32.6±2.21 K531
BKCa Rg3 15.3±3.13 Y360
hERG Rg3 0.41±0.053 S631
KCNQ Rg3 15.2±8.74 K318
Na+
Nav1.2 Rg3 32.0±6.05 I417, N418, L421
Nav1.4 Rg3 58.5±6.36 I433, N434, L437
Nav1.5 Rg3 16.1±2.87  
Ligand-gated ion channels Ginsenoside EC50 or IC50 (μmol/L)  
GABAA Rc 53.0±12.38  
Glycine Rf 49.8±9.89  
5-HT3 Rg3 27.6±4.310 V291, F292, I295
Nicotinic acetylcholine
α3β4 Rg2 60±1411  
α1β1δε Rg2 16±911  
α7 (L247A mutant) Rg3 33.1±1.312 L247
NMDA Protopanaxatriol 48±1613  

EC50 values are shown for BKCa, hERG, and KCNQ K+ channels and GABAA and glycine receptors and IC50 values are shown for the remainder and were determined in oocytes expressing these ion channels or receptors.

Site-directed mutagenesis experiments have identified ginsenoside interaction site(s), and homology docking modeling has provided three-dimensional configurations for ginsenosides and channel proteins15,16,18,24 (Figure 1). For example, the hydroxyl groups on the second carbohydrate in ginsenoside Rg3 form stable hydrogen bonds with the core amino acids of channel proteins (Figure 1). The 1st or 2nd amino acids after the K+ channel 'signature sequence' (TXGYGD) at the pore entrances have been shown to interact with ginsenoside Rg3. That is, K318 in the KCNQ K+ channel, S631 in the hERG K+ channel, Y360 in the BKCa K+ channel, K531 in the K+V1.4 channel, and K859 in the neuronal Na+ channel. Similarly, the amino acids I417, N418, and L421 in the Na+V1.2 channel, I433, N434, and L437 in the Na+V1.4 channel, L417, N428, and L431 in the L-type Ca2+ channel, V291, F292, and I295 in the 5-HT3 channel, and L247 in the α7 nicotinic acetylcholine receptor were found to interact with ginsenosides4,14,15,16,17,18,20 (Table 1 and Figure 1). Therefore, ginsenosides have regulatory effects in a broad range of ion channels with low affinities, but their common factor is that they stabilize membrane potentials and attenuate cellular activities.

Ginsenosides in ginseng pharmacology

Ginsenosides decrease the excitability of neuronal cells by inhibiting cation influx and/or by stimulating anion influx. These pharmacologic actions of ginsenosides are linked to reductions in the excitabilities of neurons, smooth muscle cells, and cardiomyocytes. Furthermore, the ginsenoside-induced modulation of Ca2+ and K+ channels results in the dilation of blood vessels via relaxation of smooth muscles25,26,27,28,29. In bradycardia, it induces relaxation of the cardiomyocytes, which explains its anti-hypertensive and cardioprotective effects30,31,32,33,34,35. In addition, ginsenoside-induced inhibition of the cation-gated NMDA receptor and neuronal Na+ channels and its stimulation of anion-gated GABAA receptors and glycine receptors explain the neuroprotective and anxiolytic effects of ginseng10,36. It is also possible that ginseng attenuation of cisplatin-induced nausea and vomiting is due its inhibition of 5-HT3 ion channels37.

Discovery of gintonin

The crude ginseng total saponin (cGTS) fraction contains approximately 50% ginsenosides by weight. Furthermore, the cGTS fraction increases intracellular Ca2+ in mammalian cells and activates endogenous Ca2+-activated Cl channels in Xenopus oocytes, whereas ginsenosides do not. These data imply that some unidentified ginseng component(s) is responsible for the Ca2+ increase7. In addition, the cGTS fraction-induced Ca2+ increases are both reversible and transient. Precise and continuing studies over the past twenty years have elucidated a signaling pathway downstream of cGTS in oocytes, namely the unknown membrane target protein-Gq/11-PLCβ3-IP3-Ca2+ release38. Later, homologous desensitization by cGTS was shown to be mediated through GRK2 and β-arrestin I in oocytes, strongly implying GPCR involvement (Figure 2)39.

Figure 2.

Figure 2

Schematic diagram of gintonin-mediated signal transduction pathways. For the desensitization, gintonin activates LPA GPCRs, which leads to activation of GRK2. The activated GRK2 phosphorylates the LPA GPCRs and then β-arrestin I is recruited. The recruited β-arrestin I inhibits GPCR-G protein coupling. For cellular responses, gintonin activates LPA GPCRs, which leads to activation of phospholipase C (PLC). The activated PLC produces IP3 and diacylglycerol (DAG). DAG activates protein kinase C (PKC), which phosphorylates Ca2+ channels. IP3 mobilizes Ca2+ from internal Ca2+ stores through IP3 receptors. The increased Ca2+ levels activate many kinases.

In 2011, the active ingredient in cGTS was finally separated by anion exchange chromatography after ginseng butanol extraction40. The novel non-saponin ingredient was designated as gintonin, where gin was derived from ginseng, ton from the tonic effects of ginseng, and in from protein41. Six different gintonin have been identified, and all six induce intracellular Ca2+ increases in mammalian cells and Xenopus oocytes, confirming that gintonin is responsible for cGTS-induced Ca2+ mobilization. Gintonin is composed of carbohydrates, lipids, and proteins, such as, ginseng major latex-like protein and ginseng ribonuclease-like storage proteins. Thus, gintonin is part of a novel class of glycolipoproteins in ginseng that induces intracellular Ca2+ increases in mammalian cells8. Ginseng contains 0.2% gintonin by weight (Table 2).

Table 2. A brief comparison of ginseng components, gintonin, and ginsenosides.

  Gintonin Ginsenosides
Molecular weight (MW) Native MW: 67 kDa Apparent MW: 13 kDa 0.6–1.3 kDa
Composition Glycolipoprotein: carbohydrates (Glucose), lipids (LPA C18:2), and ginseng proteins (GLP and GSP) Dammarane glycosides
Content in ginseng 0.2% 3%–4% (Sum of individual ginsenosides)
Target protein on cell membrane and signal cascades LPA receptors, transient [Ca2+]i elevation via PTX-sensitive and -insensitive G proteins coupled PLC pathway Non-selective interactions with ion channels and receptors, do not have signal transduction pathway
Desensitization after repeated treatment on cells Induction of rapid desensitization No desensitization

Gintonin activation of GPCRs for LPA

During additional studies with the gintonin, it was shown that phospholipase A1 had attenuating effects on gintonin. This finding highlighted the importance of position 1 esterification on the fatty acid component, which suggests that gintonin contains phospholipids8. Previously, Tigyi and Miledi demonstrated that LPA bound to serum albumin could sufficiently activate Ca2+-activated Cl channels in Xenopus oocytes, and digestion with PLA1 prevented this activation42. Furthermore, using methanol extraction, they demonstrated LPA dissociation from serum albumin42. Building upon this reference, researchers examined whether gintonin contained LPAs by liquid chromatography-electrospray ionization/multi-stage mass spectrometry (LC-ESI-MS/MS) analysis43. LC-ESI-MS/MS analysis showed that 9.5% of gintonin is composed of lysophosphatidic acids (LPAs), such as LPA C18:2, LPA C16:0, and LPA C18:1, indicating that the bioactive component(s) of gintonin (with respect to the Ca2+ response) is a LPA43.

LPA is an intercellular lipid mediator that functions as a mitogen with hormone- and growth-factor-like activities in most cell types9,44. Currently, three EDG family GPCRs (LPA1–3) and three purinergic GPCRs (LPA4–6) have been reported to act as LPA receptors9. Hwang et al demonstrated that gintonin activates LPA receptors in GPCR-expressing B103 mammalian cells and in Xenopus oocytes45. Gintonin only activates LPA receptors specifically, not other GPCRs, such as S1P, fatty acids, 5-HT1C, and muscarinic acetylcholine receptor subtypes (m1, m3, and m5)8. Furthermore, the LPA GPCRs have different affinities for gintonin, which follow a decreasing order of LPA2>LPA5>LPA1>LPA3>LPA48. Furthermore, gintonin has 3- to 130-fold greater affinity for LPA GPCRs than free LPA. This might be due to ginseng proteins in gintonin because the protein components of gintonin may function as efficient LPA carriers to effectively carry LPA to LPA receptors and protect LPA from hydrolyzing enzymes8. Based on the average molecular weight of gintonin (20 kDa), four LPA molecules could most likely bind to one molecule of ginseng protein. The LPA content in ginseng is 80 to 240-fold higher than in other plants, which in part explains its unique pharmacologic properties. Somewhat unexpectedly, the action of ginseng is now linked to LPA and its GPCRs and vice versa, and this linkage opens new dimensions for ginseng pharmacology and LPA therapeutics (Table 2).

Gintonin in ginseng pharmacology

Gintonin induces transient Ca2+ elevation and activates MAPK, PI3K, PKC, and Rho kinase via LPA receptors to evoke stimulatory cellular responses4,11 (Figure 2). Gintonin evokes cell proliferation and migration and morphological changes in human umbilical vein endothelial cells and PC12 neuronal cells4,5,41,45. These effects of gintonin are consistent with those caused by LPAs via GPCRs and diverse G proteins, such as Gαi/o, Gα12/13, and Gαq/111. Gintonin hinders the amyloidogenic pathway and induces non-amyloidogenic pathways that produce beneficial soluble APPα (sAPPα) in neurons by activating LPA receptors41. Gintonin also reduces the release of Aβ1–42 and attenuates Aβ1–40-induced cytotoxicity41. In addition, gintonin has been shown to rescue Aβ1–40-induced cognitive dysfunction in mice41. Furthermore, in a transgenic murine Alzheimer's disease model, long-term oral administration of gintonin effectively attenuated both amyloid plaque deposition and short- and long-term memory impairment41. Thus, gintonin may contribute to the memory-improving effects of ginseng, which have been proven in human trials.

Autotaxin was found as an autocrine factor released by tumors, which stimulates tumor growth and migration44. Later, autotaxin was identified as lysophospholipase D, which produces LPA from lysophosphatidylcholine46,47. Autotaxin and LPA function as mitogenic and motility signals in various cancers, including neuroblastoma, hepatoma, lung cancer, ovarian cancer, metastatic breast cancer, and melanoma44. A recent report showed that LPA C18:2, which is highly abundant in gintonin, inhibits autotaxin activity. Furthermore, Hwang et al showed that gintonin inhibits autotaxin activity in vitro and metastasis of B16/F10 melanoma cells in vivo45. Vessel formation in tumors was also reduced in gintonin-treated mice. These effects may contribute to the anti-cancer effects of ginseng45.

Yin and Yang of ginseng pharmacology

In Chinese medicine, Yin and Yang are opposite and complementary forces that combine to form a whole. Ginseng pharmacology is interesting because the two active components have opposite effects and compliment the entire effect of ginseng. For example, ginsenosides stabilize membrane potentials via the dual modulation of ion channels, but gintonin and LPA activate many cellular responses via GPCR activation. Ginsenosides inhibit Ca2+ influx, but gintonin induces a transient Ca2+ rise, and ginsenosides have low affinity for calcium but are relatively abundant in ginseng, whereas gintonin has a higher affinity for calcium but is only present at low levels (Table 2). Thus, it is highly likely that ginsenosides act as a Yin component to gintonin's Yang component to produce the effects of ginseng. Therefore, it appears that the healing effects of Panax are the result of a harmonious balance between the positive and negative actions of ginsenosides and gintonins, reminiscent of the Yin and Yang forces in Chinese medicine.

Concluding remark and perspective

Our understanding of ginseng pharmacology has advanced tremendously during the last few decades. Ginsenosides were considered to be the active ingredients of ginseng for 5 decades. However, many researchers used ginseng extract (the butanol fraction) for pharmacologic studies because individual ginsenosides were scarce and difficult to purify. As a result, other components were included in these pharmacology experiments. Gintonin is now considered as a part of the ginseng fraction, and the LPAs in this fraction are considered responsible for a variety of the biological effects of ginseng, which are mediated through GPCRs. Furthermore, it appears that the pharmacological actions of ginseng can now be explained by the complementary opposing actions of gintonin and ginsenoside. One example might be the anticancer effects of the ginseng extract. Many ginsenosides have shown clear anticancer activity in different cancer cell lines by regulation of cell proliferation13. Gintonin exhibited autotoxin inhibition in vitro and inhibition of metastasis of B16/F10 melanoma cells in vivo45. Therefore, both components in ginseng may act synergistically and complementarily for the anti-tumor efficacy of ginseng.

How ginsenosides and gintonin work together in the body or whether ginsenosides affect the actions of gintonin or vice versa remains to be established. Future studies are required to elucidate the pharmacological effects of the different actions of ginsenosides and gintonin with respect to their individual contributions and the effects of whole ginseng in biological systems. Future research will undoubtedly expand our knowledge of ginseng pharmacology and the applications of LPA. In addition, the therapeutic application of LPA might be facilitated by knowledge of the traditional usages of ginseng. Although new drug developments based on targeting LPA receptors are in the pipeline, gintonin might provide alternative therapies for pathologic conditions related to LPA and LPA receptor-related diseases.

Acknowledgments

This work was supported by the Basic Science Research Program (2011–0021144 and 2011–0021158) and the Priority Research Center Program through the National Research Foundation of Korea (NRF), which is funded by the Ministry of Education, Science, and Technology (2012–0006686), by a grant from the BK21 project funded to Seung-yeol NAH and by the MRC program (Grant No 2009–0083538) of the Korean National Research Foundation funded by the Korean government (MEST).

References

  1. Nah SY, Kim DH, Rhim H. Ginsenosides: are any of them candidates for drugs acting on the central nervous system. CNS Drug Rev. 2007;13:381–404. doi: 10.1111/j.1527-3458.2007.00023.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Lee JH, Choi SH, Lee BH, Yoon IS, Shin TJ, Pyo MK, et al. Modifications of aliphatic side chain of 20(S)-ginsenoside RG3 cause an enhancement or loss of brain Na+ channel current inhibitions. Biol Pharm Bull. 2008;31:480–6. doi: 10.1248/bpb.31.480. [DOI] [PubMed] [Google Scholar]
  3. Choi SH, Lee JH, Pyo MK, Lee BH, Shin TJ, Hwang SH, et al. Mutations Leu427, Asn428, and Leu431 residues within transmembrane domain-I-segment 6 attenuate ginsenoside-mediated L-type Ca2+ channel current inhibitions. Biol Pharm Bull. 2009;32:1224–30. doi: 10.1248/bpb.32.1224. [DOI] [PubMed] [Google Scholar]
  4. Lee BH, Lee JH, Lee SM, Jeong SM, Yoon IS, Choi SH, et al. Identification of ginsenoside interaction sites in 5-HT3A receptors. Neuropharmacology. 2007;52:1139–50. doi: 10.1016/j.neuropharm.2006.12.001. [DOI] [PubMed] [Google Scholar]
  5. Jang S, Ryu JH, Kim DH, Oh S. Changes of [3H]MK-801, [3H]muscimol and 3H]flunitrazepam binding in rat brain by the prolonged ventricular infusion of transformed ginsenosides. Neurochem Res. 2004;29:2257–66. doi: 10.1007/s11064-004-7034-2. [DOI] [PubMed] [Google Scholar]
  6. 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;323:416–24. doi: 10.1016/j.bbrc.2004.08.106. [DOI] [PubMed] [Google Scholar]
  7. Choi S, Rho SH, Jung SY, Kim SC, Park CS, Nah SY. A novel activation of Ca2+-activated Cl− channel in Xenopus oocytes by Ginseng saponins: evidence for the involvement of phospholipase C and intracellular Ca2+ mobilization. Br J Pharmacol. 2001;132:641–8. doi: 10.1038/sj.bjp.0703856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Hwang SH, Shin TJ, Choi SH, Cho HJ, Lee BH, Pyo MK, et al. Gintonin, newly identified compounds from ginseng, is novel lysophosphatidic acids-protein complexes and activates G protein-coupled lysophosphatidic acid receptors with high affinity. Mol Cells. 2012;33:151–62. doi: 10.1007/s10059-012-2216-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Choi JW, Chun J. Lysophospholipids and their receptors in the central nervous system. Biochim Biophys Acta. 2013;1831:20–32. doi: 10.1016/j.bbalip.2012.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Kim JH, Cho SY, Lee JH, Jeong SM, Yoon IS, Lee BH, et al. Neuroprotective effects of ginsenoside Rg3 against homocysteine-induced excitotoxicity in rat hippocampus. Brain Res. 2007;1136:190–9. doi: 10.1016/j.brainres.2006.12.047. [DOI] [PubMed] [Google Scholar]
  11. Shin TJ, Kim HJ, Kwon BJ, Choi SH, Kim HB, Hwang SH, et al. Gintonin, a ginseng-derived novel ingredient, evokes long-term potentiation through N-methyl-D-aspartic acid receptor activation: Involvement of LPA receptors. Mol Cells. 2012;34:563–72. doi: 10.1007/s10059-012-0254-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Choi KT. Botanical characteristics, pharmacological effects and medicinal components of Korean Panax ginseng C A Meyer. Acta Pharmacol Sin. 2008;29:1109–18. doi: 10.1111/j.1745-7254.2008.00869.x. [DOI] [PubMed] [Google Scholar]
  13. Nag SA, Qin JJ, Wang W, Wang MH, Wang H, Zhang R. Ginsenosides as anticancer agents: in vitro and in vivo activities, structure-activity relationships, and molecular mechanisms of action. Front Pharmacol. 2012;3:25. doi: 10.3389/fphar.2012.00025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Lee JH, Jeong SM, Kim JH, Lee BH, Yoon IS, Choi SH, et al. Effects of ginsenosides and their metabolites on voltage-dependent Ca2+ channel subtypes. Mol Cells. 2006;21:52–62. [PubMed] [Google Scholar]
  15. Choi SH, Shin TJ, Hwang SH, Lee BH, Kang J, Kim HJ, et al. Ginsenoside Rg(3) decelerates hERG K+ channel deactivation through Ser631 residue interaction. Eur J Pharmacol. 2011;663:59–67. doi: 10.1016/j.ejphar.2011.05.006. [DOI] [PubMed] [Google Scholar]
  16. Choi SH, Shin TJ, Lee BH, Chu DH, Choe H, Pyo MK, et al. Ginsenoside Rg3 activates human KCNQ1 K+ channel currents through interacting with the K318 and V319 residues: a role of KCNE1 subunit. Eur J Pharmacol. 2010;637:138–47. doi: 10.1016/j.ejphar.2010.04.001. [DOI] [PubMed] [Google Scholar]
  17. Lee JH, Jeong SM, Kim JH, Lee BH, Yoon IS, Choi SH, et al. Characteristics of ginsenoside Rg3-mediated brain Na+ current inhibition. Mol Pharmacol. 2005;68:1114–26. doi: 10.1124/mol.105.015115. [DOI] [PubMed] [Google Scholar]
  18. Lee JH, Lee BH, Choi SH, Yoon IS, Pyo MK, Shin TJ, et al. Ginsenoside Rg3 inhibits human Kv1.4 channel currents by interacting with the Lys531 residue. Mol Pharmacol. 2008;73:619–26. doi: 10.1124/mol.107.040360. [DOI] [PubMed] [Google Scholar]
  19. Kang DI, Lee JY, Yang JY, Jeong SM, Lee JH, Nah SY, et al. Evidence that the tertiary structure of 20(S)-ginsenoside Rg(3) with tight hydrophobic packing near the chiral center is important for Na+ channel regulation. Biochem Biophys Res Commun. 2005;333:1194–201. doi: 10.1016/j.bbrc.2005.06.026. [DOI] [PubMed] [Google Scholar]
  20. Lee BH, Choi SH, Pyo MK, Shin TJ, Hwang SH, Kim BR, et al. A role for Leu247 residue within transmembrane domain 2 in ginsenoside-mediated alpha7 nicotinic acetylcholine receptor regulation. Mol Cells. 2009;27:591–9. doi: 10.1007/s10059-009-0073-4. [DOI] [PubMed] [Google Scholar]
  21. Kim CS, Son SJ, Kim HS, Kim YD, Lee KS, Jeon BH, et al. Modulating effect of ginseng saponins on heterologously expressed HERG currents in Xenopus oocytes. Acta Pharmacol Sin. 2005;26:551–8. doi: 10.1111/j.1745-7254.2005.00116.x. [DOI] [PubMed] [Google Scholar]
  22. Choi SE, Choi S, Lee JH, Whiting PJ, Lee SM, Nah SY. Effects of ginsenosides on GABAA receptor channels expressed in Xenopus oocytes. Arch Pharm Res. 2003;26:28–33. doi: 10.1007/BF03179927. [DOI] [PubMed] [Google Scholar]
  23. Noh JH, Choi S, Lee JH, Betz H, Kim JI, Park CS, et al. Effects of ginsenosides on glycine receptor alpha1 channels expressed in Xenopus oocytes. Mol Cells. 2003;15:34–9. [PubMed] [Google Scholar]
  24. Choi SH, Shin TJ, Lee BH, Hwang SH, Lee SM, Lee BC, et al. Ginsenoside Rg3 enhances large conductance Ca2+-activated potassium channel currents: a role of Tyr360 residue. Mol Cells. 2011;31:133–40. doi: 10.1007/s10059-011-0017-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Kwan CY, Kwan TK. Effects of Panax notoginseng saponins on vascular endothelial cells in vitro. Acta Pharmacol Sin. 2000;21:1101–5. [PubMed] [Google Scholar]
  26. Li Z, Chen X, Niwa Y, Sakamoto S, Nakaya Y. Involvement of Ca2+-activated K+ channels in ginsenosides-induced aortic relaxation in rats. J Cardiovasc Pharmacol. 2001;37:41–7. doi: 10.1097/00005344-200101000-00005. [DOI] [PubMed] [Google Scholar]
  27. Kim ND, Kang SY, Park JH, Schini-Kerth VB. Ginsenoside Rg3 mediates endothelium-dependent relaxation in response to ginsenosides in rat aorta: role of K+ channels. Eur J Pharmacol. 1999;367:41–9. doi: 10.1016/s0014-2999(98)00898-x. [DOI] [PubMed] [Google Scholar]
  28. Chung I, Kim ND. Ginseng saponins enhance maxi Ca2+-activated K+ currents of the rabbit coronary artery smooth muscle cells. J Ginseng Res. 1999;23:230–4. [Google Scholar]
  29. Chung I, Lee JS. Ginsenoside Rg3 increases the ATP-sensitive K+ channel activity in the smooth muscle of the rabbit coronary artery. J Ginseng Res. 1999;23:235–8. [Google Scholar]
  30. Jeon BH, Kim CS, Kim HS, Park JB, Nam KY, Chang SJ. Effect of Korean red ginseng on blood pressure and nitric oxide production. Acta Pharmacol Sin. 2000;21:1095–100. [PubMed] [Google Scholar]
  31. He H, Xu J, Xu Y, Zhang C, Wang H, He Y, et al. Cardioprotective effects of saponins from Panax japonicus on acute myocardial ischemia against oxidative stress-triggered damage and cardiac cell death in rats. J Ethnopharmacol. 2012;140:73–82. doi: 10.1016/j.jep.2011.12.024. [DOI] [PubMed] [Google Scholar]
  32. Li HX, Han SY, Ma X, Zhang K, Wang L, Ma ZZ, et al. The saponin of red ginseng protects the cardiac myocytes against ischemic injury in vitro and in vivo. Phytomedicine. 2012;19:477–83. doi: 10.1016/j.phymed.2012.01.002. [DOI] [PubMed] [Google Scholar]
  33. Bai CX, Takahashi K, Masumiya H, Sawanobori T, Furukawa T. Nitric oxide-dependent modulation of the delayed rectifier K+ current and the L-type Ca2+ current by ginsenoside Re, an ingredient of Panax ginseng, in guinea-pig cardiomyocytes. Br J Pharmacol. 2004;142:567–75. doi: 10.1038/sj.bjp.0705814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Furukawa T, Bai CX, Kaihara A, Ozaki E, Kawano T, Nakaya Y, et al. Ginsenoside Re, a main phytosterol of Panax ginseng, activates cardiac potassium channels via a nongenomic pathway of sex hormones. Mol Pharmacol. 2006;70:1916–24. doi: 10.1124/mol.106.028134. [DOI] [PubMed] [Google Scholar]
  35. Attele AS, Wu JA, Yuan CS. Ginseng pharmacology: multiple constituents and multiple actions. Biochem Pharmacol. 1999;58:1685–93. doi: 10.1016/s0006-2952(99)00212-9. [DOI] [PubMed] [Google Scholar]
  36. Kim TW, Choi HJ, Kim NJ, Kim DH. Anxiolytic-like effects of ginsenosides Rg3 and Rh2 from red ginseng in the elevated plus-maze model. Planta Med. 2009;75:836–9. doi: 10.1055/s-0029-1185402. [DOI] [PubMed] [Google Scholar]
  37. Kim JH, Yoon IS, Lee BH, Choi SH, Lee JH, Jeong SM, et al. Effects of Korean red ginseng extract on cisplatin-induced nausea and vomiting. Arch Pharm Res. 2005;28:680–4. doi: 10.1007/BF02969358. [DOI] [PubMed] [Google Scholar]
  38. Choi S, Kim HJ, Ko YS, Jeong SW, Kim YI, Simonds WF, et al. G αq/11 coupled to mammalian phospholipase C beta 3-like enzyme mediates the ginsenoside effect on Ca2+-activated Cl− current in the Xenopus oocyte. J Biol Chem. 2001;276:48797–802. doi: 10.1074/jbc.M104346200. [DOI] [PubMed] [Google Scholar]
  39. Lee JH, Jeong SM, Lee BH, Noh HS, Kim BK, Kim JI, et al. Prevention of ginsenoside-induced desensitization of Ca2+-activated Cl− current by microinjection of inositol hexakisphosphate in Xenopus laevis oocytes: involvement of GRK2 and beta-arrestin I. J Biol Chem. 2004;279:9912–21. doi: 10.1074/jbc.M310824200. [DOI] [PubMed] [Google Scholar]
  40. Pyo MK, Shin TJ, Choi SH, Lee BH, Pyo MK, Lee JH, et al. Novel glycoproteins from ginseng. J Ginseng Res. 2011;35:92–103. [Google Scholar]
  41. Hwang SH, Shin EJ, Shin TJ, Lee BH, Choi SH, Kang J, et al. Gintonin, a ginseng-derived lysophosphatidic acid receptor ligand, attenuates Alzheimer's disease-related neuropathies: involvement of non-amyloidogenic processing. J Alzheimers Dis. 2012;31:207–23. doi: 10.3233/JAD-2012-120439. [DOI] [PubMed] [Google Scholar]
  42. Tigyi G, Miledi R. Lysophosphatidates bound to serum albumin activate membrane currents in Xenopus oocytes and neurite retraction in PC12 pheochromocytoma cells. J Biol Chem. 1992;267:21360–7. [PubMed] [Google Scholar]
  43. Yoon HR, Kim H, Cho SH. Quantitative analysis of acyl-lysophosphatidic acid in plasma using negative ionization tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci. 2003;788:85–92. doi: 10.1016/s1570-0232(02)01031-0. [DOI] [PubMed] [Google Scholar]
  44. Houben AJ, Moolenaar WH. Autotaxin and LPA receptor signaling in cancer. Cancer Metastasis Rev. 2011;30:557–65. doi: 10.1007/s10555-011-9319-7. [DOI] [PubMed] [Google Scholar]
  45. Hwang SH, Lee BH, Kim HJ, Cho HJ, Shin HC, Im KS, et al. Suppression of metastasis of intravenously-inoculated B16/F10 melanoma cells by the novel ginseng-derived ingredient, gintonin: Involvement of autotaxin inhibition. Int J Oncol. 2013;42:317–26. doi: 10.3892/ijo.2012.1709. [DOI] [PubMed] [Google Scholar]
  46. Tokumura A, Majima E, Kariya Y, Tominaga K, Kogure K, Yasuda K, et al. Identification of human plasma lysophospholipase D, a lysophosphatidic acid-producing enzyme, as autotaxin, a multifunctional phosphodiesterase. J Biol Chem. 2002;277:39436–42. doi: 10.1074/jbc.M205623200. [DOI] [PubMed] [Google Scholar]
  47. Umezu-Goto M, Kishi Y, Taira A, Hama K, Dohmae N, Takio K, et al. Autotaxin has lysophospholipase D activity leading to tumor cell growth and motility by lysophosphatidic acid production. J Cell Biol. 2002;158:227–33. doi: 10.1083/jcb.200204026. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Acta Pharmacologica Sinica are provided here courtesy of Nature Publishing Group

RESOURCES