DOI QR코드

DOI QR Code

Autophagy and its regulation by ginseng components

  • Received : 2018.11.25
  • Accepted : 2018.12.26
  • Published : 2019.07.15

Abstract

Autophagy is the sequential process whereby cell components are degraded, which can occur due to nutrient deprivation. Its regulation has an essential role in many diseases, functioning in both cell survival and cell death. Autophagy starts when mTORC1 is inhibited, resulting in the activation of several complexes to form a cargo that fuses with a lysosome, where it undergoes degradation. In this review, we describe a plant extract that is well known in Korea, namely Korean ginseng extract; we studied how its derivatives and metabolites can regulate autophagy and thus mediate the pathogenesis of certain diseases.

Keywords

1. Introduction

Autophagy is the sequential process of cytoplasm component degradation by lysosomes [1,2]. It plays an essential role in coun- tering certain diseases and conditions, such as cancer, infection, aging, heart diseases, and neurodegradation [3,4]. There are different types of autophagy, classified by their mechanisms. Certain articles have classified autophagy into two types: nonse- lective (macroautophagy) and selective (classified by the cytosolic cargo sequestered) [3,5]. Other articles have classified autophagy into three main types: macroautophagy, microautophagy, and chaperone-mediated autophagy [4,6,7]. Autophagy generally oc- curs due to nutrient deprivation and/or starvation, functioning in both cell survival and cell death [8].

Macroautophagy, which is commonly referred as autophagy, is involved in several pathways to form double membrane vesicles (autophagosomes) and to sequester cytoplasmic components through the inhibition of the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway [9,10]. This inhibition ac- tivates the downstream complex Unc-51 like autophagy activating kinase 1/2 (ULK1/2), composed of ULK1/2, Atg13, and FIP200 [11]. This complex then triggers the Beclin-1/Phosphatidylinositol 3- kinase catalytic subunit type 3 (PI3KC3) complex to initiate phagopore formation. This in turn leads to autophagosome for- mation through the microtubule light-chain 3 protein or LC3 by activation through ubiquitin-like conjugation pathways [auto- phagy-related protein (ATG) conjugates] [12]. The autophagosome then fuses with a lysosome (autolysosome), and the materials in- side are degraded. Furthermore, the proteins involved in autophagy would also be recycled at the end of the process.

While autophagy has a functional role in cell survival, it has several pathways in common with apoptosis, a pathway that causes cell death. Autophagy and apoptosis regulate each another: For example, autophagy mediates cell death by degrading the negative regulator of apoptosis signaling, particularly in the caspase signaling pathway [13], or by degrading catalase, increasing reac- tive oxygen species (ROS) accumulation, and leading to cell death [14]. On the other hand, apoptosis regulators such as Bcl-2 and Bcl- XL prevent autophagy by interacting with Beclin-1 on their BH3 domain. This can be disrupted by BH3 mimetics, for example, Bcl-2- associated death promoter (BAD), to initiate autophagy [15]. The implication of autophagy involvement in some diseases is attract- ing attention. A number of cancer treatments involve autophagy inhibition or activation. In this review, we focus on how natural sources take part in autophagy regulation, in particular a very famous plant in Korea, Korean ginseng, and its components.

2. Korean ginseng extract and its components

Korean ginseng (Panax ginseng Meyer) belongs to the Araliaceae family [16]. Korean ginseng has been found to have antidiabetic, antiinflammatory, antimicrobial, antifungal, neuroprotective, anti- oxidant, anticancer, and antitumor activity [17-22]. The plant is especially rich in ginsenosides, a group of triterpene saponins, which comprise the active components [23]. Ginsenosides are classified into two categories based on their chemical structures, 20-S-protopanaxadiol and 20-S-protopanaxatriol [24]. Some gin- senoside components have been reported to play a role in certain diseases. Some ginsenoside derivatives such as Rb1, Rb2, Rg3, Rg1, and Rb1’s metabolite compound K (Fig. 1) have been reported to have antidiabetic activity, involving the sites of activation or inhi- bition of proteins responsible for diabetes [25e29]. Some ginse- noside derivatives also have antiinflammatory activity; for example, ginsenoside Rg1 decreases nitric oxide (NO) and tumor necrosis factor (TNF)-α levels in macrophage cells [30], as well as interleukin (IL)-1β and IL-6 in vivo [31]. There is also a report that the nuclear factor (NF)-κB signaling pathway is downregulated in the presence of ginsenoside Rg3 in lung tissue of humans with asthma [32]. Ginsenoside Rp1 downregulates the insulin-like growth factor 1 receptor (IGF-1R)/protein kinase B (Akt) pathway in breast cancer cells, reducing their proliferation and colony forma- tion [33]. In addition, ginsenoside Rk3 has been found to down- regulate the expression of cyclin D1 and CDK4, followed by the upregulation of p21, in lung cancer in vitro and in vivo [34].

Fig. 1. Chemical structure of ginseng components. (A) Ginsenoside Rh2. (B) Ginsenoside F2. (C) Ginsenoside Rb1. (D) Compound K

3. Regulation of autophagy by Korean ginseng extract

The components and derivatives of Korean ginseng extract (KGE) play important roles in treating certain serious diseases, and the effects of Korean ginseng on autophagy may be one of the mechanisms (Table 1 and Fig. 2). Several studies have investigated KGE-mediated autophagy. Cha et al [35] reported that the extract had the potential to attenuate testicular dysfunction as treatment decreased molecules responsible for spermatogenesis and down- regulated the mRNA levels of sex hormone receptors including the androgen, luteinizing hormone, and follicle-stimulating hormone receptors. Concomitantly, Korean Red Ginseng extract (RGE) also modulated autophagy responses by decreasing the mTORC1 mRNA and protein levels after they had been elevated by doxycycline (DOX)-induced testicular damage [35]. Nonetheless, further study is needed to determine the stage at which autophagy regulates testicular dysfunction and vice versa.

Table 1 Target diseases and autophagy-mediated molecular targets responding to ginseng components

Fig. 2. Schematic flow chart of autophagy-mediated molecular targets regarding ginseng components. AMPK, AMP-activated protein kinase; CK, compound K; mTORC1, mammalian target of rapamycin complex 1; PI3KC3, Phosphatidylinositol 3-kinase catalytic subunit type 3; RGE, Red Ginseng extract; ULK1/2, Unc-51 like autophagy activating kinase 1/2.

On the other hand, RGE inhibited autophagic flux synergistically with doxorubicin, leading to cell death in hepatocarcinoma cell lines [36]. RGE-mediated prevention of autophagic flux increased the sensitivity of the cancer cells to the doxorubicin. Autophagic flux inhibition increases cell starvation by preventing autophago- some-lysosome fusion, which is marked by a decrease in the protein level of p62 due to autophagy cargo degradation [37,38]. When chloroquine treatment was used to block lysosome function, in turn inhibiting autophagy, RGE did not upregulate the expression levels of both LC3 and p62, which indicates that RGE upregulates LC3 in the last stage of autophagy [38].

Through regulation of autophagy, KGE may reduce immunosuppressant-associated adverse effects, such as diabetes mellitus associated with tacrolimus [39,40]. Tacrolimus-induced diabetes mellitus is characterized by the accumulation of autopha- gic vacuoles, whose conversion to autophagosomes results from the activation of LC3 [39]. These autophagosomes mediate beta cell injury, leading to diabetes mellitus. RGE can modulate this activity to prevent the sustainable formation of autophagosomes, markedly by the inhibition of LC3B as an autophagosome marker after enhancement of the colocalization of LC3B and lysosome-associated membrane protein-2A during autophagosome-lysosome fusion, to enhance the autophagy clearance [40].

4. Regulation of autophagy by ginsenoside derivatives

KGE contains many derivatives and metabolites, which have divergent roles in regulating autophagy (Table 1 and Fig. 2). One ginsenoside Rb1 metabolite, 20-O-β-D-glucopyranosil-20(S)- protopanaxadiol (compound K), has been shown to play a role in malignancy by inducing autophagy and apoptosis via activation of AMP-activated protein kinase (AMPK)/c-Jun N-terminal kinase (JNK) [41]. The parallel activity in the mitogen activated protein kinase (MAPK) and AMPK signaling pathways initiates autophagy as follows: JNK phosphorylates Bcl-2 to dissociate with Beclin-1, inhibiting mTOR [42], whereas mTOR generally is inhibited by AMPK activation to initiate autophagy processes and negatively regulate the transformation and proliferation of melanoma cells [43,44]. Compound K also has been reported to induce autophagy in human colon cancer by regulating JNK and ROS generation [45] because in cancer cells, intracellular ROS concentration is found to be increased, which could mediate autophagy [46]. Activation of autophagy is also part of atherosclerosis: Macro- phages form cholesterol-laden foam cells after engulfing oxy- genized low-density lipoproteins, resulting in the formation of atherosclerotic plaques [47]. In brief, autophagy regulates lipid metabolism in macrophages through the hydrolysis of intracel- lular lipids and cholesterol efflux promotion. Moreover, auto- phagy dysfunction may lead to the abnormalities of atherosclerotic plaque formation and progression of atheroscle- rosis [48-50].

In breast cancer, metastasis can lead to drug resistance [51,52]. Mai et al [53] found an association between apoptosis and auto- phagy with ginsenoside F2 in breast cancer stem cells. They concluded that ginsenoside F2 blocks proliferative activity via activation of the intrinsic apoptotic pathway and triggers mito- chondrial dysfunction. Moreover, it also induced the recruitment of LC3, Beclin-1, and Atg-7 in the context of autophagy, controlled by the apoptosiseautophagy conjunction controller protein p53. The upregulation of p53 phosphorylation resulted in the activation of AMPK, which inhibits mTOR as an initiator of autophagy via AMPK- TSC/1/TSC2-mTOR [54,55]. In short, ginsenoside F2 in this case has a dual role including activation of apoptosis after protective auto- phagy against breast cancer stem cells.

Another ginsenoside derivative, Rh2 (G-Rh2), was shown to induce apoptosis in leukemia cells by upregulating transforming growth factor (TGF)-β expression and regulating the mitochondria signaling pathway [56,57]. Furthermore, autophagy may play another role in leukemia by upregulating the activation of LC3-II from LC3-I, together with downregulating p62, which indicates the autophagy flux in a dose-dependent manner [58]. This regulation protects the cells against apoptosis. Conclusively, to increase the efficiency of the therapeutic effect of G-Rh2 in both acute and chronic myeloid leukemia, it is necessary to inhibit autophagy to increase cancer cell death.

5. Conclusion

Studies of KGE and its components are helping to provide an understanding of the role of autophagy in the pathogenesis of certain diseases (Fig. 2). More studies are needed to determine the role of each stage of autophagy in disease. These studies would provide a better understanding of how this extract and its com- ponents could be used to treat diseases such as cancer.

Conflicts of interest

All authors declare no conflicts of interest.

Acknowledgments

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Republic of Korea (2016R1D1A1B03932512).

Appendix A. Supplementary data

Supplementary data to this article can be found online at https://doi.org/10.1016/j.jgr.2018.12.011.

References

  1. Tooze SA, Abada A, Elazar Z. Endocytosis and autophagy: exploitation or cooperation? Cold Spring Harbor Perspect Biol 2014;6:a018358. https://doi.org/10.1101/cshperspect.a018358
  2. Maria Cuervo A. Autophagy: in sickness and in health. Trends Cell Biol 2004;14:70-7. https://doi.org/10.1016/j.tcb.2003.12.002
  3. Lamb CA, Dooley HC, Tooze SA. Endocytosis and autophagy: shared machinery for degradation. BioEssays 2013;35:34-45. https://doi.org/10.1002/bies.201200130
  4. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell 2008;132:27-42. https://doi.org/10.1016/j.cell.2007.12.018
  5. Yang Z, Klionsky DJ. An overview of the molecular mechanism of autophagy. Curr Top Microbiol Immunol 2009;335:1-32. https://doi.org/10.1007/978-3-642-00302-8_1
  6. Glick D, Barth S, Macleod KF. Autophagy: cellular and molecular mechanisms. J Pathol 2010;221:3-12. https://doi.org/10.1002/path.2697
  7. Lynch-Day MA, Klionsky DJ. The Cvt pathway as a model for selective autophagy. FEBS Lett 2010;584:1359-66. https://doi.org/10.1016/j.febslet.2010.02.013
  8. Baehrecke EH. Autophagy: dual roles in life and death? Nat Rev Mol Cell Biol 2005;6:505. https://doi.org/10.1038/nrm1666
  9. Paquette M, El-Houjeiri L, Pause A. mTOR pathways in cancer and autophagy. Cancers 2018;10:18. https://doi.org/10.3390/cancers10010018
  10. Choi AMK, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med 2013;368:651-62. https://doi.org/10.1056/NEJMra1205406
  11. Randall-Demllo S, Chieppa M, ERI R. Intestinal epithelium and autophagy: partners in gut homeostasis. Front Immunol 2013;4.
  12. Das G, Shravage BV, Baehrecke EH. Regulation and function of autophagy during cell survival and cell death. Cold Spring Harbor Perspect Biol 2012;4.
  13. Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, Dawson TM, Dawson VL, El-Deiry WS, Fulda S, et al. Molecular definitions of cell death subroutines: recommendations of the nomenclature committee on cell death 2012. Cell Death Differ 2012;19:107-20. https://doi.org/10.1038/cdd.2011.96
  14. Yu L, Wan F, Dutta S, Welsh S, Liu Z, Freundt E, Baehrecke EH, Lenardo M. Autophagic programmed cell death by selective catalase degradation. Proc Natl Acad Sci U S A 2006;103:4952-7. https://doi.org/10.1073/pnas.0511288103
  15. Maiuri MC, Le Toumelin G, Criollo A, Rain J-C, Gautier F, Juin P, Tasdemir E, Pierron G, Troulinaki K, Tavernarakis N, et al. Functional and physical interaction between bcl-x(l) and a bh3-like domain in beclin-1. EMBO J 2007;26:2527-39. https://doi.org/10.1038/sj.emboj.7601689
  16. Cho C-W, Kim Y-C, Rhee YK, Lee Y-C, Kim K-T, Hong H-D. Chemical composition characteristics of Korean straight ginseng products. J Ethn Foods 2014;1:24-8. https://doi.org/10.1016/j.jef.2014.11.007
  17. Kim JH, Yi Y-S, Kim M-Y, Cho JY. Role of ginsenosides, the main active components of Panax ginseng, in inflammatory responses and diseases. J Ginseng Res 2017;41:435-43. https://doi.org/10.1016/j.jgr.2016.08.004
  18. Attele AS, Zhou Y-P, Xie J-T, Wu JA, Zhang L, Dey L, Pugh W, Rue PA, Polonsky KS, Yuan C-S. Antidiabetic effects of Panax ginseng berry extract and the identification of an effective component. Diabetes 2002;51:1851. https://doi.org/10.2337/diabetes.51.6.1851
  19. Xue P, Yao Y, Yang X-S, Feng J, Ren G-X. Improved antimicrobial effect of ginseng extract by heat transformation. J Ginseng Res 2017;41:180-7. https://doi.org/10.1016/j.jgr.2016.03.002
  20. Zheng M, Xin Y, Li Y, Xu F, Xi X, Guo H, Cui X, Cao H, Zhang X, Han C. Ginsenosides: a potential neuroprotective agent. BioMed Res Int 2018;2018:11.
  21. Ahuja A, Kim JH, Kim J-H, Yi Y-S, Cho JY. Functional role of ginseng-derived compounds in cancer. J Ginseng Res 2018;42:248-54. https://doi.org/10.1016/j.jgr.2017.04.009
  22. Xie J-T, Shao Z-H, Vanden Hoek TL, Chang W-T, Li J, Mehendale S, Wang C-Z, Hsu C-W, Becker LB, Yin J-J, et al. Antioxidant effects of ginsenoside re in cardiomyocytes. Eur J Pharmacol 2006;532:201-7. https://doi.org/10.1016/j.ejphar.2006.01.001
  23. Cho C-W, Kim Y-C, Kang J-H, Rhee YK, Choi SY, Kim K-T, Lee Y-C, Hong H-D. Characteristic study on the chemical components of Korean curved ginseng products. J Ginseng Res 2013;37:349-54. https://doi.org/10.5142/jgr.2013.37.349
  24. Leung KW, Wong AS-T. Pharmacology of ginsenosides: a literature review. Chin Med 2010;5. 20-20. https://doi.org/10.1186/1749-8546-5-20
  25. Lee K-T, Jung TW, Lee H-J, Kim S-G, Shin Y-S, Whang W-K. The antidiabetic effect of ginsenoside Rb2 via activation of AMPK. Arch Pharmacol Res 2011;34:1201. https://doi.org/10.1007/s12272-011-0719-6
  26. Saba E, Kim S-H, Kim S-D, Park S-J, Kwak D, Oh J-H, Park C-K, Rhee MH. Alleviation of diabetic complications by ginsenoside Rg3-enriched red ginseng extract in western diet-fed ldle/e mice. J Ginseng Res 2018;42:352-5. https://doi.org/10.1016/j.jgr.2017.04.004
  27. Mohanan P, Subramaniyam S, Mathiyalagan R, Yang D-C. Molecular signaling of ginsenosides Rb1, Rg1, andRrg3 and their mode of actions. J Ginseng Res 2018;42:123-32. https://doi.org/10.1016/j.jgr.2017.01.008
  28. Shen L, Haas M, Wang DQH, May A, Lo CC, Obici S, Tso P, Woods SC, Liu M. Ginsenoside Rb1 increases insulin sensitivity by activating AMP-activated protein kinase in male rats. Physiol Rep 2015;3:e12543. https://doi.org/10.14814/phy2.12543
  29. Jiang S, Ren D, Li J, Yuan G, Li H, Xu G, Han X, Du P, An L. Effects of compound K on hyperglycemia and insulin resistance in rats with type 2 diabetes mellitus. Fitoterapia 2014;95:58-64. https://doi.org/10.1016/j.fitote.2014.02.017
  30. Song Y, Zhao F, Zhang L, Du Y, Wang T, Fu F. Ginsenoside Rg1 exerts synergistic anti-inflammatory effects with low doses of glucocorticoids in vitro. Fitoterapia 2013;91:173-9. https://doi.org/10.1016/j.fitote.2013.09.001
  31. Kim MK, Kang H, Baek CW, Jung YH, Woo YC, Choi GJ, Shin HY, Kim KS. Antinociceptive and anti-inflammatory effects of ginsenoside Rf in a rat model of incisional pain. J Ginseng Res 2018;42:183-91. https://doi.org/10.1016/j.jgr.2017.02.005
  32. Lee I-S, Uh I, Kim K-S, Kim K-H, Park J, Kim Y, Jung J-H, Jung H-J, Jang H-J. Antiinflammatory effects of ginsenoside Rg3 via $NF-{\kappa}b$ pathway in a549 cells and human asthmatic lung tissue. J Immunol Res 2016;2016. 7521601-7521601.
  33. Kang J-H, Song K-H, Woo J-K, Park MH, Rhee MH, Choi C, Oh SH. Ginsenoside Rp1 from Panax ginseng exhibits anti-cancer activity by down-regulation of the IGF-1R/Akt pathway in breast cancer cells. Plant Foods Human Nutr 2011;66:298. https://doi.org/10.1007/s11130-011-0242-4
  34. Duan Z, Deng J, Dong Y, Zhu C, Li W, Fan D. Anticancer effects of ginsenoside Rk3 on non-small cell lung cancer cells: in vitro and in vivo. Food Funct 2017;8:3723-36. https://doi.org/10.1039/C7FO00385D
  35. Cha K-M, Kopalli SR, Han SY, Lee S-H, Jeong M-S, Cho JY, Han C-G, Lee S-H, Kim S-N, Kim J-C, et al. Korean red ginseng attenuates doxorubicin-induced testicular dysfunction in rats by modulating inflammatory, oxidative, and autophagy responses. J Funct Foods 2018;40:736-43. https://doi.org/10.1016/j.jff.2017.12.008
  36. Park H-H, Choi S-W, Lee GJ, Kim Y-D, Noh H-J, Oh S-J, Yoo I, Ha Y-J, Koo G-B, Hong S-S, et al. A formulated red ginseng extract inhibits autophagic flux and sensitizes to doxorubicin-induced cell death. J Ginseng Res 2017.
  37. Takanezawa Y, Nakamura R, Kojima Y, Sone Y, Uraguchi S, Kiyono M. Cytochalasin E increased the sensitivity of human lung cancer a549 cells to bortezomib via inhibition of autophagy. Biochem Biophys Res Commun 2018;498:603-8. https://doi.org/10.1016/j.bbrc.2018.03.029
  38. Dupont N, Orhon I, Bauvy C, Codogno P. Chapter four e autophagy and autophagic flux in tumor cells. In: Galluzzi L, Kroemer G, editors. Methods in enzymology. Academic Press; 2014. p. 73-88.
  39. Lim SW, Jin L, Jin J, Yang CW. Effect of exendin-4 on autophagy clearance in beta cell of rats with tacrolimus-induced diabetes mellitus. Sci Rep 2016;6:29921. https://doi.org/10.1038/srep29921
  40. Lim SW, Jin L, Luo K, Jin J, Yang CW. Ginseng extract reduces tacrolimusinduced oxidative stress by modulating autophagy in pancreatic beta cells. Lab Invest 2017;97:1271. https://doi.org/10.1038/labinvest.2017.75
  41. Kang S, Kim J-E, Song NR, Jung SK, Lee MH, Park JS, Yeom M-H, Bode AM, Dong Z, Lee KW. The ginsenoside 20-O-${\beta}$-d-glucopyranosyl-20(S)-protopanaxadiol induces autophagy and apoptosis in human melanoma via AMPK/JNK phosphorylation. PLoS One 2014;9:e104305. https://doi.org/10.1371/journal.pone.0104305
  42. Wei Y, Pattingre S, Sinha S, Bassik M, Levine B. JNK1-mediated phosphorylation of Bcl-2 regulates starvation-induced autophagy. Mol Cell 2008;30:678-88. https://doi.org/10.1016/j.molcel.2008.06.001
  43. Nikoletopoulou V, Markaki M, Palikaras K, Tavernarakis N. Crosstalk between apoptosis, necrosis and autophagy. Biochim Biophys Acta 2013;1833:3448-59. https://doi.org/10.1016/j.bbamcr.2013.06.001
  44. Vakana E, Altman JK, Platanias LC. Targeting AMPK in the treatment of malignancies. J Cell Biochem 2012;113:404-9. https://doi.org/10.1002/jcb.23369
  45. Kim AD, Kang KA, Kim HS, Kim DH, Choi YH, Lee SJ, Kim HS, Hyun JW. 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 2013;4:e750. https://doi.org/10.1038/cddis.2013.273
  46. Ling LU, Tan KB, Lin H, Chiu GNC. The role of reactive oxygen species and autophagy in safingol-induced cell death. Cell Death Dis 2011;2:e129. https://doi.org/10.1038/cddis.2011.12
  47. Shao B-z, Han B-z, Zeng Y-x, Su D-f, Liu C. The roles of macrophage autophagy in atherosclerosis. Acta Pharmacol Sin 2016;37:150-6. https://doi.org/10.1038/aps.2015.87
  48. Ouimet M, Franklin V, Mak E, Liao X, Tabas I, Marcel Yves L. Autophagy regulates cholesterol efflux from macrophage foam cells via lysosomal acid lipase. Cell Metab 2011;13:655-67. https://doi.org/10.1016/j.cmet.2011.03.023
  49. Li W, Sultana N, Siraj N, Ward LJ, Pawlik M, Levy E, Jovinge S, Bengtsson E, Yuan X-M. Autophagy dysfunction and regulatory cystatin C in macrophage death of atherosclerosis. J Cell Mol Med 2016;20:1664-72. https://doi.org/10.1111/jcmm.12859
  50. Razani B, Feng C, Coleman T, Emanuel R, Wen H, Hwang S, Ting Jenny P, Virgin Herbert W, Kastan Michael B, Semenkovich Clay F. Autophagy links inflammasomes to atherosclerotic progression. Cell Metab 2012;15:534-44. https://doi.org/10.1016/j.cmet.2012.02.011
  51. Gottesman MM. Mechanisms of cancer drug resistance. Ann Rev Med 2002;53:615-27. https://doi.org/10.1146/annurev.med.53.082901.103929
  52. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature 2001;414:105. https://doi.org/10.1038/35102167
  53. Mai TT, Moon J, Song Y, Viet PQ, Phuc PV, Lee JM, Yi T-H, Cho M, Cho SK. Ginsenoside F2 induces apoptosis accompanied by protective autophagy in breast cancer stem cells. Cancer Lett 2012;321:144-53. https://doi.org/10.1016/j.canlet.2012.01.045
  54. Balaburski GM, Hontz RD, Murphy ME. P53 and ARF: unexpected players in autophagy. Trends Cell Biol 2010;20:363-9. https://doi.org/10.1016/j.tcb.2010.02.007
  55. Lu Z, Chen C, Wu Z, Miao Y, Muhammad I, Ding L, Tian E, Hu W, Ni H, Li R, et al. A dual role of p53 in regulating colistin-induced autophagy in PC-12 cells. Front Pharmacol 2017;8. 768-768. https://doi.org/10.3389/fphar.2017.00768
  56. Xia T, Wang J-C, Xu W, Xu L-H, Lao C-H, Ye Q-X, Fang J-P. 20(S)-ginsenoside Rh2 induces apoptosis in human leukaemia reh cells through mitochondrial signaling pathways. Biol Pharmaceut Bull 2014;37:248-54. https://doi.org/10.1248/bpb.b13-00667
  57. Chung K-S, Cho S-H, Shin J-S, Kim D-H, Choi J-H, Choi SY, Rhee YK, Hong H-D, Lee K-T. Ginsenoside Rh2 induces cell cycle arrest and differentiation in human leukemia cells by upregulating $TGF-{\beta}$ expression. Carcinogenesis 2013;34:331-40. https://doi.org/10.1093/carcin/bgs341
  58. Zhuang J, Yin J, Xu C, Mu Y, Lv S. 20(S)-ginsenoside Rh2 induce the apoptosis and autophagy in U937 and K562 cells. Nutrients 2018:10.

Cited by

  1. Calycosin induces apoptosis in adenocarcinoma HT29 cells by inducing cytotoxic autophagy mediated by SIRT1/AMPK‐induced inhibition of Akt/mTOR vol.46, pp.10, 2019, https://doi.org/10.1111/1440-1681.13133
  2. The Role of Autophagy in Skin Fibroblasts, Keratinocytes, Melanocytes, and Epidermal Stem Cells vol.140, pp.9, 2019, https://doi.org/10.1016/j.jid.2019.11.023
  3. Cissus subtetragona Planch. Ameliorates Inflammatory Responses in LPS-induced Macrophages, HCl/EtOH-induced Gastritis, and LPS-induced Lung Injury via Attenuation of Src and TAK1 vol.26, pp.19, 2021, https://doi.org/10.3390/molecules26196073