1
|
Seyedmajidi M, Bijani F, Zabihi E, Bijani A, Nouri H, Nafarzadeh S. Evaluation of apoptotic effect of crocin, cisplatin, and their combination in human oral squamous cell carcinoma cell line HN5. Dent Res J (Isfahan) 2021. [DOI: 10.4103/1735-3327.324029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
2
|
Belayachi L, Aceves-Luquero C, Merghoub N, de Mattos SF, Amzazi S, Villalonga P, Bakri Y. INDUCTION OF CELL CYCLE ARREST AND APOPTOSIS BY ORMENIS ERIOLEPIS A MORROCAN ENDEMIC PLANT IN VARIOUS HUMAN CANCER CELL LINES. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES 2017; 14:356-373. [PMID: 28573252 PMCID: PMC5446462 DOI: 10.21010/ajtcam.v14i2.37] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background: Ormenis eriolepis Coss (Asteraceae) is an endemic Moroccan subspecies, traditionally named “Hellala” or “Fergoga”. It’s usually used for its hypoglycemic effect as well as for the treatment of stomacal pain. As far as we know, there is no scientific exploration of anti tumoral activity of Ormenis eriolepis extracts. Materials and Methods: In this regard, we performed a screening of organic extracts and fractions in a panel of both hematological and solid cancer cell lines, to evaluate the potential in vitro anti tumoral activity and to elucidate the respective mechanisms that may be responsible for growth arrest and cell death induction. The plant was extracted using organic solvents, and four different extracts were screened on Jurkat, Jeko-1, TK-6, LN229, SW620, U2OS, PC-3 and NIH3T3 cells. Results: Cell viability assays revealed that, the IC50 values were (11,63±5,37μg/ml) for Jurkat, (13,33±1,67μg/ml) for Jeko-1, (41,67±1,98μg/ml) for LN229 and (19,31±4,88μg/ml) for PC-3 cells upon treatment with Oe-DF and Oe-HE respectively. Both the fraction and extract exhibited no effects on TK6 and NIH3T3. Cytometry analysis accompanied by DNA damage signaling protein levels monitoring (p-H2A.X), showed that both the Dichloromethane Fraction and Hexanic extract induce DNA double stranded breaks (DSBs) accompanied by cell cycle arrest in G1 (Jurkat, Jeko -1 and LN22) and G2/M (PC-3) phases which is agreed with the caspase activity observed. Additional experiments with selective inhibitors of stress and survival pathways (JNK, MAPK, Rho, p53, and JAK3) indicated that none of these pathways was significantly involved in apoptosis induction. The bioactive compound analysis by CG/MS indicated that the major compounds in Oe-DF were: Linoleic Acid (15,89%), Podophyllotoxin (17,89%) and Quercetin (22,95%). For Oe-HE the major molecules were: Linoleic Acid (9,76%), α-curcumene (7,07%), α-bisabolol (5,49%), Campesterol (4,41%), Stigmasterol (14,08%) and β-sitosterol (7,49%). Conclusion: Our data suggest that bioactive compounds present in Ormenis eriolepis show significant anti proliferative activity inducing cell cycle arrest and cell death operating through apoptosis pathway.
Collapse
Affiliation(s)
- Lamiae Belayachi
- Biochemistry, Immunology Laboratory, Faculty of Sciences, Mohammed V-Agdal University, Rabat, Morocco.,Cancer Cell Biology Group, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Rabat - Morocco
| | - Clara Aceves-Luquero
- Cancer Cell Biology Group, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Rabat - Morocco
| | - Nawel Merghoub
- MAScIR (Moroccan Foundation for Advanced Science, Innovation & Research)- Rabat Design Center, Rabat - Morocco
| | - Silvia Fernández de Mattos
- Cancer Cell Biology Group, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Rabat - Morocco.,Departament de Biologia Fonamental, Universitat de les Illes Balears, Illes Balears, Spain, Green Biotechnology Center, Rabat - Morocco
| | - Saaîd Amzazi
- Biochemistry, Immunology Laboratory, Faculty of Sciences, Mohammed V-Agdal University, Rabat, Morocco
| | - Priam Villalonga
- Cancer Cell Biology Group, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Rabat - Morocco.,Departament de Biologia Fonamental, Universitat de les Illes Balears, Illes Balears, Spain, Green Biotechnology Center, Rabat - Morocco
| | - Youssef Bakri
- Biochemistry, Immunology Laboratory, Faculty of Sciences, Mohammed V-Agdal University, Rabat, Morocco
| |
Collapse
|
3
|
Lee GS, Lee HS, Kim SH, Suk DH, Ryu DS, Lee DS. Anti-cancer activity of the ethylacetate fraction from Orostachys japonicus for modulation of the signaling pathway in HepG2 human hepatoma cells. Food Sci Biotechnol 2013. [DOI: 10.1007/s10068-014-0037-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
4
|
Tsai YD, Chen HJ, Hsu HF, Lu K, Liang CL, Liliang PC, Wang KW, Wang HK, Wang CP, Houng JY. Luteolin inhibits proliferation of human glioblastoma cells via induction of cell cycle arrest and apoptosis. J Taiwan Inst Chem Eng 2013. [DOI: 10.1016/j.jtice.2013.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
5
|
Crocus sativus L. (saffron) stigma aqueous extract induces apoptosis in alveolar human lung cancer cells through caspase-dependent pathways activation. BIOMED RESEARCH INTERNATIONAL 2013; 2013:417928. [PMID: 24288678 PMCID: PMC3830877 DOI: 10.1155/2013/417928] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 07/10/2013] [Accepted: 07/11/2013] [Indexed: 11/18/2022]
Abstract
Worldwide, lung cancer is the most common form of cancer. Saffron has been used in folk medicine for centuries. We investigated the potential of saffron to induce cytotoxic and apoptotic effects in lung cancer cells (A549). We also examined the caspase-dependent pathways activation of saffron-induced apoptosis against the A549 cells. A549 cells were incubated with different concentrations of saffron extract; then cell morphological changes, cell viability, and apoptosis were determined by the normal invertmicroscope, MTT assay, Annexin V and propidium iodide, and flow cytometric analysis, respectively. Activated caspases were detected by treatment of saffron in lung cancer cells using fluorescein-labeled inhibitors of polycaspases. The proliferation of the A549 cells were decreased after treatment with saffron in a dose- and time-dependent manner. The percentage of apoptotic cells increased with saffron concentrations. Saffron induced morphological changes, decreased percentage of viable cells, and induced apoptosis. Saffron could induce apoptosis in the A549 cells and activate caspase pathways. The levels of caspases involved in saffron-induced apoptosis in the A549 cells indicating caspase-dependent pathway were induced by saffron. The anticancer activity of the aqueous extract of saffron could be attributed partly to its inhibition of the cell proliferation and induction of apoptosis in cancer cells through caspase-dependent pathways activation.
Collapse
|
6
|
Ethanol Extract of Dianthus chinensis L. Induces Apoptosis in Human Hepatocellular Carcinoma HepG2 Cells In Vitro. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:573527. [PMID: 22645629 PMCID: PMC3356935 DOI: 10.1155/2012/573527] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 02/20/2012] [Indexed: 11/25/2022]
Abstract
Dianthus chinensis L. is used to treat various diseases including cancer; however, the molecular mechanism by which the ethanol extract of Dianthus chinensis L. (EDCL) induces apoptosis is unknown. In this study, the apoptotic effects of EDCL were investigated in human HepG2 hepatocellular carcinoma cells. Treatment with EDCL significantly inhibited cell growth in a concentration- and time-dependent manner by inducing apoptosis. This induction was associated with chromatin condensation, activation of caspases, and cleavage of poly (ADP-ribose) polymerase protein. However, apoptosis induced by EDCL was attenuated by caspase inhibitor, indicating an important role for caspases in EDCL responses. Furthermore, EDCL did not alter the expression of bax in HepG2 cells but did selectively downregulate the expression of bcl-2 and bcl-xl, resulting in an increase in the ratio of bax:bcl-2 and bax:bcl-xl. These results support a mechanism whereby EDCL induces apoptosis through the mitochondrial pathway and caspase activation in HepG2 cells.
Collapse
|
7
|
Nho KJ, Chun JM, Kim HK. Agrimonia pilosa ethanol extract induces apoptotic cell death in HepG2 cells. JOURNAL OF ETHNOPHARMACOLOGY 2011; 138:358-363. [PMID: 21945237 DOI: 10.1016/j.jep.2011.09.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 08/23/2011] [Accepted: 09/10/2011] [Indexed: 05/31/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Agrimonia pilosa (AP) has been used as a traditional herbal medicine for treating various cancers and diseases in Asian countries. MATERIALS AND METHODS Cell viability along with caspase-3/-7, caspase-8 and caspase-9 activity were measured to detect apoptosis. The activity of the apoptotic factors bcl-2, bcl-xl, mcl-1, XIAP, BID, BIK, caspase-3, caspase-9 and PARP were measured by Western blotting. FACS analysis was used to analyze the cell cycle. RESULTS APE inhibited the proliferation of HepG2 cells. Growth inhibition was associated with increased caspase activity and sub-G1 apoptotic fractions. When we measured the affect of APE on intracellular signaling, APE stimulated the apoptotic factors bcl-2, bcl-xl, mcl-1, XIAP, BID, BIK, caspase-3, caspase-9 and PARP in HepG2 cells. CONCLUSIONS The results indicate that APE induces programmed cell death (apoptosis) in HepG2 cells and demonstrates one of the mechanisms underlying the therapeutic effects of the extract reported in previous studies.
Collapse
Affiliation(s)
- Kyoung Jin Nho
- Center of Herbal Resources Research, Korea Institute of Oriental Medicine, Daejeon 305-811, Republic of Korea
| | | | | |
Collapse
|
8
|
Chamaejasmine induces apoptosis in human lung adenocarcinoma A549 cells through a Ros-mediated mitochondrial pathway. Molecules 2011; 16:8165-80. [PMID: 21952498 PMCID: PMC6264184 DOI: 10.3390/molecules16108165] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 09/21/2011] [Accepted: 09/23/2011] [Indexed: 11/24/2022] Open
Abstract
In the present study, the anticancer activity of chamaejasmine towards A549 human lung adenocarcinoma cells was investigated. In order to explore the underlying mechanism of cell growth inhibition of chamaejasmine, cell cycle distribution, ROS generation, mitochondrial membrane potential (Δψm) disruption, and expression of cytochrome c, Bax, Bcl-2, caspase-3, caspase-9 and PARP were measured in A549 cells. Chamaejasmine inhibited the growth of A549 cells in a time and dose-dependent manner. The IC50 value was 7.72 µM after 72 h treatment. Chamaejasmine arrested the cell cycle in the G2/M phase and induced apoptosis via a ROS-mediated mitochondria-dependent pathway. Western blot analysis showed that chamaejasmine inhibited Bcl-2 expression and induced Bax expression to desintegrate the outer mitochondrial membrane and causing cytochrome c release. Mitochondrial cytochrome c release was associated with the activation of caspase-9 and caspase-3 cascade, and active-caspase-3 was involved in PARP cleavage. All of these signal transduction pathways are involved in initiating apoptosis. To the best of our knowledge, this is the first report demonstrating the cytotoxic activity of chamaejasmine towards A549 in vitro.
Collapse
|
9
|
Cholongitas E, Papatheodoridis GV, Manesis EK, Petraki K, Tiniakos D, Hadziyannis SJ. Effect of antiviral therapy on the immunohistochemical expression of bcl-xL and bax protein in patients with HBeAg-negative chronic hepatitis B. J Med Virol 2011; 83:1165-1171. [PMID: 21567420 DOI: 10.1002/jmv.21780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The effect of antiviral treatment on apoptosis in chronic hepatitis B (CHB) has not been clarified. We evaluated the hepatic immunohistochemical expression of the pro-apoptotic bax and the antiapoptotic bcl-xL protein in HBeAg-negative CHB patients before and after treatment. In our study we included 72 paired biopsies from 36 HBeAg-negative CHB patients: 29 treated (interferon-alfa: 17, adefovir: 12) and 7 untreated. Changes in expression of apoptotic proteins (D-bax, D-bcl-xL), necroinflammation and fibrosis (D-grade/D-stage) (Ishak classification) were evaluated. We found that Bax-positive compared to bax-negative biopsies had worse necroinflammation (8.2 vs. 6.7, P = 0.05) and fibrosis score (3.9 vs. 3, P = 0.036). bcl-xL-positive compared to bcl-xL-negative biopsies had lower intralobular inflammation (1.6 vs. 2.2, P = 0.03). Decreased compared to stable/increased D-bax was associated with greater improvement in necroinflammation only in treated patients (D-grade: -4.6 vs. -1.6, P = 0.05) and greater fibrosis improvement in interferon treated patients (D-stage: -0.4 vs. 0.55, P = 0.05). Increased compared to stable/decreased total apoptotic trend [D-apoptosis: (D-bax)-(D-bcl-xL)], was associated with worsening fibrosis, particularly in adefovir treated patients (D-stage: 2.3 vs. 0, P = 0.004). In the 11 patients without significant changes from 1st to 2nd biopsy, increased apoptosis was more frequent in treated than untreated cases (P = 0.046). In multivariate analysis, bax change was independently associated with change of grade (P = 0.038) and antiviral therapy (P = 0.015). In conclusions, in HBeAg-negative CHB, histological improvement after treatment is associated with decreased hepatocyte apoptosis. In patients without substantial histological changes, treatment seems to increase the apoptosis of hepatocytes, thus having a possible protective effect on hepatocarcinogenesis.
Collapse
Affiliation(s)
- Evangelos Cholongitas
- 2nd Department of Internal Medicine, Hippokration General Hospital, National University of Athens School of Medicine, Athens, Greece.
| | | | | | | | | | | |
Collapse
|
10
|
Hsu HF, Huang KH, Lu KJ, Chiou SJ, Yen JH, Chang CC, Houng JY. Typhonium blumei extract inhibits proliferation of human lung adenocarcinoma A549 cells via induction of cell cycle arrest and apoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2011; 135:492-500. [PMID: 21470575 DOI: 10.1016/j.jep.2011.03.048] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2010] [Revised: 02/17/2011] [Accepted: 03/23/2011] [Indexed: 05/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Typhonium blumei Nicolson & Sivadasan is a traditional Chinese medicinal herb endowing with detumescence, detoxification, anti-inflammation activities, and has been used as a folk prescription on anticancer in Taiwan. AIM OF THE STUDY The purpose of this study is to investigate the inhibitory effect of Typhonium blumei (Tb) extract on the viability of different cancer cells and the apoptotic effect of this extract on A549 lung cancer cells. MATERIALS AND METHODS Human A549 cell line and other cancer cell lines were treated with different concentrations of Tb extract at different time intervals. Growth inhibition was determined by MTT assay. Apoptosis was detected by cell morphologic observation, cell cycle analysis, and immunoblot analysis on the expression of protein associated with cell death. GC-MS were used to determine the chemical constituents of this extract. RESULTS The Tb extract had cytotoxicity toward A549 lung cancer cells (IC(50)=97.7 μg/ml), LNCaP prostate cancer cells (IC(50)=124.5 μg/ml) and MCF-7 breast cancer cells (IC(50)=125.8 μg/ml). Conversely, the adverse effects of Tb extract on normal embryonic lung fibroblast MRC-5 cells (IC(50)=245.5 μg/ml) and embryonic kidney fibroblast HEK293 cells (IC(50)=251.1 μg/ml) were comparatively low. Cytometric analysis results demonstrate that A549 cells were arrested at the G2/M phase by treatment with Tb extract. The extract induced A549 cell apoptosis via the mitochondrial pathway by down-regulating Bcl-2 and Bcl-xL protein expression, up-regulating Bax, Bad and Bak protein expression, and activating caspase-9 and caspase-3. Experimental results of bioactive compound analysis indicate that dibutyl phthalate, α-linolenic acid, phytol, campesterol, stigmasterol and β-sitosterol were the major bioactive ingredients of Tb extract. Although all these compounds had good anti-proliferative effects on A549 cells, campesterol (IC(50)=2.2 μM for 24h treatment) and β-sitosterol (IC(50)=1.9 μM for 24h treatment) displayed the greatest inhibitory activity. CONCLUSIONS Experimental results of this study suggest that the Tb extract exerts potential anticancer activity through the growth inhibition and the apoptosis on A549 cells.
Collapse
Affiliation(s)
- Hsia-Fen Hsu
- Department of Medical Nutrition, I-Shou University, Kaohsiung City, Taiwan
| | | | | | | | | | | | | |
Collapse
|
11
|
Lu YS, Kashida Y, Kulp SK, Wang YC, Wang D, Hung JH, Tang M, Lin ZZ, Chen TJ, Cheng AL, Chen CS. Efficacy of a novel histone deacetylase inhibitor in murine models of hepatocellular carcinoma. Hepatology 2007; 46:1119-30. [PMID: 17654699 DOI: 10.1002/hep.21804] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) is a leading cause of cancer death worldwide, yet effective therapeutic options for advanced HCC are limited. This study was aimed at assessing the antitumor effect of a novel phenylbutyrate-derived histone deacetylase (HDAC) inhibitor, OSU-HDAC42, vis-à-vis suberoylanilide hydroxamic acid (SAHA), in in vitro and in vivo models of human HCC. OSU-HDAC42 was several times more potent than SAHA in suppressing the viability of PLC5, Huh7, and Hep3B cells with submicromolar median inhibitory concentration (IC(50)) values. With respect to SAHA, OSU-HDAC42 exhibited greater apoptogenic potency, which was associated with reduced levels of the apoptotic regulators phosphorylated Akt B-cell lymphoma-xL, survivin, cellular inhibitor of apoptosis protein 1, and cellular inhibitor of apoptosis protein 2. The in vivo efficacy of OSU-HDAC42 versus SAHA was assessed in orthotopic and subcutaneous xenograft tumor models in athymic nude mice. Daily oral treatments with OSU-HDAC42 and SAHA, both at 25 mg/kg, suppressed the growth of orthotopic PLC5 tumor xenografts by 91% and 66%, respectively, and of established subcutaneous PLC5 tumor xenografts by 85% and 56%, respectively. This differential tumor suppression correlated with the modulation of intratumoral biomarkers associated with HDAC inhibition and apoptosis regulation. Moreover, the oral administration of OSU-HDAC42 at 50 mg/kg every other day markedly suppressed ectopic tumor growth in mice bearing large tumor burdens (500 mm(3)) at the start of treatment. CONCLUSION OSU-HDAC42 is a potent, orally bioavailable inhibitor of HDAC with a broad spectrum of antitumor activity that includes targets regulating multiple aspects of cancer cell survival. These results suggest that OSU-HDAC42 has clinical value in therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Yen-Shen Lu
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy and Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Anti-proliferative and pro-apoptotic effect of Smilax glabra Roxb. extract on hepatoma cell lines. Chem Biol Interact 2007; 171:1-14. [PMID: 17996228 DOI: 10.1016/j.cbi.2007.08.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2007] [Revised: 08/28/2007] [Accepted: 08/30/2007] [Indexed: 02/01/2023]
Abstract
Smilax glabra Roxb. (SGR) is the root of a traditional Chinese herb, referred to as tu fu ling in Chinese medicine. It is an inexpensive traditional Chinese medicine commonly used for the treatment of liver diseases, and a few studies have indicated that SGR has anti-hepatocarcinogenic and anti-cancer growth activities. In the current study, raw SGR plant was extracted with Accelerate Solvent Extractor, and the molecular mechanism by which S. glabra Roxb. extract (SGRE) has an anti-proliferative effect on the human hepatoma cell lines, HepG2 and Hep3B, was determined. We showed that SGRE inhibited HepG2 and Hep3B cell growth by causing cell-cycle arrest at either S phase or S/G2 transition and induced apoptosis, as evidenced by a DNA fragmentation assay. SGRE-induced apoptosis by alternation of mitochondrial transmembrane depolarization, release of mitochondrial cytochrome c, activation of caspase-3, and cleavage of poly(ADP-ribose) polymerase. The SGRE-mediated mitochondria-caspase dependent apoptotic pathway also involved activation of p38, JNK, and ERK mitogen-activated protein kinase signaling. Isometric compounds of astilbin (flavonoids) and smilagenin (saponin) have been identified as the main chemical constituents in SGRE by HPLC-MS/MS. These results have identified, for the first time, the biological activity of SGRE in HepG2 and Hep3B cells and should lead to further development of SGR for liver disease therapy.
Collapse
|
13
|
De Toni EN, Kuntzen C, Gerbes AL, Thasler WE, Sonuc N, Mucha SR, Camaj P, Bruns C, Göke B, Eichhorst ST. P60-c-src suppresses apoptosis through inhibition of caspase 8 activation in hepatoma cells, but not in primary hepatocytes. J Hepatol 2007; 46:682-91. [PMID: 17224200 DOI: 10.1016/j.jhep.2006.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2006] [Revised: 10/05/2006] [Accepted: 10/17/2006] [Indexed: 01/09/2023]
Abstract
BACKGROUND/AIMS Failure to induce apoptosis triggered by members of the death receptor family has been described in hepatocellular carcinoma (HCC) and sensitization of malignant cells to pro-apoptotic molecules such as TRAIL has been proposed as an alternative cancer therapy. Limiting to this approach are the resistance of many tumor cells to TRAIL and safety concerns about the toxicity of TRAIL in normal hepatocytes. METHODS We here explored the possibility that the protooncogene c-Src, known to be overexpressed in a variety of tumors, could be specifically responsible for the loss of response to receptor-mediated apoptosis. RESULTS Cotreatment of several hepatoma cell lines with the Src inhibitor PP2 potently sensitized these cells to TRAIL and CD95, dramatically decreasing effective doses of TRAIL to as low as 1 ng/ml. Remarkably, Src-inhibition did not synergize with TRAIL signaling in primary hepatocytes. Specific siRNAs showed that the effect was due to blockade of p60(c-Src) and occurred through increased recruitment of caspase 8. CONCLUSIONS We provide evidence that p60(c-Src) is an important and effective suppressor of receptor-mediated apoptosis in hepatoma cells but not in primary human hepatocytes. Inhibition of Src sensitizes tumor cells to apoptosis and decreases effective doses of TRAIL to therapeutic concentrations.
Collapse
Affiliation(s)
- Enrico N De Toni
- Ludwig-Maximilians-University, University Hospital Grosshadern, Department of Medicine 2, Research Lab B 5 E01 308, Marchioninistrasse 15, D-81377 Munich, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Reifenberg K, Hildt E, Lecher B, Wiese E, Nusser P, Ott S, Yamamura KI, Rutter G, Löhler J. IFNgamma expression inhibits LHBs storage disease and ground glass hepatocyte appearance, but exacerbates inflammation and apoptosis in HBV surface protein-accumulating transgenic livers. Liver Int 2006; 26:986-93. [PMID: 16953839 DOI: 10.1111/j.1478-3231.2006.01317.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND/AIMS Interferon gamma (IFNgamma) controls hepatitis B virus replication. As systemic application may cause severe adverse effects, approaches of liver-directed IFNgamma gene therapy may represent an attractive alternative for treatment of chronic viral hepatitis B and thus needs testing in vivo in suitable animal models. METHODS We therefore crossbred Alb-1HBV transgenic mice overexpressing the large HBV surface protein (LHBs) in their livers and developing LHBs storage disease and ground glass hepatocyte appearance with SAP-IFNgamma transgenic animals previously shown to exhibit constitutive hepatic IFNgamma expression, and analyzed the resulting double-transgenic offspring. RESULTS We found that IFNgamma coexpression significantly reduced hepatic LHBs expression and thereby inhibited hepatocellular LHBs storage disease and ground glass hepatocyte appearance. The beneficial antiviral IFNgamma effects as observed in Alb1-HBV SAP-IFNgamma double-transgenic livers were associated with significantly elevated serum ALT concentrations, massive mononuclear cell infiltrates, appearance of Councilman bodies, and increased alpha-PARP (poly(ADP-ribose) polymerase cleavage). CONCLUSIONS Exacerbation of hepatic necroinflammation and increased hepatocellular apoptosis rate in IFNgamma-expressing Alb1-HBV transgenic livers suggest that special precautions be taken for testing approaches of liver-specific IFNgamma expression in patients with chronic hepatitis B.
Collapse
Affiliation(s)
- Kurt Reifenberg
- Central Laboratory Animal Facility, University of Mainz, Mainz, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Pathil A, Armeanu S, Venturelli S, Mascagni P, Weiss TS, Gregor M, Lauer UM, Bitzer M. HDAC inhibitor treatment of hepatoma cells induces both TRAIL-independent apoptosis and restoration of sensitivity to TRAIL. Hepatology 2006; 43:425-34. [PMID: 16583461 DOI: 10.1002/hep.21054] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma (HCC) displays a striking resistance to chemotherapeutic drugs or innovative tumor cell apoptosis-inducing agents such as tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Recently, we found 2 histone deacetylase inhibitors (HDAC-I), valproic acid and ITF2357, exhibiting inherent therapeutic activity against HCC. In TRAIL-sensitive cancer cells, the mechanism of HDAC-I-induced cell death has been identified to be TRAIL-dependent by inducing apoptosis in an autocrine fashion. In contrast, in HCC-derived cells, a prototype of TRAIL-resistant tumor cells, we found a HDAC-I-mediated apoptosis that works independently of TRAIL and upregulation of death receptors or their cognate ligands. Interestingly, TRAIL resistance could be overcome by a combinatorial application of HDAC-I and TRAIL, increasing the fraction of apoptotic cells two- to threefold compared with HDAC-I treatment alone, whereas any premature HDAC-I withdrawal rapidly restored TRAIL resistance. Furthermore, a tumor cell-specific downregulation of the FLICE inhibitory protein (FLIP) was observed, constituting a new mechanism of TRAIL sensitivity restoration by HDAC-I. In contrast, FLIP levels in primary human hepatocytes (PHH) from different donors were upregulated by HDAC-I. Importantly, combination HDAC-I/TRAIL treatment did not induce any cytotoxicity in nonmalignant PHH. In conclusion, HDAC-I compounds, exhibiting a favorable in vivo profile and inherent activity against HCC cells, are able to selectively overcome the resistance of HCC cells toward TRAIL. Specific upregulation of intracellular FLIP protein levels in nonmalignant hepatocytes could enhance the therapeutic window for clinical applications of TRAIL, opening up a highly specific new treatment option for advanced HCC.
Collapse
Affiliation(s)
- Anita Pathil
- Department of Internal Medicine I, Medical University Clinic, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Kuo PL, Hsu YL, Cho CY, Ng LT, Kuo YH, Lin CC. Apoptotic effects of Antrodia cinnamomea fruiting bodies extract are mediated through calcium and calpain-dependent pathways in Hep 3B cells. Food Chem Toxicol 2006; 44:1316-26. [PMID: 16600460 DOI: 10.1016/j.fct.2006.02.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2005] [Revised: 01/26/2006] [Accepted: 02/18/2006] [Indexed: 11/18/2022]
Abstract
Antrodia cinnamomea is well known in Taiwan as a traditional medicine for treating cancer and inflammation. The purpose of this study was to evaluate the apoptotic effects of ethylacetate extract from A. cinnamomea (EAC) fruiting bodies in Hep 3B, a liver cancer cell line. EAC decreased cell proliferation of Hep 3B cells by inducing apoptotic cell death. EAC treatment increased the level of calcium (Ca2+) in the cytoplasm and triggered the subsequent activation of calpain and caspase-12. EAC also initiated the mitochondrial apoptotic pathway through regulation of Bcl-2 family proteins expression, release of cytochrome c, and activation of caspase-9 in Hep 3B cells. Furthermore, the mitochondrial apoptotic pathway amplified the calpain pathway by Bid and Bax interaction and Ca2+ translocation. We have therefore concluded that the molecular mechanisms during EAC-mediated proliferation inhibition in Hep 3B cells were due to: (1) apoptosis induction, (2) triggering of Ca2+/calpain pathway, (3) disruption of mitochondrial function, and (4) apoptotic signaling being amplified by cross-talk between the calpain/Bid/Bax and Ca2+/mitochondrial apoptotic pathways.
Collapse
Affiliation(s)
- Po-Lin Kuo
- Department of Biotechnology, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| | | | | | | | | | | |
Collapse
|
17
|
Armeanu S, Bitzer M, Lauer UM, Venturelli S, Pathil A, Krusch M, Kaiser S, Jobst J, Smirnow I, Wagner A, Steinle A, Salih HR. Natural killer cell-mediated lysis of hepatoma cells via specific induction of NKG2D ligands by the histone deacetylase inhibitor sodium valproate. Cancer Res 2005; 65:6321-9. [PMID: 16024634 DOI: 10.1158/0008-5472.can-04-4252] [Citation(s) in RCA: 291] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Natural killer (NK) cells as components of the innate immunity substantially contribute to antitumor immune responses. However, the tumor-associated ligands engaging activating NK cell receptors are largely unknown. An exception are the MHC class I chain-related molecules MICA and MICB and the UL16-binding proteins (ULBP) which bind to the activating immunoreceptor NKG2D expressed on cytotoxic lymphocytes. A therapeutic induction of NKG2D ligands that primes cancer cells for NK cell lysis has not yet been achieved. By microarray studies, we found evidence that treatment of human hepatocellular carcinoma cells with the histone deacetylase inhibitor (HDAC-I) sodium valproate (VPA) mediates recognition of cancer cells by cytotoxic lymphocytes via NKG2D. VPA induced transcription of MICA and MICB in hepatocellular carcinoma cells, leading to increased cell surface, soluble and total MIC protein expression. No significant changes in the expression of the NKG2D ligands ULBP1-3 were observed. The induction of MIC molecules increased lysis of hepatocellular carcinoma cells by NK cells which was abolished by addition of a blocking NKG2D antibody. Importantly, in primary human hepatocytes, VPA treatment did not induce MIC protein expression. Taken together, our data show that the HDAC-I VPA mediates specific priming of malignant cells for innate immune effector mechanisms. These results suggest the clinical evaluation of HDAC-I in solid tumors such as hepatocellular carcinoma, especially in combination with immunotherapy approaches employing adoptive NK cell transfer.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/metabolism
- Enzyme Inhibitors/pharmacology
- Histocompatibility Antigens Class I/metabolism
- Histone Deacetylase Inhibitors
- Humans
- Killer Cells, Natural/immunology
- Ligands
- Liver Neoplasms/genetics
- Liver Neoplasms/immunology
- Liver Neoplasms/metabolism
- NK Cell Lectin-Like Receptor Subfamily K
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptors, Immunologic/biosynthesis
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Receptors, Natural Killer Cell
- Up-Regulation/drug effects
- Valproic Acid/pharmacology
Collapse
Affiliation(s)
- Sorin Armeanu
- Departments of Internal Medicine I, Internal Medicine II, and Immunology, Eberhard Karls University, Tuebingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Armeanu S, Pathil A, Venturelli S, Mascagni P, Weiss TS, Göttlicher M, Gregor M, Lauer UM, Bitzer M. Apoptosis on hepatoma cells but not on primary hepatocytes by histone deacetylase inhibitors valproate and ITF2357. J Hepatol 2005; 42:210-7. [PMID: 15664246 DOI: 10.1016/j.jhep.2004.10.020] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2004] [Revised: 09/22/2004] [Accepted: 10/01/2004] [Indexed: 12/20/2022]
Abstract
BACKGROUND/AIMS Due to a particular resistance against conventional chemotherapeutics, palliative treatment of hepatocellular carcinomas (HCC) is highly ineffective. Recent demonstration of both proliferation-inhibition and apoptosis of hepatoma cells by a histone deacetylase inhibitor (HDAC-I) treatment opens up a promising new approach. However, little is known about tumor cell death mechanisms and HDAC-I influences on healthy hepatocytes. METHODS HDAC-I substances with favourable in vivo profiles, valproate (VPA) and ITF2357, were investigated on HCC cell lines and primary human hepatocytes (PHH). Histone acetylation and apoptosis-modulating proteins were investigated by western-blotting, proliferation by sulforhodamin B binding, toxicity by enzyme release, apoptosis by FACS analysis. RESULTS VPA and ITF2357 inhibited proliferation in HCC cell lines. Both substances induced considerable cellular damage in HCC-derived cells, but PHH tolerated these substances well. A downregulation of anti- and upregulation of proapoptotic factors was found. Moreover, Bcl-X(L) transfection into HCC cells abrogated apoptosis induced by both substances, indicating that modulation of intracellular pro- and anti-apoptotic proteins is a key event in VPA or ITF2357 induced tumor-cell death. CONCLUSIONS Preferential induction of cell death in HCC-derived cell lines, without toxicity in PHH, demonstrates the potential of VPA and ITF2357 to become promising new tools in the fight against HCC.
Collapse
Affiliation(s)
- Sorin Armeanu
- Department of Internal Medicine I, Medical University Clinic, Otfried-Müller-Strasse 10, D-72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|