Category Archives: XIAP

Teng Long Bu Zhong Tang

Cancer: Colon

Action: Induces apoptosis, inhibits angiogenesis

CT26 colon carcinoma was established in BALB/c mice and treated with Teng Long Bu Zhong Tang (TLBZT), 5-Fu, or TLBZT plus 5-Fu. The tumor volumes were observed. TLBZT significantly inhibited CT26 colon carcinoma growth. TLBZT elicited apoptosis in CT26 colon carcinoma, accompanied by Caspase-3, 8, and 9 activation and PARP cleavage, and down-regulation of XIAP and Survivin. TLBZT also induced cell senescence in CT26 colon carcinoma, with concomitant up-regulation of p16 and p21 and down-regulation of RB phosphorylation.

In addition, angiogenesis and VEGF expression in CT26 colon carcinoma was significantly inhibited by TLBZT treatment. TLBZT exhibited significant anti-cancer effect, and enhanced the effects of 5-Fu in CT26 colon carcinoma, which may correlate with induction of apoptosis and cell senescence, and angiogenesis inhibition (Deng et al., 2013).

Reference

Deng S, Hu B, An HM, et al. (2013). Teng-Long-Bu-Zhong-Tang, a Chinese herbal formula, enhances anti-cancer effects of 5 – Fluorouracil in CT26 colon carcinoma. BMC Complement Altern Med, 13:128. doi: 10.1186/1472-6882-13-128.

Sanguinarine (See also chelerythrine)

Cancer:
Prostate, bladder, breast, colon, melanoma, leukemia

Action: Pro-oxidative, anti-inflammatory, apoptosis induction

AR+/AR- Prostate Cancer

Sanguinarine, a benzophenanthridine alkaloid derived from the bloodroot plant Sanguinaria canadensis (L.), has been shown to possess anti-microbial, anti-inflammatory, anti-cancer and anti-oxidant properties. It has been shown that sanguinarine possesses strong anti-proliferative and pro-apoptotic properties against human epidermoid carcinoma A431 cells and immortalized human HaCaT keratinocytes. Employing androgen-responsive human prostate carcinoma LNCaP cells and androgen-unresponsive human prostate carcinoma DU145 cells, the anti-proliferative properties of sanguinarine against prostate cancer were also examined.

The mechanism of the anti-proliferative effects of sanguinarine against prostate cancer were examined by determining the effect of sanguinarine on critical molecular events known to regulate the cell-cycle and the apoptotic machinery.

A highlight of this study was the fact that sanguinarine induced growth-inhibitory and anti-proliferative effects in human prostate carcinoma cells irrespective of their androgen status. To our knowledge, this is the first study showing the involvement of cyclin kinase inhibitor-cyclin-cyclin-dependent kinase machinery during cell-cycle arrest and apoptosis of prostate cancer cells by sanguinarine. These results suggest that sanguinarine may be developed as an agent for the management of prostate cancer (Adhami et al., 2004).

Breast Cancer

The effects of this compound were examined on reactive oxygen species (ROS) production and its association with apoptotic tumor cell death using a human breast carcinoma MDA-MB-231 cell line. Cytotoxicity was evaluated by trypan blue exclusion methods. Apoptosis was detected using DAPI staining, agarose gel electrophoresis and flow cytometer. The expression levels of proteins were determined by Western blot analyzes and caspase activities were measured using colorimetric assays.

These observations clearly indicate that ROS is involved in the early molecular events in the sanguinarine-induced apoptotic pathway. Data suggests that sanguinarine-induced ROS are key mediators of MMP collapse, which leads to the release of cytochrome c followed by caspase activation, culminating in apoptosis (Choi, Kim, Lee & Choi, 2008).

Leukemia

Sanguinarine, chelerythrine and chelidonine are isoquinoline alkaloids derived from the greater celandine. They possess a broad spectrum of pharmacological activities. It has been shown that their anti-tumor activity is mediated via different mechanisms, which can be promising targets for anti-cancer therapy.

This study focuses on the differential effects of these alkaloids upon cell viability, DNA damage, and nucleus integrity in mouse primary spleen and lymphocytic leukemic cells, L1210. Sanguinarine and chelerythrine produced a dose-dependent increase in DNA damage and cytotoxicity in both primary mouse spleen cells and L1210 cells. Chelidonine did not show a significant cytotoxicity or damage DNA in both cell types, but completely arrested growth of L1210 cells.

Data suggests that cytotoxic and DNA-damaging effects of chelerythrine and sanguinarine are more selective against mouse leukemic cells and primary mouse spleen cells, whereas chelidonine blocks proliferation of L1210 cells. The action of chelidonine on normal and tumor cells requires further investigation (Kaminsky, Lin, Filyak, & Stoika, 2008).

T-lymphoblastic Leukemia

Apoptogenic and DNA-damaging effects of chelidonine (CHE) and sanguinarine (SAN), two structurally related benzophenanthridine alkaloids isolated from Chelidonium majus, were compared. Both alkaloids induced apoptosis in human acute T-lymphoblastic leukemia MT-4 cells. Apoptosis induction by CHE and SAN in these cells were accompanied by caspase-9 and -3 activation and an increase in the pro-apoptotic Bax protein. An elevation in the percentage of MT-4 cells possessing caspase-3 in active form after their treatment with CHE or SAN was in parallel to a corresponding increase in the fraction of apoptotic cells.

The involvement of the mitochondria in apoptosis induction by both alkaloids was supported by cytochrome C elevation in cytosol, with an accompanying decrease in cytochrome C content in the mitochondrial fraction. At the same time, two alkaloids under study differed drastically in their cell-cycle phase-specific effects, since only CHE arrested MT-4 cells at the G2/M phase. It was previously demonstrated, that CHE, in contrast to SAN, does not interact directly with DNA. (Philchenkov, Kaminskyy, Zavelevich, & Stoika, 2008).

Sanguinarine, chelerythrine and chelidonine possess prominent apoptotic effects towards cancer cells. This study found that sanguinarine and chelerythrine induced apoptosis in human CEM T-leukemia cells, accompanied by an early increase in cytosolic cytochrome C that precedes caspases-8, -9 and -3 processing. Effects of sanguinarine and chelerythrine on mitochondria were confirmed by clear changes in morphology (3h), however chelidonine did not affect mitochondrial integrity.

Sanguinarine and chelerythrine also caused marked DNA damage in cells after 1h, but a more significant increase in impaired cells occurred after 6h. Chelidonine induced intensive DNA damage in 15–20% cells after 24h. Results demonstrated that rapid cytochrome C release in CEM T-leukemia cells exposed to sanguinarine or chelerythrine was not accompanied by changes in Bax, Bcl-2 and Bcl-X((L/S)) proteins in the mitochondrial fraction, and preceded activation of the initiator caspase-8 (Kaminskyy, Kulachkovskyy & Stoika, 2008).

Colorectal Cancer

The effects of sanguinarine, a benzophenanthridine alkaloid, was examined on reactive oxygen species (ROS) production, and the association of these effects with apoptotic cell death, in a human colorectal cancer HCT-116 cell line. Sanguinarine generated ROS, followed by a decrease in mitochondrial membrane potential (MMP), activation of caspase-9 and -3, and down-regulation of anti-apoptotic proteins, such as Bcl2, XIAP and cIAP-1. Sanguinarine also promoted the activation of caspase-8 and truncation of Bid (tBid).

Observations clearly indicate that ROS, which are key mediators of Egr-1 activation and MMP collapse, are involved in the early molecular events in the sanguinarine-induced apoptotic pathway acting in HCT-116 cells (Han, Kim, Yoo, & Choi, 2013).

Bladder Cancer

Although the effects of sanguinarine, a benzophenanthridine alkaloid, on the inhibition of some kinds of cancer cell growth have been established, the underlying mechanisms are not completely understood. This study investigated possible mechanisms by which sanguinarine exerts its anti-cancer action in cultured human bladder cancer cell lines (T24, EJ, and 5637). Sanguinarine treatment resulted in concentration-response growth inhibition of the bladder cancer cells by inducing apoptosis.

Taken together, the data provide evidence that sanguinarine is a potent anti-cancer agent, which inhibits the growth of bladder cancer cells and induces their apoptosis through the generation of free radicals (Han et al., 2013).

Melanoma

Sanguinarine is a natural isoquinoline alkaloid derived from the root of Sanguinaria canadensis and from other poppy fumaria species, and is known to have a broad spectrum of pharmacological properties. Current study has found that sanguinarine, at low micromolar concentrations, showed a remarkably rapid killing activity against human melanoma cells. Sanguinarine disrupted the mitochondrial transmembrane potential (ΔΨ m), released cytochrome C and Smac/DIABLO from mitochondria to cytosol, and induced oxidative stress. Thus, pre-treatment with the thiol anti-oxidants NAC and GSH abrogated the killing activity of sanguinarine. Collectively, data suggests that sanguinarine is a very rapid inducer of human melanoma caspase-dependent cell death that is mediated by oxidative stress (Burgeiro, Bento, Gajate, Oliveira, & Mollinedo, 2013).

References

Adhami YM, Aziz MH, Reagan-Shaw SR, et al. (2004). Sanguinarine causes cell-cycle blockade and apoptosis of human prostate carcinoma cells via modulation of cyclin kinase inhibitor-cyclin-cyclin-dependent kinase machinery. Mol Cancer Ther, 3:933


Burgeiro A, Bento AC, Gajate C, Oliveira PJ, Mollinedo F. (2013). Rapid human melanoma cell death induced by sanguinarine through oxidative stress. European Journal of Pharmacology, 705(1-3), 109-18. doi: 10.1016/j.ejphar.2013.02.035.


Choi WY, Kim GY, Lee WH, Choi YH. (2008). Sanguinarine, a benzophenanthridine alkaloid, induces apoptosis in MDA-MB-231 human breast carcinoma cells through a reactive oxygen species-mediated mitochondrial pathway. Chemotherapy, 54(4), 279-87. doi: 10.1159/000149719.


Han MH, Kim GY, Yoo YH, Choi YH. (2013). Sanguinarine induces apoptosis in human colorectal cancer HCT-116 cells through ROS-mediated Egr-1 activation and mitochondrial dysfunction. Toxicology Letters, 220(2), 157-66. doi: 10.1016/j.toxlet.2013.04.020.


Han MH, Park C, Jin CY, et al. (2013). Apoptosis induction of human bladder cancer cells by sanguinarine through reactive oxygen species-mediated up-regulation of early growth response gene-1. PLoS One, 8(5), e63425. doi: 10.1371/journal.pone.0063425.


Kaminskyy V, Lin KW, Filyak Y, Stoika R. (2008). Differential effect of sanguinarine, chelerythrine and chelidonine on DNA damage and cell viability in primary mouse spleen cells and mouse leukemic cells. Cell Biology International, 32(2), 271-277.


Kaminskyy V, Kulachkovskyy O, Stoika R. (2008) A decisive role of mitochondria in defining rate and intensity of apoptosis induction by different alkaloids. Toxicology Letters, 177(3), 168-81. doi: 10.1016/j.toxlet.2008.01.009.


Philchenkov A, Kaminskyy V, Zavelevich M, Stoika R. (2008). Apoptogenic activity of two benzophenanthridine alkaloids from Chelidonium majus L. does not correlate with their DNA-damaging effects. Toxicology In Vitro, 22(2), 287-95.

Saikosaponin

Cancers:
Cervical, colon, liver, lung, ovarian, liver, breast, hepatocellular

Action: Anti-angiogenic, anti-metastatic, chemo-sensitizer, pro-oxidative, cell-cycle arrest

T cell-mediated autoimmune, induces apoptosis, immune regulating, radio-sensitizer

Induces Apoptosis

Long dan xie gan tang, a well known Chinese herbal formulation, is commonly used by patients with chronic liver disease in China. Accumulated anecdotal evidence suggests that Long dan tang may have beneficial effects in patients with hepatocellular carcinoma. Long dan tang is comprised of five herbs: Gentiana root, Scutellaria root, Gardenia fruit, Alisma rhizome, and Bupleurum root. The cytotoxic effects of compounds from the five major ingredients isolated from the above plants, i.e. gentiopicroside, baicalein, geniposide, alisol B acetate and saikosaponin-d, respectively, on human hepatoma Hep3B cells, were investigated.

Annexin V immunofluorescence detection, DNA fragmentation assays and FACScan analysis of propidium iodide-staining cells showed that gentiopicroside, baicalein, and geniposide had little effect, whereas alisol B acetate and saikosaponin-d profoundly induced apoptosis in Hep3B cells. Alisol B acetate, but not saikosaponin-d, induced G2/M arrest of the cell-cycle as well as a significant increase in caspase-3 activity. Interestingly, baicalein by itself induced an increase in H(2)O(2) generation and the subsequent NF-kappaB activation; furthermore, it effectively inhibited the transforming growth factor-beta(1) (TGF-beta(1))-induced caspase-3 activation and cell apoptosis.

Results suggest that alisol B acetate and saikosaponin-d induced cell apoptosis through the caspase-3-dependent and -independent pathways, respectively. Instead of inducing apoptosis, baicalein inhibits TGF-beta(1)-induced apoptosis via increase in cellular H(2)O(2) formation and NF-kappaB activation in human hepatoma Hep3B cells (Chou, Pan, Teng & Guh, 2003).

Breast

Saikosaponin-A treatment of MDA-MB-231 for 3 hours and of MCF-7 cells for 2 hours, respectively, caused an obvious increase in the sub G1 population of cell-cycles.

Apoptosis in MDA-MB-231 cells was independent of the p53/p21 pathway mechanism and was accompanied by an increased ratio of Bax to Bcl-2 and c-myc levels and activation of caspase-3. In contrast, apoptosis of MCF-7 cells may have been initiated by the Bcl-2 family of proteins and involved p53/p21 dependent pathway mechanism, and was accompanied by an increased level of c-myc protein. The apoptosis of both MDA-MB-231 and MCF-7 cells showed a difference worthy of further research (Chen, Chang, Chung, & Chen, 2003).

Hepatocellular Carcinoma

The signaling pathway mediating induction of p15(INK4b) and p16(INK4a) during HepG2 growth inhibition triggered by the phorbol ester tumor promoter TPA (12-O-tetradecanoylphorbol 13-acetate) and the Chinese herbal compund Saikosaponin A was investigated.

Expressions of proto-oncogene c-jun, junB and c-fos were induced by TPA and Saikosaponin A between 30 minutes to 6 hours of treatment. Pre-treatment of 20 microg/ml PD98059, an inhibitor of MEK (the upstream kinase of ERK), prevents the TPA and Saikosaponin A triggered HepG2 growth inhibition by 50% and 30%, respectively. In addition, AP-1 DNA-binding assay, using non-isotopic capillary electrophoresis and laser-induced fluorescence (CE/LIF), demonstrated that the AP-1-related DNA-binding activity was significantly induced by TPA and Saikosaponin A, which can be reduced by PD98059 pre-treatment.

Results suggest that activation of ERK, together with its downstream transcriptional machinery, mediated p15(INK4b) and p16(INK4a) expression that led to HepG2 growth inhibition (Wen-Sheng, 2003).

The effects of Saikosaponin D (SSd) on syndecan-2, matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases-2 (TIMP-2) in livers of rats with hepatocellular carcinoma (HCC) was investigated.

The model group had more malignant nodules than the SSd group. Model-group HCC cells were grade III; SSd-group HCC cells were grades I-II. Controls showed normal hepatic cell phenotypes and no syndecan-2+ staining. Syndecan-2+ staining was greater in the model group (35.2%, P < or = 0.001) than in controls or the SSd group (16.5%, P < or = 0.001). The model group had more intense MMP-2+ staining than controls (0.37 vs 0.27, P< or =0.01) or the SSd group (0.31 vs 0.37, P< or =0.05); and higher MMP-13+ staining (72.55%) than in controls (12.55%, P< or =0.001) and SSd group (20.18%, P< or =0.01).

The model group also had more TIMP-2+ staining (57.2%) than controls (20.9%, P< or =0.001) and SSd group (22.7%, P< or=0.001). Controls and SSd group showed no difference in TIMP-2+ rates.

SSd inhibited HCC development, and downregulated expression of syndecan-2, MMP-2, MMP-13 and TIMP-2 in rat HCC liver tissue (Jia et al., 2012).

T Cell-mediated Autoimmune

Saikosaponin-d (Ssd) is a triterpene saponin derived from the medicinal plant, Bupleurum falcatum L. (Umbelliferae). Previous findings showed that Ssd exhibits a variety of pharmacological and immunomodulatory activities including anti-inflammatory, anti-bacterial, anti-viral and anti-cancer effects.

Results demonstrated that Ssd not only suppressed OKT3/CD28-costimulated human T cell proliferation, it also inhibited PMA, PMA/Ionomycin and Con A-induced mouse T cell activation in vitro. The inhibitory effect of Ssd on PMA-induced T cell activation was associated with down-regulation of NF-kappaB signaling through suppression of IKK and Akt activities. In addition, Ssd suppressed both DNA binding activity and the nuclear translocation of NF-AT and activator protein 1 (AP-1) of the PMA/Ionomycin-stimulated T cells. The cell surface markers, such as IL-2 receptor (CD25), were also down-regulated along with decreased production of pro-inflammatory cytokines of IL-6, TNF-alpha and IFN-gamma.

Results indicate that the NF-kappaB, NF-AT and AP-1 (c-Fos) signaling pathways are involved in the T cell inhibition evoked by Ssd. Ssd could be a potential candidate for further study in treating T cell-mediated autoimmune conditions (Wong, Zhou, Cheung, Li, & Liu, 2009).

Cervical Cancer

Saikosaponin-a and -d, two naturally occurring compounds derived from Bupleurum radix, have been shown to exert anti-cancer activity in several cancer cell lines. However, the effect of a combination of saikosaponins with chemotherapeutic drugs have never been addressed. Investigated as to whether these two saikosaponins have chemo-sensitization effect on cisplatin-induced cancer cell cytotoxicity was carried out.

Two cervical cancer cell lines, HeLa and Siha, an ovarian cancer cell line, SKOV3, and a non-small-cell lung cancer cell line, A549, were treated with saikosaponins or cisplatin individually or in combination. Cell death was quantitatively detected by the release of lactate dehydrogenase (LDH) using a cytotoxicity detection kit. Cellular ROS was analyzed by flow cytometry. Apoptosis was evaluated by AO/EB staining, flow cytometry after Anexin V and PI staining, and Western blot for caspase activation. ROS scavengers and caspase inhibitor were used to determine the roles of ROS and apoptosis in the effects of saikosaponins on cisplatin-induced cell death.

Both saikosaponin-a and -d sensitized cancer cells to cisplatin-induced cell death in a dose-dependent manner, which was accompanied with induction of reactive oxygen species (ROS) accumulation.

Results suggest that saikosaponins sensitize cancer cells to cisplatin through ROS-mediated apoptosis, and the combination of saikosaponins with cisplatin could be an effective therapeutic strategy (Wang et al., 2010).

Colon Cancer

Saikosaponin-a (SSa)-induced apoptosis of HCC cells was associated with proteolytic activation of caspase-9, caspase-3, and PARP cleavages and decreased levels of IAP family members, such as XIAP and c-IAP-2, but not of survivin. SSa treatment also enhanced the activities of caspase-2 and caspase-8, Bid cleavage, and the conformational activation of Bax. Moreover, inhibition of caspase-2 activation by the pharmacological inhibitor z-VDVAD-fmk, or by knockdown of protein levels using a si-RNA, suppressed SSa-induced caspase-8 activation, Bid cleavage, and the conformational activation of Bax. Although caspase-8 is an initiator caspase like caspase-2, the inhibition of caspase-8 activation by knockdown using a si-RNA did not suppress SSa-induced caspase-2 activation.

Results suggest that sequential activation of caspase-2 and caspase-8 is a critical step in SSa-induced apoptosis (Kim & Hong, 2011).

Immune Regulating

Tumor necrosis factor-alpha (TNF- α ) was reported as an anti-cancer therapy due to its cytotoxic effect against an array of tumor cells. However, its undesirable responses of TNF- α on activating NF- κB signaling and pro-metastatic property limit its clinical application in treating cancers. Therefore, sensitizing agents capable of overcoming this undesirable effect must be valuable for facilitating the usage of TNF- α -mediated apoptosis therapy for cancer patients. Previously, saikosaponin-d (Ssd), a triterpene saponin derived from the medicinal plant, Bupleurum falcatum L. (Umbelliferae), exhibited a variety of pharmacological activities such as anti-inflammatory, anti-bacterial, anti-viral and anti-cancer.

Investigation found that Ssd could potentially inhibit activated T lymphocytes via suppression of NF- κ B, NF-AT and AP-1 signaling. Ssd significantly potentiated TNF- α -mediated cell death in HeLa and HepG2 cancer cells via suppression of TNF- α -induced NF- κ B activation and its target genes expression involving cancer cell proliferation, invasion, angiogenesis and survival. Also, Ssd revealed a significant potency in abolishing TNF- α -induced cancer cell invasion and angiogenesis in HUVECs while inducing apoptosis via enhancing the loss of mitochondrial membrane potential in HeLa cells.

Collectively, findings indicate that Ssd has significant potential to be developed as a combined adjuvant remedy with TNF- α for cancer patients (Wong et al., 2013).

Radio-sensitizer

Saikosaponin-d (SSd), a monomer terpenoid purified from the Chinese herbal drug Radix bupleuri, has multiple effects, including anti-cancer properties. Treatment with SSd alone and radiation alone inhibited cell growth and increased apoptosis rate at the concentration used. These effects were enhanced when SSd was combined with radiation. Moreover, SSd potentiated the effects of radiation to induce G0/G1 arrest in SMMC-7721 hepatocellular carcinoma cells, and reduced the G2/M-phase population under hypoxia. SSd potentiates the effects of radiation on SMMC-7721 cells; thus, it is a promising radio-sensitizer. The radio-sensitizing effect of SSd may contribute to its effect on the G0/G1 and G2/M checkpoints of the cell-cycle (Wang et al., 2013).

References

Chen JC, Chang NW, Chung JG, Chen KC. (2003). Saikosaponin-A induces apoptotic mechanism in human breast MDA-MB-231 and MCF-7 cancer cells. The American Journal of Chinese Medicine, 31(3), 363-77.


Chou CC, Pan SL, Teng CM, Guh JH. (2003). Pharmacological evaluation of several major ingredients of Chinese herbal medicines in human hepatoma Hep3B cells. European Journal of Pharmaceutical Sciences, 19(5), 403-12.


Jia X, Dang S, Cheng Y, et al. (2012). Effects of saikosaponin-d on syndecan-2, matrix metalloproteinases and tissue inhibitor of metalloproteinases-2 in rats with hepatocellular carcinoma. Journal of Traditional Chinese Medicine, 32(3), 415-22.


Kim BM, Hong SH. (2011). Sequential caspase-2 and caspase-8 activation is essential for saikosaponin a-induced apoptosis of human colon carcinoma cell lines. Apoptosis, 16(2), 184-197. doi: 10.1007/s10495-010-0557-x.


Wang BF, Dai ZJ, Wang XJ, et al. (2013). Saikosaponin-d increases the radiosensitivity of smmc-7721 hepatocellular carcinoma cells by adjusting the g0/g1 and g2/m checkpoints of the cell-cycle. BMC Complementary and Alternative Medicine, 13:263. doi:10.1186/1472-6882-13-263


Wang Q, Zheng XL, Yang L, et al. (2010). Reactive oxygen species-mediated apoptosis contributes to chemo-sensitization effect of saikosaponins on cisplatin-induced cytotoxicity in cancer cells. Journal of Experimental & Clinical Cancer Research, 9(29), 159. doi: 10.1186/1756-9966-29-159.


Wen-Sheng, W. (2003). ERK signaling pathway is involved in p15INK4b/p16INK4a expression and HepG2 growth inhibition triggered by TPA and Saikosaponin A. Oncogene, 22(7), 955-963.


Wong VK, Zhang MM, Zhou H, et al. (2013). Saikosaponin-d Enhances the Anti-cancer Potency of TNF- α via Overcoming Its Undesirable Response of Activating NF-Kappa B Signaling in Cancer Cells. Evidence-based Complementary and Alternative Medicine, 2013(2013), 745295. doi: 10.1155/2013/745295.


Wong VK, Zhou H, Cheung SS, Li T, Liu L. (2009). Mechanistic study of saikosaponin-d (Ssd) on suppression of murine T lymphocyte activation. Journal of Cellular Biochemistry, 107(2), 303-15. doi: 10.1002/jcb.22126.

Rosmarinic Acid

Cancer: Leukemia

Action: Anti-oxidative, MDR

Leukemia

Because tumor necrosis factor-alpha (TNF-alpha) is well known to induce inflammatory responses, its clinical use is limited in cancer treatment. Rosmarinic acid (RA), a naturally occurring polyphenol flavonoid, has been reported to inhibit TNF-alpha-induced NF-kappaB activation in human dermal fibroblasts. Investigation found that RA treatment significantly sensitizes TNF-alpha-induced apoptosis in human leukemia U937 cells through the suppression of nuclear transcription factor-kappaB (NF-kappaB) and reactive oxygen species (ROS). This inhibition was correlated with suppression of NF-kappaB-dependent anti-apoptotic proteins (IAP-1, IAP-2, and XIAP). RA treatment also normalized TNF-alpha-induced ROS generation. Additionally, ectopic Bcl-2 expressing U937 reversed combined treatment-induced cell death, cytochrome c release into cytosol, and collapse of mitochondrial potential.

Results demonstrated that RA inhibits TNF-alpha-induced ROS generation and NF-kappaB activation, and enhances TNF-alpha-induced apoptosis (Moon, Kim, Lee, Choi, & Kim, 2010).

MDR

The intracellular accumulation of adriamycin, rhodamine123 (Rh123), and the expression of P-glycoprotein (P-gp) were assayed by flow cytometry. The influence of RA on the transcription of MDR1 gene was determined by reverse transcription-polymerase chain reaction. The results showed that RA could reverse the MDR of SGC7901/Adr cells, increase the intracellular accumulation of Adr and Rh123, and decrease the transcription of MDR1 gene and the expression of P-gp in SGC7901/Adr cells (Li et al., 2013).

Anti-cancer

Rosmarinic acid (RA), one of the major components of polyphenol, possesses attractive remedial features. Supplementation with RA significantly reduced the formation of aberrant crypt foci (ACF) and ACF multiplicity in 1,2-dimethylhydrazine (DMH) treated rats. Moreover RA supplementation prevented the alterations in circulatory anti-oxidant enzymes and colonic bacterial enzymes activities. Overall, results showed that all three doses of RA inhibited carcinogenesis, though the effect of the intermediary dose of 5 mg/kg b.w. was more pronounced (Karthikkumar et al., 2012).

References

Karthikkumar V, Sivagami G, Vinothkumar R, Rajkumar D, Nalini N. (2012). Modulatory efficacy of rosmarinic acid on premalignant lesions and anti-oxidant status in 1,2-dimethylhydrazine induced rat colon carcinogenesis. Environ Toxicol Pharmacol, 34(3):949-58. doi: 10.1016/j.etap.2012.07.014.


Li FR, Fu YY, Jiang DH, et al. (2013). Reversal effect of rosmarinic acid on Multi-drug resistance in SGC7901/Adr cell. J Asian Nat Prod Res, 15(3):276-85. doi: 10.1080/10286020.2012.762910.


Moon DO, Kim MO, Lee JD, Choi YH, Kim GY. (2010). Rosmarinic acid sensitizes cell death through suppression of TNF-alpha-induced NF-kappaB activation and ROS generation in human leukemia U937 cells. Cancer Letters, 288(2), 183-191. doi: 10.1016/j.canlet.2009.06.033.

Baicalin & Baicalein

Cancer:
Myeloma, liver, colorectal., breast, prostate, oral., hepatoma, ovarian

Action: Anti-cancer, cardiovascular disease, cytostatic, cardio-protective against Doxorubicin, anti-inflammatory, angiogenesis

Baicalin and baicalein are naturally occurring flavonoids that are found in the roots and leaves of some Chinese medicinal plants (including Scutellaria radix, Scutellaria rivularis (Benth.); Scutellaria baicalensis (Georgi) and Scutellaria lateriflora (L.)) are thought to have anti-oxidant activity and possible anti-angiogenic, anti-cancer, anxiolytic, anti-inflammatory and neuroprotective activities. In particular, Scutellaria baicalensis is one of the most popular and multi-purpose herbs used in China traditionally for treatment of inflammation, hypertension, cardiovascular diseases, and bacterial and viral infections (Ye et al., 2002; Zhang et al., 2011a).

Anti-cancer

Accumulating evidence demonstrates that Scutellaria also possesses potent anti-cancer activities. The bioactive components of Scutellaria have been confirmed to be flavones, wogonin, baicalein and baicalin. These phytochemicals are not only cytostatic but also cytotoxic to various human tumor cell lines in vitro and inhibit tumor growth in vivo. Most importantly, they show almost no or minor toxicity to normal epithelial and normal peripheral blood and myeloid cells. The anti-tumor functions of these flavones are largely due to their abilities to scavenge oxidative radicals, to attenuate NF-kappaB activity, to inhibit several genes important for regulation of the cell-cycle, to suppress COX-2 gene expression and to prevent viral infections (Li, 2008).

Multiple Myeloma

In the search for a more effective adjuvant therapy to treat multiple myeloma (MM), Ma et al. (2005) investigated the effects of the traditional Chinese herbal medicines Huang-Lian-Jie-Du-Tang (HLJDT), Gui-Zhi-Fu-Ling-Wan (GZFLW), and Huang-Lian-Tang (HLT) on the proliferation and apoptosis of myeloma cells. HLJDT inhibited the proliferation of myeloma cell lines and the survival of primary myeloma cells, especially MPC-1- immature myeloma cells, and induced apoptosis in myeloma cell lines via a mitochondria-mediated pathway by reducing mitochondrial membrane potential and activating caspase-9 and caspase-3.

Further experiments confirmed that Scutellaria radix was responsible for the suppressive effect of HLJDT on myeloma cell proliferation, and the baicalein in Scutellaria radix showed strong growth inhibition and induction of apoptosis in comparison with baicalin or wogonin. Baicalein as well as baicalin suppressed the survival in vitro of MPC-1- immature myeloma cells rather than MPC-1+ myeloma cells from myeloma patients.

Baicalein inhibited the phosphorylation of IkB-alpha, which was followed by decreased expression of the IL-6 and XIAP genes and activation of caspase-9 and caspase-3. Therefore, HLJDT and Scutellaria radix have an anti-proliferative effect on myeloma cells, especially MPC-1- immature myeloma cells, and baicalein may be responsible for the suppressive effect of Scutellaria radix by blocking IkB-alpha degradation (Ma, 2005).

Hepatoma

The effects of the flavonoids from Scutellaria baicalensis Georgi (baicalein, baicalin and wogonin) in cultured human hepatoma cells (Hep G2, Hep 3B and SK-Hep1) were compared by MTT assay and flow cytometry. All three flavonoids dose-dependently decreased the cell viabilities accompanying the collapse of mitochondrial membrane potential and the depletion of glutathione content. However, the influence of baicalein, baicalin or wogonin on cell-cycle progression was different.

All three flavonoids resulted in prominent increase of G2/M population in Hep G2 cells, whereas an accumulation of sub G1 (hypoploid) peak in Hep 3B cells was observed. In SK-Hep1 cells, baicalein and baicalin resulted in a dramatic boost in hypoploid peak, but wogonin mainly in G1 phase accumulation. These data, together with the previous findings in other hepatoma cell lines, suggest that baicalein, baicalin and wogonin might be effective candidates for inducing apoptosis or inhibiting proliferation in various human hepatoma cell lines (Chang, 2002).

Long dan xie gan tang (pinyin) is one of the most commonly used herbal formulas by patients with chronic liver disease in China. Accumulated anecdotal evidence suggests that Long dan tang may have beneficial effects in patients with hepatocellular carcinoma. Long dan tang is comprised of five herbs: Gentiana root, Scutellaria root, Gardenia fruit, Alisma rhizome, and Bupleurum root. The cytotoxic effects of compounds from the five major ingredients isolated from the above plants, i.e. gentiopicroside, baicalein, geniposide, alisol B acetate and saikosaponin-d, were investigated, respectively, on human hepatoma Hep3B cells..

Interestingly, baicalein by itself induced an increase in H(2)O(2) generation and the subsequent NF-kappaB activation; furthermore, it effectively inhibited the transforming growth factor-beta(1) (TGF-beta(1))-induced caspase-3 activation and cell apoptosis. Results suggest that alisol B acetate and saikosaponin-d induced cell apoptosis through the caspase-3-dependent and -independent pathways, respectively. Instead of inducing apoptosis, baicalein inhibits TGF-beta(1)-induced apoptosis via increase in cellular H(2)O(2) formation and NF-kappaB activation in human hepatoma Hep3B cells (Chou, Pan, Teng & Guh, 2003).

Ovarian Cancer

Ovarian cancer is one of the primary causes of death for women all through the Western world. Two kinds of ovarian cancer (OVCAR-3 and CP-70) cell lines and a normal ovarian cell line (IOSE-364) were selected to be investigated in the inhibitory effect of baicalin and baicalein on cancer cells. Largely, baicalin and baicalein inhibited ovarian cancer cell viability in both ovarian cancer cell lines with LD50 values in the range of 45-55 µM for baicalin and 25-40 µM for baicalein. On the other hand, both compounds had fewer inhibitory effects on normal ovarian cells viability with LD50 values of 177 µM for baicalin and 68 µM for baicalein.

Baicalin decreased expression of VEGF (20 µM), cMyc (80 µM), and NFkB (20 µM); baicalein decreased expression of VEGF (10 µM), HIF-1α (20 µM), cMyc (20 µM), and NFkB (40 µM). Therefore baicalein is more effective in inhibiting cancer cell viability and expression of VEGF, HIF-1α, cMyc, and NFκB in both ovarian cancer cell lines. It seems that baicalein inhibited cancer cell viability through the inhibition of cancer promoting genes expression including VEGF, HIF-1α, cMyc, and NFκB.

Overall, this study showed that baicalein and baicalin significantly inhibited the viability of ovarian cancer cells, while generally exerting less of an effect on normal cells. They have potential for chemoprevention and treatment of ovarian cancers (Chen, 2013).

Breast Cancer

Baicalin was found to be a potent inhibitor of mammary cell line MCF-7 and ductal breast epithelial tumor cell line T-47D proliferation, as well as having anti-proliferative effects on other cancer types such as the human head and neck cancer epithelial cell lines CAL-27 and FaDu. Overall, baicalin inhibited the proliferation of human breast cancer cells and CAL-27 and FaDu cells with effective potency (Franek, 2005).

Breast Cancer, Cell Invasion

The effect of Baicalein on cell viability of the human breast cancer MDA-MB-231 cell line was tested by MTT. 50, 100 µmol·L-1 of Baicalein inhibited significantly cell invasion(P0.01) and migration(P0.01) compared with control groups. The inhibitory rates were 50% and 77% in cell migration and 15% and 44% in cell invasion, respectively. 50 µmol·L-1 of Baicalein significantly inhibited the level of MMP 2 expression. 100 µmol·L-1 of Baicalein significantly inhibited the level of MMP 9 and uPA expressions.

Baicalein inhibits invasion and migration of MDA-MB-231 cells. The mechanisms may be involved in the direct inhibition of cell invasion and migration abilities, and the inhibition of MMP 2, MMP 9, and uPA expressions (Wang et al., 2010).

The proliferation of MDA-MB-231 cell line human breast adenocarcinoma was inhibited by baicalin in a dose-and time-dependent manner and the IC50 was 151 µmol/L. The apoptotic rate of the baicalin-treated MDA-MB-231 cells increased significantly at 48 hours. Flow cytometer analysis also revealed that most of the baicalin-treated MDA-MB-231 cells were arrested in the G2/M phase. Typically apoptotic characteristics such as condensed chromatin and apoptotic bodies were observed after being treated with baicalin for 48 hours.

The results of RT-PCR showed that the expression of bax was up-regulated; meanwhile, the expression of bcl-2 was down-regulated. Baicalin could inhibit the proliferation of MDA-MB-231 cells through apoptosis by regulating the expression of bcl-2, bax and intervening in the process of the cell-cycle (Zhu et al., 2008).

Oral Cancer

As an aryl hydrocarbon receptor (AhR) ligand, baicalein at high concentrations blocks AhR-mediated dioxin toxicity. Because AhR had been reported to play a role in regulating the cell-cycle, it is suspected that the anti-cancer effect of baicalein is associated with AhR. The molecular mechanism involved in the anti-cancer effect of baicalein in oral cancer cells HSC-3 has been investigated, including whether such an effect would be AhR-mediated. Results revealed that baicalein inhibited cell proliferation and increased AhR activity in a dose-dependent manner. Cell-cycle was arrested at the G1 phase and the expression of CDK4, cyclin D1, and phosphorylated retinoblastoma (pRb) was decreased.

When cells were pre-treated with LiCl, the inhibitor of GSK-3β, the decrease of cyclin D1 was blocked and the reduction of pRb was recovered. The data indicates that in HSC-3 the reduction of pRb is mediated by baicalein both through activation of AhR and facilitation of cyclin D1 degradation, which causes cell-cycle arrest at the G1 phase, and results in the inhibition of cell proliferation (Cheng, 2012).

Anti-inflammatory

Baicalin has also been examined for its effects on LPS-induced nitric oxide (NO) production and iNOS and COX-2 gene expressions in RAW 264.7 macrophages. The results indicated that baicalin inhibited LPS-induced NO production in a concentration-dependent manner without a notable cytotoxic effect on these cells. The decrease in NO production was consistent with the inhibition by baicalin of LPS-induced iNOS gene expression (Chen, 2001)

Angiogenesis Modulation

The modulation of angiogenesis is one possible mechanism by which baicalin may act in the treatment of cardiovascular diseases. This may be elucidated by investigating the effects of baicalin on the expression of vascular endothelial growth factor (VEGF), a critical factor for angiogenesis. The effects of baicalin and an extract of S. baicalensis on VEGF expression were tested in several cell lines. Both agents induced VEGF expression in all cells without increasing expression of hypoxia-inducible factor-1alpha (HIF-1alpha).

Their ability to induce VEGF expression was suppressed once ERRalpha expression was knocked down by siRNA, or ERRalpha-binding sites were deleted in the VEGF promoter. It was also found that both agents stimulated cell migration and vessel sprout formation from the aorta. These results therefore implicate baicalin and S. baicalensis in angiogenesis by inducing VEGF expression through the activation of the ERRalpha pathway (Zhang, 2011b).

Colon Cancer

The compounds of baicalein and wogonin, derived from the Chinese herb Scutellaria baicalensis, were studied for their effect in suppressing the viability of HT-29 human colon cancer cells. Following treatment with baicalein or wogonin, several apoptotic events were observed, including DNA fragmentation, chromatin condensation and increased cell-cycle arrest at the G1 phase. Baicalein and wogonin decreased Bcl-2 expression, whereas the expression of Bax was increased in a dose-dependent manner when compared to the control.

The results indicated that baicalein induced apoptosis via Akt activation, in a p53-dependent manner, in HT-29 colon cancer cells. Baicalein may serve as a chemo-preventive, or therapeutic, agent for HT-29 colon cancer (Kim et al., 2012).

Cardio-protective

The cardiotoxicity of doxorubicin limits its clinical use in the treatment of a variety of malignancies. Previous studies suggest that doxorubicin-associated cardiotoxicity is mediated by reactive oxygen species (ROS)-induced apoptosis. Baicalein attenuated phosphorylation of JNK induced by doxorubicin. Co-treatment of cardiomyocytes with doxorubicin and JNK inhibitor SP600125 (10 µM; 24 hours) reduced JNK phosphorylation and enhanced cell survival., suggesting that the baicalein protection against doxorubicin cardiotoxicity was mediated by JNK activation. Baicalein adjunct treatment confers anti-apoptotic protection against doxorubicin-induced cardiotoxicity without compromising its anti-cancer efficacy (Chang et al., 2011).

Prostate Cancer

There are four compounds capable of inhibiting prostate cancer cell proliferation in Scutellaria baicalensis: baicalein, wogonin, neobaicalein, and skullcapflavone. Comparisons of the cellular effects induced by the entire extract versus the four-compound combination produced comparable cell-cycle changes, levels of growth inhibition, and global gene expression profiles (r(2) = 0.79). Individual compounds exhibited anti-androgenic activities with reduced expression of the androgen receptor and androgen-regulated genes. In vivo, baicalein (20 mg/kg/d p.o.) reduced the growth of prostate cancer xenografts in nude mice by 55% at 2 weeks compared with placebo and delayed the average time for tumors to achieve a volume of approximately 1,000 mm(3) from 16 to 47 days (P < 0.001).

Most of the anti-cancer activities of S. baicalensis can be recapitulated with four purified constituents that function in part through inhibition of the androgen receptor signaling pathway (Bonham et al., 2005)

Cancer: Acute lymphocytic leukemia, lymphoma and myeloma

Action: Cell-cycle arrest, induces apoptosis

Scutellaria baicalensis (S.B.) is a widely used Chinese herbal medicine. S.B inhibited the growth of acute lymphocytic leukemia (ALL), lymphoma and myeloma cell lines by inducing apoptosis and cell cycle arrest at clinically achievable concentrations. The anti-proliferative effectwas associated with mitochondrial damage, modulation of the Bcl family of genes, increased level of the CDK inhibitor p27KIP1 and decreased level of c-myc oncogene. HPLC analysis of S.B. showed it contains 21% baicalin and further studies confirmed it was the major anti-cancer component of S.B. Thus, Scutellaria baicalensis should be tested in clinical trials for these hematopoietic malignancies (Kumagai et al., 2007).

References

Bonham M, Posakony J, Coleman I, Montgomery B, Simon J, Nelson PS. (2005). Characterization of chemical constituents in Scutellaria baicalensis with antiandrogenic and growth-inhibitory activities toward prostate carcinoma. Clin Cancer Res, 11(10):3905-14.


Chang WH Chen CH Lu FJ. (2002). Different Effects of Baicalein, Baicalin and Wogonin on Mitochondrial Function, Glutathione Content and cell-cycle Progression in Human Hepatoma Cell Lines. Planta Med, 68(2):128-32. doi: 10.1055/s-2002-20246


Chang WT, Li J, Huang HH, et al. (2011). Baicalein protects against doxorubicin-induced cardiotoxicity by attenuation of mitochondrial oxidant injury .and JNK activation. J Cell Biochem. doi: 10.1002/jcb.23201.


Chen J, Li Z, Chen AY, Ye X, et al. (2013). Inhibitory effect of baicalin and baicalein on ovarian cancer cells. Int J Mol Sci, 14(3):6012-25. doi: 10.3390/ijms14036012.


Chen YC, Shen SC, Chen LG, Lee TJ, Yang LL. (2001). Wogonin, baicalin, and baicalein inhibition of inducible nitric oxide synthase and cyclooxygenase-2 gene expressions induced by nitric oxide synthase inhibitors and lipopolysaccharide. Biochem Pharmacol,61(11):1417-27. doi:10.1016/S0006-2952(01)00594-9


Cheng YH, Li LA, Lin P, et al. (2012). Baicalein induces G1 arrest in oral cancer cells by enhancing the degradation of cyclin D1 and activating AhR to decrease Rb phosphorylation. Toxicol Appl Pharmacol, 263(3):360-7. doi: 10.1016/j.taap.2012.07.010.


Chou CC, Pan SL, Teng CM, & Guh JH. (2003). Pharmacological evaluation of several major ingredients of Chinese herbal medicines in human hepatoma Hep3B cells. European Journal of Pharmaceutical Sciences, 19(5), 403-12.


Franek KJ, Zhou Z, Zhang WD, Chen WY. (2005). In vitro studies of baicalin alone or in combination with Salvia miltiorrhiza extract as a potential anti-cancer agent. Int J Oncol, 26(1):217-24.


Kim SJ, Kim HJ, Kim HR, et al. (2012). Anti-tumor actions of baicalein and wogonin in HT-29 human colorectal cancer cells. Molecular Medicine Reports, 6(6):1443-1449. doi: 10.3892/mmr.2012.1085.


Li-Weber M. (2009). New therapeutic aspects of flavones: The anti-cancer properties of Scutellaria and its main active constituents Wogonin, Baicalein and Baicalin. Cancer Treat Rev, 35(1):57-68. doi: 10.1016/j.ctrv.2008.09.005.


Ma Z, Otsuyama K, Liu S, et al. (2005). Baicalein, a component of Scutellaria radix from Huang-Lian-Jie-Du-Tang (HLJDT), leads to suppression of proliferation and induction of apoptosis in human myeloma cells. Blood, 105(8):3312-8. doi:10.1182/blood-2004-10-3915.


Wang Xf, Zhou Qm, Su Sb. (2010). Experimental study on Baicalein inhibiting the invasion and migration of human breast cancer cells. Zhong Guo Yao Li Xue Tong Bao, 26(6): 745-750.


Zhang XW, Li WF, Li WW, et al. (2011a). Protective effects of the aqueous extract of Scutellaria baicalensis against acrolein-induced oxidative stress in cultured human umbilical vein endothelial cells. Pharm Biol, 49(3): 256–261. doi:10.3109/13880209.2010.501803.


Ye F, Xui L, Yi J, Zhang, W, Zhang DY. (2002). Anti-cancer activity of Scutellaria baicalensis and its potential mechanism. J Altern Complement Med, 8(5):567-72.


Zhang K, Lu J, Mori T, et al. (2011b). Baicalin increases VEGF expression and angiogenesis by activating the ERR{alpha}/PGC-1{alpha} pathway.[J]. Cardiovascular Research, 89(2):426-435.


Zhu Gq, Tang Lj, Wang L, Su Jj, et al. (2008). Study on Baicalin Induced Apoptosis of Human Breast Cancer Cell Line MDA-MB-231. An Hui Zhong Yi Xue Yuan Xue Bao, 27(2):20-23

Kumagai T, et al. (2007) Scutellaria baicalensis, a herbal medicine: Anti-proliferative and apoptotic activity against acute lymphocytic leukemia, lymphoma and myeloma cell lines. Leukemia Research 31 (2007) 523-530

Ginsenoside (See also Rg3)

Cancer:
Breast, colorectal., brain, leukemia, acute myeloid leukemia (AML), melanoma, lung, glioblastoma, prostate, fibroblast carcinoma

Action: Multi-drug resistance, apoptosis, anti-cancer, chemotherapy sensitizer, CYP450 regulating, inhibits growth and metastasis, down-regulates MMP-9, enhances 5-FU, anti-inflammatory

Inhibits Growth and Metastasis

Ginsenosides, belonging to a group of saponins with triterpenoid dammarane skeleton, show a variety of pharmacological effects. Among them, some ginsenoside derivatives, which can be produced by acidic and alkaline hydrolysis, biotransformation and steamed process from the major ginsenosides in ginseng plant, perform stronger activities than the major primeval ginsenosides on inhibiting growth or metastasis of tumor, inducing apoptosis and differentiation of tumor and reversing multi-drug resistance of tumor. Therefore ginsenoside derivatives are promising as anti-tumor active compounds and drugs (Cao et al., 2012).

Ginsenoside content can vary widely depending on species, location of growth, and growing time before harvest. The root, the organ most often used, contains saponin complexes. These are often split into two groups: the Rb1 group (characterized by the protopanaxadiol presence: Rb1, Rb2, Rc and Rd) and the Rg1 group (protopanaxatriol: Rg1, Re, Rf, and Rg2). The potential health effects of ginsenosides include anti-carcinogenic, immunomodulatory, anti-inflammatory, anti-allergic, anti-atherosclerotic, anti-hypertensive, and anti-diabetic effects as well as anti-stress activity and effects on the central nervous system (Christensen, 2009).

Ginsenosides are considered the major pharmacologically active constituents, and approximately 12 types of ginsenosides have been isolated and structurally identified. Ginsenoside Rg3 was metabolized to ginsenoside Rh2 and protopanaxadiol by human fecal microflora (Bae et al., 2002). Ginsenoside Rg3 and the resulting metabolites exhibited potent cytotoxicity against tumor cell lines (Bae et al., 2002).

Screen-Shot-2014-03-28-at-11.53.41-am1

Ginseng Extracts (GE); Methanol-(alc-GE) or Water-extracted (w-GE) and ER+ Breast Cancer

Ginseng root extracts and the biologically active ginsenosides have been shown to inhibit proliferation of human cancer cell lines, including breast cancer. However, there are conflicting data that suggest that ginseng extracts (GEs) may or may not have estrogenic action, which might be contraindicated in individuals with estrogen-dependent cancers. The current study was designed to address the hypothesis that the extraction method of American ginseng (Panax quinquefolium) root will dictate its ability to produce an estrogenic response using the estrogen receptor (ER)-positive MCF-7 human breast cancer cell model. MCF-7 cells were treated with a wide concentration range of either methanol-(alc-GE) or water-extracted (w-GE) ginseng root for 6 days.

An increase in MCF-7 cell proliferation by GE indicated potential estrogenicity. This was confirmed by blocking GE-induced MCF-7 cell proliferation with ER antagonists ICI 182,780 (1 nM) and 4-hydroxytamoxifen (0.1 microM). Furthermore, the ability of GE to bind ERalpha or ERbeta and stimulate estrogen-responsive genes was examined. Alc-GE, but not w-GE, was able to increase MCF-7 cell proliferation at low concentrations (5-100 microg/mL) when cells were maintained under low-estrogen conditions. The stimulatory effect of alc-GE on MCF-7 cell proliferation was blocked by the ER antagonists ICI 182,780 or 4-hydroxyta-moxifen. At higher concentrations of GE, both extracts inhibited MCF-7 and ER-negative MDA-MB-231 cell proliferation regardless of media conditions.

These data indicate that low concentrations of alc-GE, but not w-GE, elicit estrogenic effects, as evidenced by increased MCF-7 cell proliferation, in a manner antagonized by ER antagonists, interactions of alc-GE with estrogen receptors, and increased expression of estrogen-responsive genes by alc-GE. Thus, discrepant results between different laboratories may be due to the type of GE being analyzed for estrogenic activity (King et al., 2006).

Anti-cancer

Previous studies suggested that American ginseng and notoginseng possess anti-cancer activities. Using a special heat-preparation or steaming process, the content of Rg3, a previously identified anti-cancer ginsenoside, increased significantly and became the main constituent in the steamed American ginseng. As expected, using the steamed extract, anti-cancer activity increased significantly. Notoginseng has a very distinct saponin profile compared to that of American ginseng. Steaming treatment of notoginseng also significantly increased anti-cancer effect (Wang et al., 2008).

Steam Extraction; Colorectal Cancer

After steaming treatment of American ginseng berries (100-120 ¡C for 1 h, and 120 ¡C for 0.5-4 h), the content of seven ginsenosides, Rg1, Re, Rb1, Rc, Rb2, Rb3, and Rd, decreased; the content of five ginsenosides, Rh1, Rg2, 20R-Rg2, Rg3, and Rh2, increased. Rg3, a previously identified anti-cancer ginsenoside, increased significantly. Two h of steaming at 120 ¡C increased the content of ginsenoside Rg3 to a greater degree than other tested ginsenosides. When human colorectal cancer cells were treated with 0.5 mg/mL steamed berry extract (120 ¡C 2 hours), the anti-proliferation effects were 97.8% for HCT-116 and 99.6% for SW-480 cells.

After staining with Hoechst 33258, apoptotic cells increased significantly by treatment with steamed berry extract compared with unheated extracts. The steaming of American ginseng berries hence augments ginsenoside Rg3 content and increases the anti-proliferative effects on two human colorectal cancer cell lines (Wang et al., 2006).

Glioblastoma

The major active components in red ginseng consist of a variety of ginsenosides including Rg3, Rg5 and Rk1, each of which has different pharmacological activities. Among these, Rg3 has been reported to exert anti-cancer activities through inhibition of angiogenesis and cell proliferation.

It is essential to develop a greater understanding of this novel compound by investigating the effects of Rg3 on a human glioblastoma cell line and its molecular signaling mechanism. The mechanisms of apoptosis by ginsenoside Rg3 were related with the MEK signaling pathway and reactive oxygen species. These data suggest that ginsenoside Rg3 is a novel agent for the chemotherapy of GBM (Choi et al., 2013).

Colon Cancer; Chemotherapy

Rg3 can inhibit the activity of NF-kappaB, a key transcriptional factor constitutively activated in colon cancer that confers cancer cell resistance to chemotherapeutic agents. Compared to treatment with Rg3 or chemotherapy alone, combined treatment was more effective (i.e., there were synergistic effects) in the inhibition of cancer cell growth and induction of apoptosis and these effects were accompanied by significant inhibition of NF-kappaB activity.

NF-kappaB target gene expression of apoptotic cell death proteins (Bax, caspase-3, caspase-9) was significantly enhanced, but the expression of anti-apoptotic genes and cell proliferation marker genes (Bcl-2, inhibitor of apoptosis protein (IAP-1) and X chromosome IAP (XIAP), Cox-2, c-Fos, c-Jun and cyclin D1) was significantly inhibited by the combined treatment compared to Rg3 or docetaxel alone.

These results indicate that ginsenoside Rg3 inhibits NF-kappaB, and enhances the susceptibility of colon cancer cells to docetaxel and other chemotherapeutics. Thus, ginsenoside Rg3 could be useful as an anti-cancer or adjuvant anti-cancer agent (Kim et al., 2009).

Prostate Cancer; Chemo-sensitizer

Nuclear factor-kappa (NF-kappaB) is also constitutively activated in prostate cancer, and gives cancer cells resistance to chemotherapeutic agents. Rg3 has hence also been found to increase susceptibility of prostate (LNCaP and PC-3, DU145) cells against chemotherapeutics; prostate cancer cell growth as well as activation of NF-kappaB was examined. It has been found that a combination treatment of Rg3 (50 microM) with a conventional agent docetaxel (5 nM) was more effective in the inhibition of prostate cancer cell growth and induction of apoptosis as well as G(0)/G(1) arrest accompanied with the significant inhibition of NF-kappaB activity, than those by treatment of Rg3 or docetaxel alone.

The combination of Rg3 (50 microM) with cisplatin (10 microM) and doxorubicin (2 microM) was also more effective in the inhibition of prostate cancer cell growth and NF-kappaB activity than those by the treatment of Rg3 or chemotherapeutics alone. These results indicate that ginsenoside Rg3 inhibits NF-kappaB, and enhances the susceptibility of prostate cancer cells to docetaxel and other chemotherapeutics. Thus, ginsenoside Rg3 could be useful as an anti-cancer agent (Kim et al., 2010).

Colon Cancer

Ginsenosides may not only be useful in themselves, but also for their downstream metabolites. Compound K (20-O-( β -D-glucopyranosyl)-20(S)-protopanaxadiol) is an active metabolite of ginsenosides and induces apoptosis in various types of cancer cells. This study investigated the role of autophagy in compound K-induced cell death of human HCT-116 colon cancer cells. Compound K activated an autophagy pathway characterized by the accumulation of vesicles, the increased positive acridine orange-stained cells, the accumulation of LC3-II, and the elevation of autophagic flux.

Compound K-provoked autophagy was also linked to the generation of intracellular reactive oxygen species (ROS); both of these processes were mitigated by the pre-treatment of cells with the anti-oxidant N-acetylcysteine.   Moreover, compound K activated the c-Jun NH2-terminal kinase (JNK) signaling pathway, whereas down-regulation of JNK by its specific inhibitor SP600125 or by small interfering RNA against JNK attenuated autophagy-mediated cell death in response to compound K.

Notably, compound K-stimulated autophagy as well as apoptosis was induced by disrupting the interaction between Atg6 and Bcl-2. Taken together, these results indicate that the induction of autophagy and apoptosis by compound K is mediated through ROS generation and JNK activation in human colon cancer cells (Kim et al., 2013b).

Lung Cancer; SCC

Korea white ginseng (KWG) has been investigated for its chemo-preventive activity in a mouse lung SCC model. N-nitroso-trischloroethylurea (NTCU) was used to induce lung tumors in female Swiss mice, and KWG was given orally. KWG significantly reduced the percentage of lung SCCs from 26.5% in the control group to 9.1% in the KWG group and in the meantime, increased the percentage of normal bronchial and hyperplasia. KWG was also found to greatly reduce squamous cell lung tumor area from an average of 9.4% in control group to 1.5% in the KWG group.

High-performance liquid chromatography/mass spectrometry identified 10 ginsenosides from KWG extracts, Rb1 and Rd being the most abundant as detected in mouse blood and lung tissue. These results suggest that KWG could be a potential chemo-preventive agent for lung SCC (Pan et al., 2013).

Leukemia

Rg1 was found to significantly inhibit the proliferation of K562 cells in vitro and arrest the cells in G2/M phase. The percentage of positive cells stained by SA-beta-Gal was dramatically increased (P < 0.05) and the expression of cell senescence-related genes was up-regulated. The observation of ultrastructure showed cell volume increase, heterochromatin condensation and fragmentation, mitochondrial volume increase, and lysosomes increase in size and number. Rg1 can hence induce the senescence of leukemia cell line K562 and play an important role in regulating p53-p21-Rb, p16-Rb cell signaling pathway (Cai et al., 2012).

Leukemia, Lymphoma

It has been found that Rh2 inhibits the proliferation of human leukemia cells concentration- and time-dependently with an IC(50) of ~38 µM. Rh2 blocked cell-cycle progression at the G(1) phase in HL-60 leukemia and U937 lymphoma cells, and this was found to be accompanied by the down-regulations of cyclin-dependent kinase (CDK) 4, CDK6, cyclin D1, cyclin D2, cyclin D3 and cyclin E at the protein level. Treatment of HL-60 cells with Rh2 significantly increased transforming growth factor- β (TGF- β ) production, and co-treatment with TGF- β neutralizing antibody prevented the Rh2-induced down-regulations of CDK4 and CDK6, up-regulations of p21(CIP1/WAF1) and p27(KIP1) levels and the induction of differentiation. These results demonstrate that the Rh2-mediated G(1) arrest and the differentiation are closely linked to the regulation of TGF- β production in human leukemia cells (Chung et al., 2012).

NSCLC

Ginsenoside Rh2, one of the components in ginseng saponin, has been shown to have anti-proliferative effect on human NSCLC cells and is being studied as a therapeutic drug for NSCLC. MicroRNAs (miRNAs) are small, non-coding RNA molecules that play a key role in cancer progression and prevention.

A unique set of changes in the miRNA expression profile in response to Rh2 treatment in the human NSCLC cell line A549 has been identified using miRNA microarray analysis. These miRNAs are predicted to have several target genes related to angiogenesis, apoptosis, chromatic modification, cell proliferation and differentiation. Thus, these results may assist in the better understanding of the anti-cancer mechanism of Rh2 in NSCLC (An et al., 2012).

Ginsenoside Concentrations

Ginsenosides, the major chemical composition of Chinese white ginseng (Panax ginseng C. A. Meyer), can inhibit tumor, enhance body immune function, prevent neurodegeneration. The amount of ginsenosides in the equivalent extraction of the nanoscale Chinese white ginseng particles (NWGP) was 2.5 times more than that of microscale Chinese white ginseng particles (WGP), and the extractions from NWGP (1000 microg/ml) reached a high tumor inhibition of 64% exposed to human lung carcinoma cells (A549) and 74% exposed to human cervical cancer cells (Hela) after 72 hours. Thia work shows that the nanoscale Chinese WGP greatly improves the bioavailability of ginsenosides (Ji et al., 2012).

Chemotherapy Side-effects

Pre-treatment with American ginseng berry extract (AGBE), a herb with potent anti-oxidant capacity, and one of its active anti-oxidant constituents, ginsenoside Re, was examined for its ability to counter cisplatin-induced emesis using a rat pica model. In rats, exposure to emetic stimuli such as cisplatin causes significant kaolin (clay) intake, a phenomenon called pica. We therefore measured cisplatin-induced kaolin intake as an indicator of the emetic response.

Rats were pre-treated with vehicle, AGBE (dose range 50–150 mg/kg, IP) or ginsenoside Re (2 and 5 mg/kg, IP). Rats were treated with cisplatin (3 mg/kg, IP) 30 min later. Kaolin intake, food intake, and body weight were measured every 24 hours, for 120 hours.

A significant dose-response relationship was observed between increasing doses of pre-treatment with AGBE and reduction in cisplatin-induced pica. Kaolin intake was maximally attenuated by AGBE at a dose of 100 mg/kg. Food intake also improved significantly at this dose (P<0.05). pre-treatment ginsenoside (5 mg/kg) also decreased kaolin intake >P<0.05). In vitro studies demonstrated a concentration-response relationship between AGBE and its ability to scavenge superoxide and hydroxyl.

Pre-treatment with AGBE and its major constituent, Re, hence attenuated cisplatin-induced pica, and demonstrated potential for the treatment of chemotherapy-induced nausea and vomiting. Significant recovery of food intake further strengthens the conclusion that AGBE may exert an anti-nausea/anti-emetic effect (Mehendale et al., 2005).

MDR

Because ginsenosides are structurally similar to cholesterol, the effect of Rp1, a novel ginsenoside derivative, on drug resistance using drug-sensitive OVCAR-8 and drug-resistant NCI/ADR-RES and DXR cells. Rp1 treatment resulted in an accumulation of doxorubicin or rhodamine 123 by decreasing MDR-1 activity in doxorubicin-resistant cells. Rp1 synergistically induced cell death with actinomycin D in DXR cells. Rp1 appeared to redistribute lipid rafts and MDR-1 protein.

Rp1 reversed resistance to actinomycin D by decreasing MDR-1 protein levels and Src phosphorylation with modulation of lipid rafts. Addition of cholesterol attenuated Rp1-induced raft aggregation and MDR-1 redistribution. Rp1 and actinomycin D reduced Src activity, and overexpression of active Src decreased the synergistic effect of Rp1 with actinomycin D. Rp1-induced drug sensitization was also observed with several anti-cancer drugs, including doxorubicin. These data suggest that lipid raft-modulating agents can be used to inhibit MDR-1 activity and thus overcome drug resistance (Yun et al., 2013).

Hypersensitized MDR Breast Cancer Cells to Paclitaxel

The effects of Rh2 on various tumor-cell lines for its effects on cell proliferation, induction of apoptosis, and potential interaction with conventional chemotherapy agents were investigated. Jia et al., (2004) showed that Rh2 inhibited cell growth by G1 arrest at low concentrations and induced apoptosis at high concentrations in a variety of tumor-cell lines, possibly through activation of caspases. The apoptosis induced by Rh2 was mediated through glucocorticoid receptors. Most interestingly, Rh2 can act either additively or synergistically with chemotherapy drugs on cancer cells. Particularly, it hypersensitized multi-drug-resistant breast cancer cells to paclitaxel.

These results suggest that Rh2 possesses strong tumor-inhibiting properties, and potentially can be used in treatments for multi-drug-resistant cancers, especially when it is used in combination with conventional chemotherapy agents.

MDR; Leukemia, Fibroblast Carcinoma

It was previously reported that a red ginseng saponin, 20(S)-ginsenoside Rg3 could modulate MDR in vitro and extend the survival of mice implanted with ADR-resistant murine leukemia P388 cells. A cytotoxicity study revealed that 120 microM of Rg3 was cytotoxic against a multi-drug-resistant human fibroblast carcinoma cell line, KB V20C, but not against normal WI 38 cells in vitro. 20 microM Rg3 induced a significant increase in fluorescence anisotropy in KB V20C cells but not in the parental KB cells. These results clearly show that Rg3 decreases the membrane fluidity thereby blocking drug efflux (Kwon et al., 2008).

MDR

Ginsenoside Rb1 is a representative component of panaxadiol saponins, which belongs to dammarane-type tritepenoid saponins and mainly exists in family araliaceae. It has been reported that ginsenoside Rb1 has diverse biological activities. The research development in recent decades on its pharmacological effects of cardiovascular system, anti-senility, reversing multi-drug resistance of tumor cells, adjuvant anti-cancer chemotherapy, and promoting peripheral nerve regeneration have been established (Jia et al., 2008).

Enhances Cyclophosphamide

Cyclophosphamide, an alkylating agent, has been shown to possess various genotoxic and carcinogenic effects, however, it is still used extensively as an anti-tumor agent and immunosuppressant in the clinic. Previous reports reveal that cyclophosphamide is involved in some secondary neoplasms.

C57BL/6 mice bearing B16 melanoma and Lewis lung carcinoma cells were respectively used to estimate the anti-tumor activity in vivo. The results indicated that oral administration of Rh(2) (5, 10 and 20 mg/kg body weight) alone has no obvious anti-tumor activity and genotoxic effect in mice, while Rh(2) synergistically enhanced the anti-tumor activity of cyclophosphamide (40 mg/kg body weight) in a dose-dependent manner.

Rh(2) decreased the micronucleus formation in polychromatic erythrocytes and DNA strand breaks in white blood cells in a dose-dependent way. These results suggest that ginsenoside Rh(2) is able to enhance the anti-tumor activity and decrease the genotoxic effect of cyclophosphamide (Wang, Zheng, Liu, Li, & Zheng, 2006).

Down-regulates MMP-9, Anti-metastatic

The effects of the purified ginseng components, panaxadiol (PD) and panaxatriol (PT), were examined on the expression of matrix metalloproteinase-9 (MMP-9) in highly metastatic HT1080 human fibrosarcoma cell line. A significant down-regulation of MMP-9 by PD and PT was detected by Northern blot analysis; however, the expression of MMP-2 was not changed by treatment with PD and PT. The results of the in vitro invasion assay revealed that PD and PT reduced tumor cell invasion through a reconstituted basement membrane in the transwell chamber. Because of the similarity of chemical structure between PD, PT and dexamethasone (Dexa), a synthetic glucocorticoid, we investigated whether the down-regulation of MMP-9 by PD and PT were mediated by the nuclear translocation of glucocorticoid receptor (GR). Increased GR in the nucleus of HT1080 human fibrosarcoma cells treated by PD and PT was detected by immunocytochemistry.

Western blot and gel retardation assays confirmed the increase of GR in the nucleus after treatment with PD and PT. These results suggest that GR-induced down-regulation of MMP-9 by PD and PT contributes to reduce the invasive capacity of HT1080 cells (Park et al., 1999).

Enhances 5-FU; Colorectal Cancer

Panaxadiol (PD) is the purified sapogenin of ginseng saponins, which exhibit anti-tumor activity. The possible synergistic anti-cancer effects of PD and 5-FU on a human colorectal cancer cell line, HCT-116, have been investigated.

The significant suppression on HCT-116 cell proliferation was observed after treatment with PD (25 microM) for 24 and 48 hours. Panaxadiol (25 microM) markedly (P < 0.05) enhanced the anti-proliferative effects of 5-FU (5, 10, 20 microM) on HCT-116 cells compared to single treatment of 5-FU for 24 and 48 hours.

Flow cytometric analysis on DNA indicated that PD and 5-FU selectively arrested cell-cycle progression in the G1 phase and S phase (P < 0.01), respectively, compared to the control condition. Combination use of 5-FU with PD significantly (P < 0.001) increased cell-cycle arrest in the S phase compared to that treated by 5-FU alone.

The combination of 5-FU and PD significantly enhanced the percentage of apoptotic cells when compared with the corresponding cell groups treated by 5-FU alone (P < 0.001). Panaxadiol hence enhanced the anti-cancer effects of 5-FU on human colorectal cancer cells through the regulation of cell-cycle transition and the induction of apoptotic cells (Li et al., 2009).

Colorectal Cancer

The possible synergistic anti-cancer effects of Panaxadiol (PD) and Epigallocatechin gallate (EGCG), on human colorectal cancer cells and the potential role of apoptosis in the synergistic activities, have been investigated.

Cell growth was suppressed after treatment with PD (10 and 20   µm) for 48   h. When PD (10 and 20   µm) was combined with EGCG (10, 20, and 30   µm), significantly enhanced anti-proliferative effects were observed in both cell lines. Combining 20   µm of PD with 20 and 30   µm of EGCG significantly decreased S-phase fractions of cells. In the apoptotic assay, the combination of PD and EGCG significantly increased the percentage of apoptotic cells compared with PD alone (p   <   0.01).

Data from this study suggested that apoptosis might play an important role in the EGCG-enhanced anti-proliferative effects of PD on human colorectal cancer cells (Du et al., 2013).

Colorectal Cancer; Irinotecan

Cell cycle analysis demonstrated that combining irinotecan treatment with panaxadiol significantly increased the G1-phase fractions of cells, compared with irinotecan treatment alone. In apoptotic assays, the combination of panaxadiol and irinotecan significantly increased the percentage of apoptotic cells compared with irinotecan alone (P<0.01). Increased activity of caspase-3 and caspase-9 was observed after treating with panaxadiol and irinotecan.

Data from this study suggested that caspase-3- and caspase-9-mediated apoptosis may play an important role in the panaxadiol enhanced anti-proliferative effects of irinotecan on human colorectal cancer cells (Du et al., 2012).

Anti-inflammatory

Ginsenoside Re inhibited IKK- β phosphorylation and NF- κ B activation, as well as the expression of pro-inflammatory cytokines, TNF- α and IL-1 β , in LPS-stimulated peritoneal macrophages, but it did not inhibit them in TNF- α – or PG-stimulated peritoneal macrophages. Ginsenoside Re also inhibited IRAK-1 phosphorylation induced by LPS, as well as IRAK-1 and IRAK-4 degradations in LPS-stimulated peritoneal macrophages.

Orally administered ginsenoside Re significantly inhibited the expression of IL-1 β and TNF- α on LPS-induced systemic inflammation and TNBS-induced colitis in mice. Ginsenoside Re inhibited colon shortening and myeloperoxidase activity in TNBS-treated mice. Ginsenoside Re reversed the reduced expression of tight-junction-associated proteins ZO-1, claudin-1, and occludin. Ginsenoside Re (20 mg/kg) inhibited the activation of NF- κ B in TNBS-treated mice. On the basis of these findings, ginsenoside Re may ameliorate inflammation by inhibiting the binding of LPS to TLR4 on macrophages (Lee et al., 2012).

Induces Apoptosis

Compound K activated an autophagy pathway characterized by the accumulation of vesicles, the increased positive acridine orange-stained cells, the accumulation of LC3-II, and the elevation of autophagic flux. Compound K activated the c-Jun NH2-terminal kinase (JNK) signaling pathway, whereas down-regulation of JNK by its specific inhibitor SP600125 or by small interfering RNA against JNK attenuated autophagy-mediated cell death in response to compound K. Compound K also provoked apoptosis, as evidenced by an increased number of apoptotic bodies and sub-G1 hypodiploid cells, enhanced activation of caspase-3 and caspase-9, and modulation of Bcl-2 and Bcl-2-associated X protein expression (Kim et al., 2013b).

Lung Cancer

AD-1, a ginsenoside derivative, concentration-dependently reduces lung cancer cell viability without affecting normal human lung epithelial cell viability. In A549 and H292 lung cancer cells, AD-1 induces G0/G1 cell-cycle arrest, apoptosis and ROS production. The apoptosis can be attenuated by a ROS scavenger – N-acetylcysteine (NAC). In addition, AD-1 up-regulates the expression of p38 and ERK phosphorylation. Addition of a p38 inhibitor, SB203580, suppresses the AD-1-induced decrease in cell viability. Furthermore, genetic silencing of p38 attenuates the expression of p38 and decreases the AD-1-induced apoptosis.

These data support development of AD-1 as a potential agent for lung cancer therapy (Zhang et al., 2013).

Pediatric AML

In this study, Chen et al. (2013) demonstrated that compound K, a major ginsenoside metabolite, inhibited the growth of the clinically relevant pediatric AML cell lines in a time- and dose-dependent manner. This growth-inhibitory effect was attributable to suppression of DNA synthesis during cell proliferation and the induction of apoptosis was accompanied by DNA double strand breaks. Findings suggest that as a low toxic natural reagent, compound K could be a potential drug for pediatric AML intervention and to improve the outcome of pediatric AML treatment.

Melanoma

Jeong et al. (2013) isolated 12 ginsenoside compounds from leaves of Panax ginseng and tested them in B16 melanoma cells. It significantly reduced melanin content and tyrosinase activity under alpha-melanocyte stimulating hormone- and forskolin-stimulated conditions. It significantly reduced the cyclic AMP (cAMP) level in B16 melanoma cells, and this might be responsible for the regulation down of MITF and tyrosinase. Phosphorylation of a downstream molecule, a cAMP response-element binding protein, was significantly decreased according to Western blotting and immunofluorescence assay. These data suggest that A-Rh4 has an anti-melanogenic effect via the protein kinase A pathway.

Leukemia

Rg1 can significantly inhibit the proliferation of leukemia cell line K562 in vitro and arrest the cells in G2/M phase. The percentage of positive cells stained by SA-beta-Gal was dramatically increased (P < 0.05) and the expression of cell senescence-related genes was up-regulated. The observation of ultrastructure showed cell volume increase, heterochromatin condensation and fragmentation, mitochondrial volume increase, and lysosomes increase in size and number (Cai et al., 2012).

Ginsenosides and CYP 450 Enzymes

In vitro experiments have shown that both crude ginseng extract and total saponins at high concentrations (.2000 mg/ml) inhibited CYP2E1 activity in mouse and human microsomes (Nguyen et al., 2000). Henderson et al. (1999) reported the effects of seven ginsenosides and two eleutherosides (active components of the ginseng root) on the catalytic activity of a panel of cDNA-expressed CYP isoforms (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4) using 96-well plate fluorometrical assay.

Of the constituents tested, Ginsenoside Rd caused weak inhibitory activity against CYP3A4, CYP2D6, CYP2C19,and CYP2C9, but ginsenoside Re and ginsenoside Rf (200 mM) produced a 70% and 54%increase in the activity of CYP2C9 and CYP3A4, respectively. The authors suggested that the activating effects of ginsenosides on CYP2C9 and CYP3A4 might be due to a matrix effect caused by the test compound fluorescing at the same wavelength as the metabolite of the marker substrates. Chang et al. (2002) reported the effects of two types of ginseng extract and ginsenosides (Rb1, Rb2, Rc, Rd, Re, Rf, and Rg1) on CYP1 catalytic activities.

The ginseng extracts inhibited human recombinant CYP1A1, CYP1A2, and CYP1B1 activities in a concentration-dependent manner. Rb1, Rb2, Rc, Rd, Re, Rf, and Rg1 at low concentrations had no effect on CYP1 activities, but Rb1, Rb2, Rc, Rd, and Rf at a higher ginsenoside concentration (50 mg/ml) inhibited these activities. These results indicated that various ginseng extracts and ginsenosides inhibited CYP1 activity in an enzyme-selective and extract-specific manner (Zhou et al., 2003).

References

An IS, An S, Kwon KJ, Kim YJ, Bae S. (2012). Ginsenoside Rh2 mediates changes in the microRNA expression profile of human non-small-cell lung cancer A549 cells. Oncol Rep, 29(2):523-8. doi: 10.3892/or.2012.2136.



Bae EA, Han MJ, Choo MK et al. (2002). Metabolism of 20(S)- and 20(R)-ginsenoside R-g3 by human intestinal bacteria and its relation to in vitro biological activities. Biol. Pharm. Bull, 25:58–63.


Cai S, Zhou Y, Liu J, et al. (2012). Experimental study on human leukemia cell line K562 senescence induced by ginsenoside Rg1. Zhongguo Zhong Yao Za Zhi, 37(16):2424-8.


Cao M, Yu HS, Song XB, Ma BP. (2012) Advances in the study of derivatization of ginsenosides and their anti-tumor structure-activity relationship. Yao Xue Xue Bao, 47(7):836-43.


Chang TKH, Chen J, Benetton SA et al. (2002). In vitro effect of standardized ginseng extracts and individual ginsenosides on the catalytic activity of human CYP1A1, CYP1A2, and CYP1B1. Drug Metab. Dispos, 30:378–384.


Chen Y, Xu Y, Zhu Y, Li X. (2013). Anti-cancer effects of ginsenoside compound k on pediatric acute myeloid leukemia cells. Cancer Cell Int, 13(1):24. doi: 10.1186/1475-2867-13-24.


Choi YJ, Lee HJ, Kang DW, et al. (2013). Ginsenoside Rg3 induces apoptosis in the U87MG human glioblastoma cell line through the MEK signaling pathway and reactive oxygen species. Oncol Rep, 30(3): 1362-1370. doi: 10.3892/or.2013.2555.


Christensen LP. (2009). Ginsenosides chemistry, biosynthesis, analysis, and potential health effects. Adv Food Nutr Res., 55:1-99. doi: 10.1016/S1043-4526(08)00401-4.


Chung KS, Cho SH, Shin JS, et al. (2013). Ginsenoside Rh2 induces Cell-cycle arrest and differentiation in human leukemia cells by upregulating TGF- β expression. Carcinogenesis, 34(2):331-40. doi: 10.1093/carcin/bgs341.


Du GJ, Wang CZ, Zhang ZY, et al. (2012) Caspase-mediated pro-apoptotic interaction of panaxadiol and irinotecan in human colorectal cancer cells. J Pharm Pharmacol, 64(5):727-34. doi: 10.1111/j.2042-7158.2012.01463.x.


Du GJ, Wang CZ, Qi LW, et al. (2013). The synergistic apoptotic interaction of panaxadiol and epigallocatechin gallate in human colorectal cancer cells. Phytother Res, 27(2):272-7. doi: 10.1002/ptr.4707.


Henderson GL, Harkey MR, Gershwin, ME, et al. (1999). Effects of ginseng components on c-DNA-expressed cytochrome P450 enzyme catalytic activity. Life Sci, PL209–PL214.


Jeong YM, Oh WK, Tran TL, et al. (2013). Aglycone of Rh4 inhibits melanin synthesis in B16 melanoma cells: possible involvement of the protein kinase A pathway. Biosci Biotechnol Biochem, 77(1):119-25.


Ji Y, Rao Z, Cui J, et al. (2012). Ginsenosides extracted from nanoscale Chinese white ginseng enhances anti-cancer effect. J Nanosci Nanotechnol, 12(8):6163-7.


Jia WW, Bu X, Philips D, et al. (2004). Rh2, a compound extracted from ginseng, hypersensitizes Multi-drug-resistant tumor cells to chemotherapy. Can J Physiol Pharmacol, 82(7):431-7.


Jia JM, Wang ZQ, Wu LJ, Wu YL. (2008). Advance of pharmacological study on ginsenoside Rb1. Zhongguo Zhong Yao Za Zhi, 33(12):1371-7.


Kim YJ, Yamabe N, Choi P, et al. (2013a) Efficient Thermal Deglycosylation of Ginsenoside Rd and Its Contribution to the Improved Anti-cancer Activity of Ginseng. J Agric Food Chem.


Kim AD, Kang KA, Kim HS, et al. (2013b). A ginseng metabolite, compound K, induces autophagy and apoptosis via generation of reactive oxygen species and activation of JNK in human colon cancer cells. Cell Death Dis, 4:e750. doi: 10.1038/cddis.2013.273.


Kim SM, Lee SY, Cho JS, et al. (2010). Combination of ginsenoside Rg3 with docetaxel enhances the susceptibility of prostate cancer cells via inhibition of NF-kappaB. Eur J Pharmacol, 631(1-3):1-9. doi: 10.1016/j.ejphar.2009.12.018.


Kim SM, Lee SY, Yuk DY, et al. (2009). Inhibition of NF-kappaB by ginsenoside Rg3 enhances the susceptibility of colon cancer cells to docetaxel. Arch Pharm Res, 32:755–765. doi: 10.1007/s12272-009-1515-4.


King ML, Adler SR, Murphy LL. (2006). Extraction-dependent effects of American ginseng (Panax quinquefolium) on human breast cancer cell proliferation and estrogen receptor activation. Integr Cancer Ther, 5(3):236-43.


Kwon HY, Kim EH, Kim SW, et al. (2008). Selective toxicity of ginsenoside Rg3 on Multi-drug-resistant cells by membrane fluidity modulation. Arch Pharm Res, 31(2):171-7.


Lee IA, Hyam SR, Jang SE, Han MJ, Kim DH. (2012). Ginsenoside Re ameliorates inflammation by inhibiting the binding of lipopolysaccharide to TLR4 on macrophages. J Agric Food Chem, 60(38):9595-602.


Li XL, Wang CZ, Mehendale SR, et al. (2009). Panaxadiol, a purified ginseng component, enhances the anti-cancer effects of 5-fluorouracil in human colorectal cancer cells. Cancer Chemother Pharmacol, 64(6):1097-104. doi: 10.1007/s00280-009-0966-0.


Mehendale S, Aung H, Wang A, et al. (2005). American ginseng berry extract and ginsenoside Re attenuate cisplatin-induced kaolin intake in rats. Cancer Chemotherapy and Pharmacology, 56(1):63-9. doi: 10.1007/s00280-004-0956-1.


Nguyen TD, Villard PH, Barlatier A et al. (2000). Panax vietnamensis protects mice against carbon tetrachloride-induced hepatotoxicity without any modification of CYP2E1 gene expression. Planta Med, 66:714–719.


Pan J, Zhang Q, Li K, et al. (2013). Chemoprevention of lung squamous cell carcinoma by ginseng. Cancer Prev Res (Phila), 6(6):530-9. doi: 10.1158/1940-6207.CAPR-12-0366.


Park MT, Cha HJ, Jeong JW, et al. (1999). Glucocorticoid receptor-induced down-regulation of MMP-9 by ginseng components, PD and PT contributes to inhibition of the invasive capacity of HT1080 human fibrosarcoma cells. Mol Cells, 9(5):476-83.


Wang CZ and Yuan CS. (2008). Potential Role of Ginseng in the Treatment of Colorectal Cancer. Am. J. Chin. Med, 36:1019. doi: 10.1142/S0192415X08006545


Wang Z, Zheng Q, Liu K, Li G, Zheng R. (2006). Ginsenoside Rh(2) enhances anti-tumor activity and decreases genotoxic effect of cyclophosphamide. Basic Clin Pharmacol Toxicol, 98(4):411-5.


Wang CZ, Zhang B, Song WX, et al. (2006). Steamed American ginseng berry: ginsenoside analyzes and anti-cancer activities. Journal of agricultural and food chemistry, 54(26):9936-42.


Yun UJ, Lee JH, Koo KH, et al. (2013). Lipid raft modulation by Rp1 reverses Multi-drug resistance via inactivating MDR-1 and Src inhibition. Biochem Pharmacol, 85(10):1441-53. doi: 10.1016/j.bcp.2013.02.025.


Zhang LH, Jia YL, Lin XX, et al. (2013). AD-1, a novel ginsenoside derivative, shows anti-lung cancer activity via activation of p38 MAPK pathway and generation of reactive oxygen species. Biochim Biophys Acta, 1830(8):4148-59. doi: 10.1016/j.bbagen.2013.04.008.


Zhou Sf, Gao Yh, Jiang Wq et al. (2003) Interactions of Herbs with Cytochrome P450. DRUG METABOLISM REVIEWS, 35(1):35–98.