Ginsenoside Absorption Rate and Extent Enhancement of Black Ginseng (CJ EnerG) over Red Ginseng in Healthy Adults

Saebyul Yoo, Bom-I Park, Do-Hyun Kim, Sooyoung Lee, Seung-Hoon Lee, Wang-Seob Shim, Yong Ki Seo, Kimoon Kang, Kyung-Tae Lee, Sung-Vin Yim, Do Yu Soung, Bo-Hyung Kim, Saebyul Yoo, Bom-I Park, Do-Hyun Kim, Sooyoung Lee, Seung-Hoon Lee, Wang-Seob Shim, Yong Ki Seo, Kimoon Kang, Kyung-Tae Lee, Sung-Vin Yim, Do Yu Soung, Bo-Hyung Kim

Abstract

Red ginseng (RG) and black ginseng (BG, CJ EnerG) were prepared from fresh ginseng using one and nine cycles of steaming and drying, respectively. This process reduces the molecular weight (MW) of ginsenoside-active compounds in ginseng by removing sugar moieties from their dammaranes. We compared the pharmacokinetic characteristics of ginsenosides between BG comprising mainly low-MW ginsenosides (Rg3, Rg5, Rk1, and Rh1) and RG that predominantly contains high-MW ginsenosides (Rb1, Rb2, Rc, Rd, Re, and Rg1). The safety profiles and tolerability were also studied using a randomized, double-blind, single-dose, crossover clinical trial. A combination of Rb1, Rg1, and Rg3, well-known representative and functional RG components, exhibited a 1 h faster absorption rate (Tmax) and 58% higher exposure (24 h area under the concentration-time curve, AUC24) in BG than in RG. Furthermore, the combination of Rg3, Rg5, and Rk1, the major and most efficient components in BG, displayed 824% higher absorption (AUC24) in BG than in RG. The total ginsenoside showed a 5 h rapid intestinal absorption (Tmax) and 79% greater systemic exposure (AUC24) in BG than in RG. No clinically significant findings were observed in terms of safety or tolerability. Thus, BG extract was more effective than RG extract.

Keywords: black ginseng; ginsenosides; pharmacokinetics.

Conflict of interest statement

B.-I.P., Y.K.S., K.K. and D.Y.S. are employees of the CJ CheilJedang Corporation. B.-I.P. analyzed and visualized RG and BG ginsenosides. She also wrote the figure legend (Figure 1). Y.K.S., K.K. and D.Y.S. were involved in the conceptualization, validation, and funding acquisition. D.Y.S. also reviewed the manuscript. The other authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Multiple steaming cycles and gut microbial activity reduce the molecular weight (MW) of ginsenosides in ginseng. Ginsenosides are tetracyclic triterpene (dammarane)-type saponins and are divided into protopanaxadiol (PPD) type (e.g., Rb1, Rb2, Rc, Rd, Rg3, Rk1, Rg5, CK, Rh2, and PPD) and protopanaxatriol (PPT) type (e.g., Re, Rg1, Rh1, and PPT). Based on their size, ginsenosides are divided into three groups: (1) high molecular weight (HMW) (>800) ginsenosides found in red ginseng (RG) such as Rb1 (MW: 1109), Rb2 (MW: 1079), Rc (MW: 1079), Rd (MW: 1079), Re (MW: 947), and Rg1 (MW: 801); (2) low MW (LMW) (>637,

Figure 2

Chromatograms obtained from high-performance liquid…

Figure 2

Chromatograms obtained from high-performance liquid chromatography analysis of ginsenosides in ( A )…

Figure 2
Chromatograms obtained from high-performance liquid chromatography analysis of ginsenosides in (A) black ginseng and (B) red ginseng extracts. Detection at a wavelength of 203 nm.

Figure 3

Flowchart showing subject disposition.

Figure 3

Flowchart showing subject disposition.

Figure 3
Flowchart showing subject disposition.

Figure 4

Mean (± standard error (SE))…

Figure 4

Mean (± standard error (SE)) plasma concentration–time profiles of ginsenosides after administration of…

Figure 4
Mean (± standard error (SE)) plasma concentration–time profiles of ginsenosides after administration of a single dose of 9 g of red ginseng (RG) (red empty symbol and dashed line) and black ginseng (BG) (black filled symbol and solid line) extract. The inserted bar plot showing the mean (±SE) for the 0 to 24 h area under the concentration–time curve (AUC24) of RG (red color) and BG (black color) extract: (A) Rb1; (B) Rb2; (C) Rc; (D) Rd; (E) Rg3; (F) Rg5; (G) Rk1; (H) Rh2; (I) CK; (J) PPD; (K) Re; (L) Rg1; (M) Rh1; (N) PPT. * Statistically significant at p < 0.05.

Figure 5

Pharmacokinetic characteristics of functional ginsenoside…

Figure 5

Pharmacokinetic characteristics of functional ginsenoside groups after the administration of a single dose…

Figure 5
Pharmacokinetic characteristics of functional ginsenoside groups after the administration of a single dose of red ginseng (RG) (red color) and black ginseng (BG) (black color) extracts: (AE) Functional ginsenoside group with Rb1, Rg1 and Rg3; (FJ) functional ginsenoside group with Rg3, Rg5, and Rk1; (A,F) mean (± standard error (SE)) plasma concentration–time profile of group 1; (B,G) mean (±SE) for Cmax; (C,H) median (inter-quartile range (IQR)) boxes for Tmax (lines for 1.5 times the IQR and dots for outliers); (D,I) mean (±SE) for area under the concentration–time curve AUC from 0 to 24 h (AUC24); (E,J) mean (±SE) for AUC from 0 to the actual time point relevant to the last concentration above the lower limit of quantification (LLOQ) (AUClast). * Statistically significant at p < 0.05.

Figure 6

Pharmacokinetic characteristics of total ginsenosides…

Figure 6

Pharmacokinetic characteristics of total ginsenosides after the administration of a single dose of…

Figure 6
Pharmacokinetic characteristics of total ginsenosides after the administration of a single dose of red ginseng (RG) (red color) and black ginseng (BG) (black color) extracts: (A) mean (±standard error (SE)) plasma concentration–time profile; (B) mean (±SE) for Cmax; (C) median (inter-quartile range (IQR)) boxes for Tmax (lines for 1.5 times the IQR and dots for outliers); (D) mean (±SE) for area under the concentration–time curve from 0 to 24 h (AUC24); (E) mean (±SE) for AUC from 0 to the actual point in time with respect to the last concentration above the lower limit of quantification (LLOQ) (AUClast). * Statistically significant at p < 0.05.
Figure 2
Figure 2
Chromatograms obtained from high-performance liquid chromatography analysis of ginsenosides in (A) black ginseng and (B) red ginseng extracts. Detection at a wavelength of 203 nm.
Figure 3
Figure 3
Flowchart showing subject disposition.
Figure 4
Figure 4
Mean (± standard error (SE)) plasma concentration–time profiles of ginsenosides after administration of a single dose of 9 g of red ginseng (RG) (red empty symbol and dashed line) and black ginseng (BG) (black filled symbol and solid line) extract. The inserted bar plot showing the mean (±SE) for the 0 to 24 h area under the concentration–time curve (AUC24) of RG (red color) and BG (black color) extract: (A) Rb1; (B) Rb2; (C) Rc; (D) Rd; (E) Rg3; (F) Rg5; (G) Rk1; (H) Rh2; (I) CK; (J) PPD; (K) Re; (L) Rg1; (M) Rh1; (N) PPT. * Statistically significant at p < 0.05.
Figure 5
Figure 5
Pharmacokinetic characteristics of functional ginsenoside groups after the administration of a single dose of red ginseng (RG) (red color) and black ginseng (BG) (black color) extracts: (AE) Functional ginsenoside group with Rb1, Rg1 and Rg3; (FJ) functional ginsenoside group with Rg3, Rg5, and Rk1; (A,F) mean (± standard error (SE)) plasma concentration–time profile of group 1; (B,G) mean (±SE) for Cmax; (C,H) median (inter-quartile range (IQR)) boxes for Tmax (lines for 1.5 times the IQR and dots for outliers); (D,I) mean (±SE) for area under the concentration–time curve AUC from 0 to 24 h (AUC24); (E,J) mean (±SE) for AUC from 0 to the actual time point relevant to the last concentration above the lower limit of quantification (LLOQ) (AUClast). * Statistically significant at p < 0.05.
Figure 6
Figure 6
Pharmacokinetic characteristics of total ginsenosides after the administration of a single dose of red ginseng (RG) (red color) and black ginseng (BG) (black color) extracts: (A) mean (±standard error (SE)) plasma concentration–time profile; (B) mean (±SE) for Cmax; (C) median (inter-quartile range (IQR)) boxes for Tmax (lines for 1.5 times the IQR and dots for outliers); (D) mean (±SE) for area under the concentration–time curve from 0 to 24 h (AUC24); (E) mean (±SE) for AUC from 0 to the actual point in time with respect to the last concentration above the lower limit of quantification (LLOQ) (AUClast). * Statistically significant at p < 0.05.

References

    1. Christensen L.P. Ginsenosides: Chemistry, biosynthesis, analysis, and potential health effects. Adv. Food Nutr. Res. 2008;55:1–99.
    1. Kang K.S., Kim H.Y., Yamabe N., Nagai R., Yokozawa T. Protective effect of sun ginseng against diabetic renal damage. Biol. Pharm. Bull. 2006;29:1678–1684. doi: 10.1248/bpb.29.1678.
    1. Qi L.-W., Wang C.-Z., Yuan C.-S. American ginseng: Potential structure–function relationship in cancer chemoprevention. Biochem. Pharmacol. 2010;80:947–954. doi: 10.1016/j.bcp.2010.06.023.
    1. Yun T.-K. Panax ginseng—A non-organ-specific cancer preventive? Lancet Oncol. 2001;2:49–55. doi: 10.1016/S1470-2045(00)00196-0.
    1. Kim D.-W., Kim Y.-J., Lee Y.-J., Min J.-W., Kim S.-Y., Yang D.-C. Conversion of ginsenosides by 9 repetitive steamings and dryings process of Korean ginseng root and its inhibition of BACE-1 activity. J. Physiol. Pathol. Korean Med. 2008;22:1557–1561.
    1. Chen W., Guo Y., Yang W., Zheng P., Zeng J., Tong W. Protective effect of ginsenoside Rb1 on integrity of blood–brain barrier following cerebral ischemia. Exp. Brain Res. 2015;233:2823–2831. doi: 10.1007/s00221-015-4352-3.
    1. Gao X.-Q., Yang C.-X., Chen G.-J., Wang G.-Y., Chen B., Tan S.-K., Liu J., Yuan Q.-L. Ginsenoside Rb1 regulates the expressions of brain-derived neurotrophic factor and caspase-3 and induces neurogenesis in rats with experimental cerebral ischemia. J. Ethnopharmacol. 2010;132:393–399. doi: 10.1016/j.jep.2010.07.033.
    1. Salim K.N., McEwen B.S., Chao H.M. Ginsenoside Rb1 regulates ChAT, NGF and trkA mRNA expression in the rat brain. Mol. Brain Res. 1997;47:177–182. doi: 10.1016/S0169-328X(97)00042-9.
    1. Wu Y., Xia Z.-Y., Dou J., Zhang L., Xu J.-J., Zhao B., Lei S., Liu H.-M. Protective effect of ginsenoside Rb1 against myocardial ischemia/reperfusion injury in streptozotocin-induced diabetic rats. Mol. Biol. Rep. 2011;38:4327–4335. doi: 10.1007/s11033-010-0558-4.
    1. Zhang J., Qu Z., Liu Y., Deng H. Preliminary study on antiamnestic mechanism of ginsenoside Rg1 and Rb1. Chin. Med. J. 1990;103:932–938. doi: 10.1016/0014-2999(90)94602-T.
    1. Zhang Y.-J., Zhang X.-L., Li M.-H., Iqbal J., Bourantas C.V., Li J.-J., Su X.-Y., Muramatsu T., Tian N.-L., Chen S.-L. The Ginsenoside Rg1 Prevents Transverse Aortic Constriction–Induced Left Ventricular Hypertrophy and Cardiac Dysfunction by Inhibiting Fibrosis and Enhancing Angiogenesis. J. Cardiovasc. Pharmacol. 2013;62:50–57. doi: 10.1097/FJC.0b013e31828f8d45.
    1. Zhou W., Chai H., Lin P.H., Lumsden A.B., Yao Q., Chen C. Ginsenoside Rb1 blocks homocysteine-induced endothelial dysfunction in porcine coronary arteries. J. Vasc. Surg. 2005;41:861–868. doi: 10.1016/j.jvs.2005.01.054.
    1. Zhou Y., Li H.-Q., Lu L., Fu D.-L., Liu A.-J., Li J.-H., Zheng G.-Q. Ginsenoside Rg1 provides neuroprotection against blood brain barrier disruption and neurological injury in a rat model of cerebral ischemia/reperfusion through downregulation of aquaporin 4 expression. Phytomedicine. 2014;21:998–1003. doi: 10.1016/j.phymed.2013.12.005.
    1. Kim E.-H., Kim S.-W., Park S.-J., Kim S., Yu K.-M., Kim S.G., Lee S.H., Seo Y.-K., Cho N.-H., Kang K. Greater efficacy of black ginseng (CJ EnerG) over red ginseng against lethal influenza A virus infection. Nutrients. 2019;11:1879. doi: 10.3390/nu11081879.
    1. Lee Y.Y., Saba E., Irfan M., Kim M., Yi-Le Chan J., Jeon B.S., Choi S.K., Rhee M.H. The anti-inflammatory and anti-nociceptive effects of Korean black ginseng. Phytomedicine. 2019;54:169–181. doi: 10.1016/j.phymed.2018.09.186.
    1. Lee M.R., Ma J.Y., Sung C.K. Chronic dietary ginseng extract administration ameliorates antioxidant and cholinergic systems in the brains of aged mice. J. Ginseng Res. 2017;41:615–619. doi: 10.1016/j.jgr.2017.06.002.
    1. Jin S., Jeon J.-H., Lee S., Kang W.Y., Seong S.J., Yoon Y.-R., Choi M.-K., Song I.-S. Detection of 13 ginsenosides (Rb1, Rb2, Rc, Rd, Re, Rf, Rg1, Rg3, Rh2, F1, Compound K, 20(S)-Protopanaxadiol, and 20(S)-Protopanaxatriol) in human plasma and application of the analytical method to human pharmacokinetic studies following two week-repeated administration of red ginseng extract. Molecules. 2019;24:2618.
    1. Health Functional Food Code (No. 2020-92). MFDS. [(accessed on 23 September 2020)]; Available online: .
    1. Lee B., Sur B., Park J., Kim S.-H., Kwon S., Yeom M., Shim I., Lee H., Hahm D.-H. Ginsenoside rg3 alleviates lipopolysaccharide-induced learning and memory impairments by anti-inflammatory activity in rats. Biomol. Ther. 2013;21:381–390. doi: 10.4062/biomolther.2013.053.
    1. Li W., Yan M.-H., Liu Y., Liu Z., Wang Z., Chen C., Zhang J., Sun Y.-S. Ginsenoside Rg5 ameliorates cisplatin-induced nephrotoxicity in mice through inhibition of inflammation, oxidative stress, and apoptosis. Nutrients. 2016;8:566. doi: 10.3390/nu8090566.
    1. Hu J.-N., Xu X.-Y., Li W., Wang Y.-M., Liu Y., Wang Z., Wang Y.-P. Ginsenoside Rk1 ameliorates paracetamol-induced hepatotoxicity in mice through inhibition of inflammation, oxidative stress, nitrative stress and apoptosis. J. Ginseng Res. 2019;43:10–19. doi: 10.1016/j.jgr.2017.07.003.
    1. Kim J.-K., Choi M.S., Jeung W., Ra J., Yoo H.H., Kim D.-H. Effects of gut microbiota on the pharmacokinetics of protopanaxadiol ginsenosides Rd, Rg3, F2, and compound K in healthy volunteers treated orally with red ginseng. J. Ginseng Res. 2020;44:611–618. doi: 10.1016/j.jgr.2019.05.012.
    1. Xie J.-T., Mehendale S.R., Li X., Quigg R., Wang X., Wang C.-Z., Wu J.A., Aung H.H., Rue P.A., Bell G.I. Anti-diabetic effect of ginsenoside Re in ob/ob mice. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2005;1740:319–325. doi: 10.1016/j.bbadis.2004.10.010.
    1. Wang Z., Li Y.-F., Han X.-Y., Sun Y.-S., Zhang L.-X., Liu W., Liu X.-X., Li W., Liu Y.-Y. Kidney protection effect of ginsenoside re and its underlying mechanisms on cisplatin-induced kidney injury. Cell. Physiol. Biochem. 2018;48:2219–2229. doi: 10.1159/000492562.
    1. Zhou X.-M., Cao Y.-L., Dou D.-Q. Protective effect of ginsenoside-Re against cerebral ischemia/reperfusion damage in rats. Biol. Pharm. Bull. 2006;29:2502–2505. doi: 10.1248/bpb.29.2502.
    1. Xie C.-L., Li J.-H., Wang W.-W., Zheng G.-Q., Wang L.-X. Neuroprotective effect of ginsenoside-Rg1 on cerebral ischemia/reperfusion injury in rats by downregulating protease-activated receptor-1 expression. Life Sci. 2015;121:145–151. doi: 10.1016/j.lfs.2014.12.002.
    1. Quan L.-H., Cheng L.-Q., Kim H.-B., Kim J.-H., Son N.-R., Kim S.-Y., Jin H.-O., Yang D.-C. Bioconversion of ginsenoside Rd into compound K by Lactobacillus pentosus DC101 isolated from Kimchi. J. Ginseng Res. 2010;34:288–295. doi: 10.5142/jgr.2010.34.4.288.
    1. Liu L., Zhu X.-M., Wang Q.-J., Zhang D.-L., Fang Z.-M., Wang C.-Y., Wang Z., Sun B.-S., Wu H., Sung C.-K. Enzymatic preparation of 20(S, R)-protopanaxadiol by transformation of 20(S, R)-Rg3 from black ginseng. Phytochemistry. 2010;71:1514–1520. doi: 10.1016/j.phytochem.2010.05.007.
    1. Hou J., Xue J., Wang C., Liu L., Zhang D., Wang Z., Li W., Zheng Y., Sung C. Microbial transformation of ginsenoside Rg3 to ginsenoside Rh2 by Esteya vermicola CNU 120806. World J. Microbiol. Biotechnol. 2012;28:1807–1811. doi: 10.1007/s11274-011-0946-5.
    1. Shi Q., Li J., Feng Z., Zhao L., Luo L., You Z., Li D., Xia J., Zuo G., Chen D. Effect of ginsenoside Rh2 on the migratory ability of HepG2 liver carcinoma cells: Recruiting histone deacetylase and inhibiting activator protein 1 transcription factors. Mol. Med. Rep. 2014;10:1779–1785. doi: 10.3892/mmr.2014.2392.
    1. Li S., Gao Y., Ma W., Guo W., Zhou G., Cheng T., Liu Y. EGFR signaling-dependent inhibition of glioblastoma growth by ginsenoside Rh2. Tumour Biol. 2014;35:5593–5598. doi: 10.1007/s13277-014-1739-x.
    1. Baatar D., Siddiqi M.Z., Im W.T., Ul Khaliq N., Hwang S.G. Anti-Inflammatory Effect of Ginsenoside Rh2-Mix on Lipopolysaccharide-Stimulated RAW 264.7 Murine Macrophage Cells. J. Med. Food. 2018;21:951–960. doi: 10.1089/jmf.2018.4180.

Source: PubMed

3
Tilaa