Cancer Letters

Cancer Letters

Volume 230, Issue 2, 18 December 2005, Pages 228-238
Cancer Letters

Caspase-3-dependent protein kinase C delta activity is required for the progression of Ginsenoside-Rh2-induced apoptosis in SK-HEP-1 cells

https://doi.org/10.1016/j.canlet.2004.12.043Get rights and content

Abstract

Ginsenoside-Rh2 (G-Rh2) has been shown to induce apoptosis in a variety of cell types. In this study, we show that G-Rh2-induced apoptosis is accompanied by the mitochondrial release of cytochrome c and activation of caspase-3 in the human hepatoma cell line, SK-HEP-1. Furthermore, protein kinase C delta (PKCδ) activity was markedly up-regulated in a lipid activator-independent manner with kinetics similar to those of PKCδ and PARP cleavages during the apoptotic progression. Pre-treatment of cells with the caspase-3 specific inhibitor (z-DEVD-fmk) effectively prevented the G-Rh2-induced proteolytic activation of PKCδ. Moreover, rottlerin, a specific PKCδ inhibitor blocked G-Rh2-induced proapoptotic effects on the cells including the release of cytochrome c, activation of caspase-3 activity, and proteolytic cleavage and activation of PKCδ. These results suggest that G-Rh2-induced apoptosis is functionally linked to mitochondrial dysfunction and caspase-3 activity is regulated by positive feedback with PKCδ via the mitochondrial pathway.

Introduction

Apoptosis, or programmed cell death, is a physiological process of cell elimination that not only is essential for normal tissue homeostasis, but also is critical in disease states [1]. Apoptosis is characterized by distinct morphological changes such as membrane blebbing, nuclear condensation, and fragmentation of genomic DNA [2]. Most of these morphological changes are conducted by caspases, a family of aspartate-specific cysteine proteases, which play essential roles in apoptotic cells [3]. Elimination of caspase activity retards, or even prevents, apoptosis [4].

In mammalian cells, apoptosis is initiated primarily by two pathways. One involves ligation of transmembrane death receptors such as Fas and TNFα receptors by their specific ligands [5], [6]. This stimulation results in the recruitment and activation of caspase-8, which activates downstream effector caspases such as caspases-3, -6 and -7 [7]. The other pathway depends on mitochondrial dysfunction [8]. Depolarization of the mitochondria leads to the release of cytochrome c which binds to Apaf-1 to recruit and activate pro-caspase-9, leading eventually to the activation of downstream caspases [9], [10].

In earlier reports, it has been shown that protein kinase Cδ (PKCδ) is a substrate of caspase-3 and, consequently, the kinase becomes constitutively activated in the progression of apoptosis [11], [12], [13]. PKCs comprise a family of Serine/Threonine protein kinases which are involved in intracellular signaling to regulate growth, differentiation and apoptosis [14]. There are 12 known isoenzymes of PKC that are classified into three categories: the conventional PKCs (cPKCs: α, β1, β2, γ), which are calcium dependently activated in the presence of diacylglycerol (DAG) or 12-O-tetradecanoylphorbol-13-acetate (TPA), the novel PKCs (nPKCs: δ, ε, θ, η), which are calcium independently activated by DAG or TPA, and the atypical PKCs (aPKCs: ζ, λ/ι), which are calcium-independent and not activated by DAG or TPA.

The individual roles of PKCs in the regulation of apoptosis have been reported in various systems. In most systems, PKCα [15], [16], ε [17] and ι [18] act as anti-apoptotic kinases, whereas PKCθ [19], μ [20] and δ [21] act as pro-apoptotic kinases. PKCδ is activated and translocated to the plasma membrane in a number of cell types by various apoptotic stimuli, including H2O2 [22], etoposide [23], [24], ionizing radiation [25], Ara-C [26], TNFα [27], UV irradiation [28], and ceramide [29]. Additionally, it has been shown that PKCδ is cleaved in the third variable (V3) region into a catalytically active fragment by caspase-3 in response to apoptotic stimuli, such as ionizing radiation [25], DNA damaging drugs [12], [20], and oxidative stress [11]. Furthermore, overexpression of the catalytic fragment of PKCδ potentiates apoptosis induced by various apoptotic agents [11], [23]. However, other studies have shown that proteolytic cleavage of PKCδ is not required for its activation during apoptosis in CHO cells treated with H2O2 [30], LNCap cells treated with phorbol esters [31], or HaCaT cells stimulated by UV radiation [32].

Ginsenoside-Rh2 (G-Rh2) is a diol-type ginseng saponin with a dammarane skeleton that is isolated from the root of Panax ginseng, C.A. Meyer [33]. It has been shown that G-Rh2 inhibits cell growth in MCF-7 human breast cancer cells [50] and SK-HEP-1 human hepatoma cells [37], and this agent is also capable of inducing apoptosis in various cells including C6 rat glioma cells [34], SK-N-BE(2) human neuroblastoma cells [35], and A375-S2 human malignant melanoma cells [36]. G-Rh2 rapidly up-regulates JNK1 activity [38] and induces proteolytic activation of pro-caspase-3 during apoptotic progression [39]. In SK-N-BE(2) cells, activity of PKC subtypes α, β, γ and δ was progressively increased by G-Rh2 treatment, while PKCε was gradually down-regulated. But, in C6Bu-1 cells, no significant changes in PKC subtypes were observed, suggesting that G-Rh2 may induce apoptosis via different pathways in different cell types [35]. However, the underlying mechanism by which G-Rh2 induces apoptosis is not clearly understood yet.

In this study, we investigated the involvement of PKCδ in G-Rh2 induced apoptosis in SK-HEP-1 cells. Here, we show that PKCδ is proteolytically activated by caspase-3 during G-Rh2-induced apoptosis, while other Serine/Threonine kinase activities, such as PKA or PKCα, remain unaltered. Furthermore, inhibition of G-Rh2-induced PKCδ activity efficiently prevents release of cytochrome c from mitochondria and also prevents activation of caspase-3. Thus, we suggest that caspase-3 and PKCδ form a positive feedback loop for their own activation, via the mitochondrial pathway during G-Rh2-induced apoptosis.

Section snippets

Materials

Ginsenoside-Rh2 was purchased from the Korea Ginseng and Tobacco Institute (Daejeon, Korea) and stored as 20 mM stock solution in dimethyl sulfoxide (DMSO) at −20 °C. KT5720, Go6976, Rottlerin, and z-DEVD-fmk were purchased from Calbiochem (La Jolla, CA, USA). All the compounds used in cell treatments were dissolved in DMSO and added to the medium at a dilution such that the total DMSO concentration was never higher than 0.1%. Polyclonal rabbit antibodies for cytochrome c, poly(ADP-ribosyl)

Ginsenoside-Rh2 induces apoptosis via mitochondrial release of cytochrome c in SK-HEP-1 cells

To investigate the mechanism of G-Rh2-induced apoptosis, we first examined the kinetic patterns of apoptotic cell death of SK-HEP-1 cells after treatment with 20 μM G-Rh2 in serum free medium (Fig. 1A). The cells showed apoptotic morphologies such as membrane blebbing, cell rounding-up and chromatin condensations by 2 h after treatment (Fig. 1B). Moreover, the proteolytic cleavage of PARP to its 85 kDa fragment, a cellular marker of apoptosis, was detected by immunoblotting after 2 h of G-Rh2

Discussion

In this study, we show that PKCδ is proteolytically activated in a caspase-3 dependent manner during G-Rh2-induced apoptosis in SK-HEP1 cells. Moreover, PKCδ activity is also involved in activation of caspase-3 activity during apoptotic progression, resulting in a positive feedback loop. In addition, we show that G-Rh2-induced PKCδ activity is functionally associated with cytochrome c release from mitochondria.

There exist two main pathways of cell death depending on the cell type and the

Acknowledgements

This work was supported by Grant R01-2000-000-00113-0 (2001) from Korea Research Foundation and by National Research Laboratory Fund (M10104000129-02J0000-05910) from the Ministry of Science and Technology, Korea.

References (50)

  • H. Konishi et al.

    Opposing effects of protein kinase C delta and protein kinase B alpha on H(2)O(2)-induced apoptosis in CHO cells

    Biochem. Biophys. Res. Commun.

    (1999)
  • M.E. Reyland et al.

    Protein kinase C delta is essential for etoposide-induced apoptosis in salivary gland acinar cells

    J. Biol. Chem.

    (1999)
  • Y. Emoto et al.

    Activation of protein kinase Cdelta in human myeloid leukemia cells treated with 1-beta-d-arabinofuranosylcytosine

    Blood

    (1996)
  • N. Chen et al.

    Translocation of protein kinase Cepsilon and protein kinase Cdelta to membrane is required for ultraviolet B-induced activation of mitogen-activated protein kinases and apoptosis

    J. Biol. Chem.

    (1999)
  • H. Sawai et al.

    Ceramide-induced translocation of protein kinase C-delta and -epsilon to the cytosol. Implications in apoptosis

    J. Biol. Chem.

    (1997)
  • T. Fujii et al.

    Involvement of protein kinase C delta (PKCdelta) in phorbol ester-induced apoptosis in LNCaP prostate cancer cells. Lack of proteolytic cleavage of PKCdelta

    J. Biol. Chem.

    (2000)
  • M. Fukunaga et al.

    UV-induced tyrosine phosphorylation of PKC delta and promotion of apoptosis in the HaCaT cell line

    Biochem. Biophys. Res. Commun.

    (2001)
  • A.S. Attele et al.

    Ginseng pharmacology: multiple constituents and multiple actions

    Biochem. Pharmacol.

    (1999)
  • H.E. Kim et al.

    Ginsenoside RH-2 induces apoptotic cell death in rat C6 glioma via a reactive oxygen- and caspase-dependent but Bcl-X(L)-independent pathway

    Life Sci.

    (1999)
  • K.Y. Lee et al.

    Ginsenoside-Rh2 blocks the cell cycle of SK-HEP-1 cells at the G1/S boundary by selectively inducing the protein expression of p27kip1

    Cancer Lett.

    (1996)
  • Y.M. Ham et al.

    The c-Jun N-terminal kinase 1 activity is differentially regulated by specific mechanisms during apoptosis

    J. Biol. Chem.

    (2003)
  • J.A. Park et al.

    Activation of caspase-3 protease via a Bcl-2-insensitive pathway during the process of ginsenoside Rh2-induced apoptosis

    Cancer Lett.

    (1997)
  • M. Gschwendt et al.

    Rottlerin, a novel protein kinase inhibitor

    Biochem. Biophys. Res. Commun.

    (1994)
  • P.K. Majumder et al.

    Mitochondrial translocation of protein kinase C delta in phorbol ester-induced cytochrome c release and apoptosis

    J. Biol. Chem.

    (2000)
  • G. Nowak

    Protein kinase C-alpha and ERK1/2 mediate mitochondrial dysfunction, decreases in active Na+ transport, and cisplatin-induced apoptosis in renal cells

    J. Biol. Chem.

    (2002)
  • Cited by (49)

    • The ways for ginsenoside Rh2 to fight against cancer: the molecular evidences in vitro and in vivo.

      2023, Journal of Ginseng Research
      Citation Excerpt :

      However, more evidences should be provided by further in vivo studies to certify this inference. The apoptosis induced by GRh2 could be observed in a majority of common cancer models, and like other conventional anticancer drugs, caspases regulation plays a critical role in this apoptotic process (Fig. 1) [23–33]. Caspases are a conserved family of enzymes that can cleave an aspartate residue in their substrates [34].

    • Anticancer property of ginsenoside Rh2 from ginseng

      2020, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Rh2 treatment results in the disruption of lipid rafts [90] and the internalization of rafts and caveolae [99]. Other mechanisms include an increased release of mitochondrial cytochrome c [14,31,78,80] and lysosomal Cat B [77], and a reduction of the mitochondrial membrane potential [14,31,78,85,89,91]. The up-regulation/activation of the mRNA/protein expression of pro-apoptotic factors caspase-9 [19,31,33,65,70,85,91], caspase-8 [18,19,33,56,70,78,92], caspase-7 [78], caspase-4 [95], caspase-3 [14,28,29,31,34,35,56,62,70,78–80,83,85,86,91,92,101], cleaved caspase-3 [18,22,23,53,65,103], caspase-2 [92], caspase-1 [101], Bax [7,8,14,18,22,34,36,46,65,78,86,99,101,102], Bim [46,99], Bak [46,78], Bid [7], Fas [12,18,33,90,94], FasL [18], TNFR1 [33], p53 [23,33,36,66,94,101], p38 [22,39], p-p38 [22,28], p21 [23], TRAIL-RI/RII (DR4/DR5) death receptor [18,92], orphan nuclear receptor Nur77 [18], activating transcription factor 4 (ATF4) [95], activating transcription factor 2 (ATF2) [58], activator protein-1 (AP-1) [58], CCAAT/enhancer-binding protein homologous protein (CHOP) [95], Bcl-2-associated X protein [91], cleaved poly adenosine diphosphate-ribose polymerase (PARP) [103], CASP5, GADD45A, HRK, and TNFRSF9 [87] and/or the down-regulation/suppression of the mRNA/protein level of anti-apoptotic factors Bcl-2 [7,8,12,14,22,27,34,36,46,65,91,94,102], cyclin D1 [8,22,23], c-Myc [20,58], transcription factors 4 [20] and E2F1 [58], Survivin [6,65], c-IAP1, cIPA2, X-IAP [6], MMP3 [8], Bcl-Xl, Mcl-1 [46], BCLAF1, IGF1R, CD27, TNFSF10, and TP73 [87] also participate in Rh2 induced apoptosis.

    • Inhibitory mechanism of ginsenoside Rh3 on granulocyte–macrophage colony-stimulating factor expression in UV-B–irradiated murine SP-1 keratinocytes

      2020, Journal of Ginseng Research
      Citation Excerpt :

      In this regard, we suggest that ginsenoside Rh3 may be a bioactive compound targeting PKCδ and a potential agent to suppress UV-B–induced skin pigmentation. An earlier report has shown that ginsenoside Re has a protective role in methamphetamine-induced apoptosis via PKCδ inhibition in SH-SY5Y neuroblastoma cells [40]. In contrast, other groups suggested that ginsenoside Rh2 induced PKCδ activity and apoptosis in SK-HEP-1 hepatoma cells [41].

    • Antitumor and immunomodulatory effects of ginsenoside Rh2 and its octyl ester derivative in H22 tumor-bearing mice

      2017, Journal of Functional Foods
      Citation Excerpt :

      However, the in vivo antitumor activity of Rh2-O has not currently been demonstrated using animal or human studies. Historically, the antitumor activity of ginsenosides has mainly been attributed to two mechanisms: inducing tumor cells apoptosis and direct killing tumor cells (Ham et al., 2006; Kim, Kim, Han, Hyun, & Kim, 2007; Lee et al., 1996; Oh, Chun, Joo, Oh, & Lee, 2005). However, some studies had also reported that ginseng and ginsenosides could inhibit the growth of tumors by enhancing their immune response (Kenarova, Neychev, Hadjiivaova, & Petkov, 1990; Kim, Germolec, & Luster, 1990; Lee & Han, 2006; Lee et al., 2004; Wu, Ru, Chen, Ma, & Li, 2014).

    View all citing articles on Scopus
    View full text