Ginseng
Phytochemicals

Author: elisa giorda
Date: 05/12/2010

Description

Panax ginseng (roots and its extract) is well known as the most important herbal medicine in the Orient, particularly in Korea, China, Japan and also in North America (typically in cooler climates) for more 2000 years, as a medicinal plant with mysterious powers, traditionally used to revitalize the body and mind, increased physical strength, prevent aging and increased vigor. But now it seems that the scientists have discovered new important application. Among the several kinds of Panax ginseng products, Korean red ginseng has the most potent multiple pharmacological actions for treating various human diseases as cardiovascular disease, diabetes mellitus, rheumatoid arthritis, against stress, fatigue, cancer, hypertension, atherosclerotic disorders , ischemic injury, sexual dysfunction,…

Characteristics

The botanical classifications of Korean ginseng are as follows:
Phylum: Embryophyta Siphonogama
Subphylum: Angiospermae
Class: Dicotyledoneae
Subclass: Archichlamydeae
Order: Umbelliflorae
Family: Araliaceae
Genus: Panax

The genus name Panax is a compound of the Greek words “pan” and “axos”. “Pan” means “all” and “axos” means “treat”, which represents “treats all diseases”.
It is compound by ginsenosides, eleutherosides, sterols, flavonoids, peptides, vitamins, polyacetylenes, minerals, b-elemine and choline.

Active components of Korean Ginseng

Ginseng saponins
The key active ingredients of ginseng are known to be a compound carbohydrate (a compound of alcohol or phenol and sugar) called saponin or ginsenosides. This property excites and suppresses the central nervous system, adjusts metabolism, lowers blood glucose, improves muscular activities, stimulates the endocrine system and maintains hormone levels properly.
The name saponin is derived from the phenomenon in which fine foam comes from its solution like a piece of soap. In general, glycosides form a very high polarization compound that causes hemolysis.
Ginsenosides are:
Ginsenoside-Rg1 (C42H72O14 2H2O)
Ginsenoside-Rb1 (C54H92O23 3H2O)
Ginsenoside-Rf (C42H72O14 2H2O)
Based on their chemical structures, saponin compounds are divided into three groups such as protopanaxadiol (PD), protopanaxatriol (PT), and oleanane.
Korean ginseng has better pharmacological efficacy compared with other ginseng, because it has ginsenosides Ra1, Ra2, Ra3, malonyl- G-Rb1, malonyl-G-Rb2, malonyl-G-Rc, malonyl-GRd, Rs1, Rs2, Rs3, Rg3, Rg5, Rh2, K-R2, Rf, Rf2, 20®- G-Rg2, Rg6, 20®-G-Rh1, 20(E)-G-F4, Rh4, K-R1, and polyacetyleneginsenoside-Ro.

Figure SEQ Figura \* ARABIC 1. Structure of ginsenosides.

Figure SEQ Figura \* ARABIC 2. Chemical structures of 20(S)-ginsenosides Rg3, ginsenosides Rh2, Rg1, Re and Rc.

Non-saponin active components
Korean ginseng has important substances other than ginsenoside that show anti-tumor, anti-oxidative, anti-diabetic and anti-inflammatory effects, and even hematosis action.
These components are panaxydol, panaxynol and panaxytriol, have showed anti-tumor and anti oxidative effects.
Phenol compounds such as maltol show anti-aging effects as a lipid per-oxidation inhibitor. Maltol is a phenol compound specific to Korean red ginseng. Carbohydrates are extracted in a water solution, and make up 60%–70% of total extracts. Monosaccharide, disaccharide, and trisaccharide and polysaccharide display a hypoglycemic effect, increased immune system and anti-ulcerative activity.

Pharmacological effects of Korean ginseng

Scientifically proven pharmacological effects of Korean ginseng are as follows:
Efficacy of improving cerebral functions

Korean ginseng (extract and saponin component) is found to have efficacy to increase attention, learning skill, to improve memory, intellectual ability and to reduce memory loss.
Anti-diabetic efficacy

Ginseng saponin and ginsenoside Rh2 reduce high blood glucose. Korean ginseng includes insulin secretion stimulating activity and insulin-like activity substance.

Efficacy of adjusting blood pressure

It is rumored that taking ginseng leads to high blood pressure. Recently, facts about the biochemical and pharmacological activities of ginseng related to blood pressure control are being gradually revealed by modern medical studies of effective ingredients and the efficacy of ginseng.
Korean red ginseng can exert an antihypertensive effect, elevating NO concentration and reducing cardiac frequency.

Efficacy of anti-fatigue and anti-stress

It is observed that Korean ginseng enforces the power, increases resistance to stress and prevents against the fatigue.
Ginsenoside Rb1 is the component, at least a major component, in ginseng with anti-stress activity: Rb1 may block stress response at a very early stage and consequently prevents adverse effects of stress.
Panax ginseng extract has potential protective effects against the oxidative stress.

Efficacy of improving liver functions

Korean ginseng has the activity of prompting the detoxification of toxic substances, protecting the liver from damage, and generating and promoting the regeneration and recovery of the liver. It has also an anti-hepatitis activity.

Efficacy of relieving pain

Korean ginseng was found to have an action inhibiting the Ca channel that exists on sensory neurons and is related to the pain transmission neuron.

Efficacy on climacteric disorders

Tests showed that the administration (3 g/day, 2 months) of Korean red ginseng in climacteric disorders was efficient.

Efficacy of improving male sexual dysfunctions

Korean red ginseng was evaluated as effective for the improvement of sexual function and for defends males against sexual behavior disorders caused by stress. A clinical test indicated that the group who were given Korean red ginseng showed better effects in erectile function, sexual desire and satisfaction of patients.

Efficacy of inhibiting AIDS virus (HIV) growth

AIDS is a chronic disease, and an examination of the anti-HIV activity of Korean red ginseng components revealed that HIV growth inhibition activity was observed in crude saponin components.
The possibility that the occurrence of the deleted nef gene might be associated with long-term intake of Korean red ginseng.

Efficacy of anti-oxidation and anti-aging activity

Korean ginseng has the anti-oxidation effect of inhibiting the increase in harmful free radical formation and lipid peroxidation.

Efficacy of increasing immunization functions

Korean ginseng extracts have the ability to revive cellular immune response.

Efficacy of preventing cancer and activate antitumor
Immunity

Studies of Korean ginseng intake and cancer cases show that those who take Korean ginseng are less likely to contract various cancers such as cancer of the stomach, liver , lung, breast, skin melanoma and human ovarian carcinoma, than those who do not take it.

- Efficacy of inhibiting cancer cell growth

Saponin and non-saponin ingredients of ginseng contain activity inhibiting a variety of cancer cell growth.
It was suggested that ginsenoside Rp1 can be used for anticancer action.

- Efficacy of inhibiting the metastasis of cancer cells

Efficacy of inhibiting metastasis of cancer was observed for example in lung tumor.

- Efficacy of increasing anti-tumor activity of anticancer drug

Combination of Korean ginseng with mitomycin C increased the anti-cancer effect of Korean ginseng in comparison to the treatment of the anti-tumor drug (mitomycin C) alone.

- Efficacy of inhibiting tolerance formation and reducing
side effects of anti-tumor drugs

Efficacy of increasing cytotoxicity against tumor cells and reducing the side effects of anti-tumor drugs with the combination of Korean ginseng with anti-tumor drugs by inhibiting resistance formation that is a hindrance to anti-tumor chemotherapy.

Figure SEQ Figura \* ARABIC 3. Schematic overview of ginsenosides-mediated genomic and non-genomic pathways. Ginsenosides possess a steroid- like skeleton composed of four trans-rings with different degrees of glyco-substitution. They are amphipathic in nature and can exhibit their actions at different cellular locations; such as the plasma membrane, cytosol and nucleus. Through the nongenomic pathway (indicated by red arrows), (i) they can initiate their actions by binding with the transmembrane receptors (e.g. ATPase pump, ion transporters and channels, voltage-gated channels and G-proteins) and subsequently activating the associated downstream signaling cascades. Moreover, they can intercalate into the plasma membrane resulting in an alteration of membrane fluidity and a trigger of a series of cellular responses. (ii) binding with steroid hormone receptors (SHRs) including glucocorticoid receptor (GR), estrogen receptor (ER), progesterone receptor (PR), androgen receptor (AR) and mineralocorticoid receptor (MR) present inside or outside the nucleus by using their hydrophobic backbone is another alternative to trigger downstream cellular responses. Those activated (phosphorylated) SHRs can activate the target molecules through a signaling cascade that brings about various cellular responses. (iii) the ligand-bound SHRs can translocate into the nucleus, where they regulate gene transcription by binding with the specific Response Elements (XRE). This is the so called 'genomic pathway' (indicated by blue arrows). Consequently, the altered gene products can affect the final cellular responses.

Figure SEQ Figura \* ARABIC 4. Schematic overview of ginsenoside Rg1-mediated angiogenic action in HUVEC. Ginsenoside Rg1, which acts as a functional ligand of glucocorticoid receptor (GR) (either cytosol GR or membrane-bound GR-mGR), promotes angiogenesis through both non-genomic and genomic pathways. Through the non-genomic pathway, it increases nitric oxide (NO) production via the PI3K-Akt pathway: GR → phosphatidylinositol-3 kinase (PI3K)/Akt pathway → endothelial nitric oxide synthase (eNOS) pathway. Rg1 also increases vascular endothelial growth factor (VEGF) production through the GR → PI3K/Akt → GSK3β → β-catenin/TCF pathway. Gene expression profiling data indicated that Rg1 could increase the expression of a group of genes (e.g. Rho A, RhoB, IQGAP1, LAMA4, CALM2 and Vav2) which are related to cell-cell adhesion, migration and cytoskeletal remodeling.

CYP450:
Cytocrome P450 is an enzymatic family of hemoprotein located primarly in hepatocytes. They are involved in the organism detossification, because they are able to act on hexogen substrate (drugs and external toxines) and on endogen one. The most important isoforms of CyP450 involved in drugs metabolism are CYP3A4, CYP2D6, CYP2C9, CYP1A2 e CYP2E1.
Herbal remedies may interact with medicinal drugs, altering the pharmacokinetic characteristics, so studies on effects on CYP450 have been tested.
Elevated cytochrome P450 activity, translated into a more rapid metabolic rate, may result in a decrease in drug plasma concentrations to sub therapeutic levels and total loss of the pharmacological effect.
Conversely, suppression of cytochrome P450 activity may trigger a rise in plasma drug levels leading to an undesirable exaggerated pharmacological effect and the appearance of toxic symptoms commensurate with overdose. However, the reported effects of ginseng on P450 activities in the clinical trials are somewhat contradictory, but now we know that ginseng can interacts with some drugs.

Pgp:

P-glycoprotein (Pgp) is a 170 kDa phosphorylated glycoprotein encoded by human MDR1 gene. It is responsible for the systemic disposition of numerous structurally and pharmacologically unrelated lipophilic and amphipathic drugs, carcinogens, toxins, and other xenobiotics in many organs, such as the intestine, liver, kidney, and brain.
Like cytochrome P450s (CYP3A4), Pgp is vulnerable to inhibition, activation, or induction by herbal constituents, as ginsenosides.

Molecular Mechanisms:

Recent research has focused on their value in human cancer prevention and treatment. Scientists have discovered new ginseng applications, for example it seems to be an antitumor herb.

Colorectal cancer:
Recent studies showed that the combination of 5-FU ( one of the most widely used chemotherapeutic agents in first-line therapy for colorectal cancer) and Panaxadiol significantly enhanced the percentage of apoptotic cells in human colorectal cancer ( through the regulation of cell cycle transition and the induction of apoptotic cells) when compared with the corresponding cell groups treated by 5-FU alone.

Prostate cancer:
Scientists have found that 25-hydroxyprotopanaxadiol(25-OH-PPD), but not 25-hydroxyprotopanaxatriol(25-OH-PPT), from Panax ginseng has an anti-cancer activity in vitro: it inhibited prostate cancer cell growth and proliferation, induced apoptosis, and led to arrest in the G1 phase of the cell cycle.

Bladder cancer:
Ginseng seems to inhibit benzoapirene, a carcinogenic compound involved in bladder cancer, acting, probably, a chemo preventive action.
The chemo preventive effect of Panax ginseng may be due, in part, to ginsenosides’s ability to compete with aryl hydrocarbon receptors (that play an important roles in carcinogenesis, development, differentiation).

Interactions:

It is wrong to think that natural therapies cannot have adverse effects; in fact it is known that herbal medicines can give serious clinical interactions when co-administered with prescription medicines. For example, ginseng may interact with Warfarin, an anticoagulant drug; it induces a decrease of protrombine time.

Figure5. WARFARIN

Ginseng, moreover, may interacts with oral hypoglycemic drugs, so, without a check on glucose hematic levels, it may induce hypoglycemia.
Ginseng can also give similar effects to the estrogenic ones, because its compounds have a chemical structure like testosterone, estrogens and glucocorticoides. So it must not to be used in pregnancy and in women that use substitutive hormonal therapy.
It seems to interacts with imatinib through CYP3A4, increasing hepatotoxic effects. So, it is necessary to check liver function.
It may increase haematic digossina level, and it may hind with antidepressive effects (as phenelzine).
Moreover, simultaneous administration of ginseng with anti-HIV drugs may cause significant elevation in plasma drug levels, for example it may interact with ritonavir.
.

Figure 6. Chemical structure of ritonavir

Adverse events:
Ginseng may cause insomnia, headache, and tremulousness, irritability. These events are not so frequent and they occurred only if ginseng is somministrated at elevated dosages and for long time. It is important to take ginseng for no more than 45-60 days consecutively.

Bibliography:
-J Ethnopharmacol. 2010 Sep 8. [Epub ahead of print]
Cho SM, Shimizu M, Lee CJ, Han DS, Jung CK, Jo JH, Kim YM.
- Drug Metab Pharmacokinet.2010-,25(3):307-13
-Planta Med.2010 Oct 26. Epub ahead of print
-Pharmacology.2010;85(6):350-6. Epub 2010 Jun 4
-J Ethnopharmacol. 2010 Aug 5. [Epub ahead of print]
Park HJ, Han ES, Park DK.
-J Cell Biochem. 2010 Jul 27. [Epub ahead of print]
Wong VK, Cheung SS, Li T, Jiang ZH, Wang JR, Dong H, Yi XQ, Zhou H, Liu L.
-Phytochemistry. 2010 Sep;71(13):1514-20. Epub 2010 Jun 22.
Liu L, Zhu XM, Wang QJ, Zhang DL, Fang ZM, Wang CY, Wang Z, Sun BS, Wu H, Sung
CK.
- Biol Pharm Bull. 2010;33(3):468-72.
Kwak YS, Kyung JS, Kim JS, Cho JY, Rhee MH.
-BMC Cancer. 2009 Dec 18;9:449.
Cho SH, Chung KS, Choi JH, Kim DH, Lee KT.
-Tetrahedron Lett. 2008 Dec 8;49(50):7178-7179
Ng F, Yun H, Lei X, Danishefsky SJ, Fahey J, Stephenson K, Flexner C, Lee L.
- Curr Vasc Pharmacol. 2009 Jul;7(3):293-302.
JM, Yao Q, Chen C.
- Cancer Chemother Pharmacol. 2009 Nov;64(6):1097-104. Epub 2009 Mar 11.
Li XL, Wang CZ, Mehendale SR, Sun S, Wang Q, Yuan CS.
- Acta Pharmacol Sin. 2009 Jan;30(1):61-9. Epub 2008 Dec 8.
Chen RJ, Chung TY, Li FY, Lin NH, Tzen JT.
- J Ethnopharmacol. 2009 Jan 21;121(2):304-12. Epub 2008 Nov 12.
Park SE, Park C, Kim SH, Hossain MA, Kim MY, Chung HY, Son WS, Kim GY, Choi YH,
Kim ND.
- Acta Pharmacol Sin. 2008 Sep;29(9):1109-18. Biol Pharm Bull. 2008 May;31(5):826-30.
Choi KT.
- Biol Pharm Bull. 2008 May;31(5):826-30.
Kim YJ, Kwon HC, Ko H, Park JH, Kim HY, Yoo JH, Yang HO.
- Prostate. 2008 Jun 1;68(8):809-19.
Wang W, Rayburn ER, Hao M, Zhao Y, Hill DL, Zhang R, Wang H.
- Molecules. 2007 Sep 5;12(9):2140-50.
Lei J, Li X, Gong XJ, Zheng YN.
- Biol Pharm Bull. 2007 Sep;30(9):1674-9.
Kim YM, Namkoong S, Yun YG, Hong HD, Lee YC, Ha KS, Lee H, Kwon HJ, Kwon YG, Kim
YM.
- Cell Biol Int. 2007 Oct;31(10):1265-73. Epub 2007 May 21.
Ming YL, Song G, Chen LH, Zheng ZZ, Chen ZY, Ouyang GL, Tong QX.
- Chin Med. 2007 May 15;2:6.
Yue PY, Mak NK, Cheng YK, Leung KW, Ng TB, Fan DT, Yeung HW, Wong RN.
- J Agric Food Chem. 2007 Apr 18;55(8):2816-23. Epub 2007 Mar 24.
Kim HY, Kim K.
- Cancer Chemother Pharmacol. 2007 Jun;60(1):69-79. Epub 2006 Sep 29.
Wang CZ, Luo X, Zhang B, Song WX, Ni M, Mehendale S, Xie JT, Aung HH, He TC, Yuan
CS.
- Biochem Biophys Res Commun. 2006 Oct 27;349(3):987-94. Epub 2006 Aug 30.
Min JK, Kim JH, Cho YL, Maeng YS, Lee SJ, Pyun BJ, Kim YM, Park JH, Kwon YG.
- Acta Pharmacol Sin. 2006 Aug;27(8):1000-6.
Ma ZC, Gao Y, Wang YG, Tan HL, Xiao CR, Wang SQ.
- Biochem Pharmacol. 2006 Aug 14;72(4):437-45. Epub 2006 May 12.
Yue PY, Wong DY, Wu PK, Leung PY, Mak NK, Yeung HW, Liu L, Cai Z, Jiang ZH, Fan
TP, Wong RN.
- J Cell Biochem. 2006 Aug 15;98(6):1471-81.
Zhang HS, Wang SQ.
- Life Sci. 2006 Jun 6;79(2):175-81. Epub 2006 Feb 17.
Ma ZC, Gao Y, Wang J, Zhang XM, Wang SQ.
- Biosci Biotechnol Biochem. 2005 May;69(5):891-5.
Kim JY, Lee HJ, Kim JS, Ryu JH.
- Acta Pharmacol Sin. 2005 Feb;26(2):143-9.
Cheng Y, Shen LH, Zhang JT.
- Chin Med J (Engl). 2004 Jun;117(6):871-6.
JP, Ma ZC, Yang J, Huang J, Wang SR, Wang SQ.
- J Invest Dermatol. 2003 Sep;121(3):607-13.
Lee EH, Cho SY, Kim SJ, Shin ES, Chang HK, Kim DH, Yeom MH, Woe KS, Lee J, Sim
YC, Lee TR.
- Mutat Res. 2003 Feb-Mar;523-524:75-85.
Keum YS, Han SS, Chun KS, Park KK, Park JH, Lee SK, Surh YJ.
- J Korean Med Sci. 2001 Dec;16 Suppl:S38-41.
Surh YJ, Na HK, Lee JY, Keum YS.
- Cancer Lett. 2000 Mar 13;150(1):41-8.
Keum YS, Park KK, Lee JM, Chun KS, Park JH, Lee SK, Kwon H, Surh YJ.
- Cancer Lett. 1999 Dec 1;147(1-2):11-6.
Konoshima T, Takasaki M, Ichiishi E, Murakami T, Tokuda H, Nishino H, Duc NM,
Kasai R, Yamasaki K.
- Biol Pharm Bull. 1999 Oct;22(10):1150-2.
Konoshima T, Takasaki M, Tokuda H.
- Biol Pharm Bull. 1998 Aug;21(8):834-8.
Konoshima T, Takasaki M, Tokuda H, Nishino H, Duc NM, Kasai R, Yamasaki K.
- Immunopharmacology. 1998 Jan;38(3):287-94.
Sonoda Y, Kasahara T, Mukaida N, Shimizu N, Tomoda M, Takeda T.
-Ann Pharmacother. 2010 May;44(5):926-8. Epub 2010 Mar 23
Bilgi N, Bell K, Ananthakrishnan AN, Atallah E.
-Planta Med. 2010 Feb;76(3):245-50. Epub 2009 Sep 11.
Xia CH, Sun JG, Wang GJ, Shang LL, Zhang XX, Zhang R, Peng Y, Wang XJ, Hao HP,
Xie L, Roberts MS.
-Drugs. 2009;69(13):1777-98
Izzo AA, Ernst E.
-Drug Metabol Drug Interact. 2009;24(1):17-35.
Wanwimolruk S, Wong K, Wanwimolruk P.
-Planta Med. 2009 Jun;75(8):797-802. Epub 2009 Mar 5.
Lai L, Hao H, Liu Y, Zheng C, Wang Q, Wang G, Chen X.
-Eur J Pharmacol. 2008 Dec 28;601(1-3):73-8. Epub 2008 Nov 11.
Wang Y, Ye X, Ma Z, Liang Q, Lu B, Tan H, Xiao C, Zhang B, Gao Y.
- PLoS One. 2008 Jul 16;3(7):e2697.
Hao M, Zhao Y, Chen P, Huang H, Liu H, Jiang H, Zhang R, Wang H.
- Planta Med. 2007 Jul;73(8):731-41. Epub 2007 Jul 5.
Etheridge AS, Black SR, Patel PR, So J, Mathews JM.
-Toxicol Sci. 2006 Jun;91(2):356-64. Epub 2006 Mar 17.
Liu Y, Zhang JW, Li W, Ma H, Sun J, Deng MC, Yang L.
-Planta Med. 2006 Feb;72(2):126-31.
Liu Y, Ma H, Zhang JW, Deng MC, Yang L.
- Drugs Aging. 2005;22(6):525-39.
Gurley BJ, Gardner SF, Hubbard MA, Williams DK, Gentry WB, Cui Y, Ang CY.
-Toxicol Rev. 2004;23(4):239-49.
Delgoda R, Westlake AC.
-J Pharm Pharmacol. 2003 Nov;55(11):1553-9.
Saruwatari J, Nakagawa K, Shindo J, Nachi S, Echizen H, Ishizaki T.
- J Clin Pharmacol. 2003 Jun;43(6):643-8.
Anderson GD, Rosito G, Mohustsy MA, Elmer GW.
- Clin Pharmacol Ther. 2002 Sep;72(3):276-87.
Gurley BJ, Gardner SF, Hubbard MA, Williams DK, Gentry WB, Cui Y, Ang CY.
-Xenobiotica. 2002 Jun;32(6):451-78.
Ioannides C.
-J Nutr Health Aging. 1998;2(3):162-6.Lee DW, Yu BP.
Lee DW, Yu BP.
-Life Sci. 1999;65(15):PL209-14.
Henderson GL, Harkey MR, Gershwin ME, Hackman RM, Stern JS, Stresser DM.
-Drug Chem Toxicol. 1987;10(3-4):227-36.
Lee FC, Park JK, Ko JH, Lee JS, Kim KY, Kim EK.
-Oncologist. 2007 Aug;12(8):927-41.
Marchetti S, Mazzanti R, Beijnen JH, Schellens JH.
-Integr Cancer Ther. 2009 Mar;8(1):29-36. Epub 2009 Jan 27.
Engdal S, Klepp O, Nilsen OG.
-Am J Ther. 2004 Jul-Aug;11(4):262-77.
Patel J, Buddha B, Dey S, Pal D, Mitra AK.
- Expert Opin Drug Metab Toxicol. 2009 Jul;5(7):789-801.
Zhang W, Han Y, Lim SL, Lim LY.
-Clin Pharmacokinet. 2008;47(6):383-97.
Butterweck V, Derendorf H.
-Nephrology (Carlton). 2008 Jun;13(4):337-47. Epub 2008 Mar 17.
Nowack R.
-Curr Pharm Des. 2006;12(35):4649-64.
Yang XX, Hu ZP, Duan W, Zhu YZ, Zhou SF.
-Clin Infect Dis. 2006 Oct 15;43(8):1052-9. Epub 2006 Sep 8.
Lee LS, Andrade AS, Flexner C.
-Drug Metab Rev. 2006;38(3):477-514.
van den Bout-van den Beukel CJ, Koopmans PP, van der Ven AJ, De Smet PA, Burger
DM.
-Drug Metab Rev. 2004 Feb;36(1):57-104.
Zhou S, Lim LY, Chowbay B.

AddThis Social Bookmark Button