1
|
Xiao RR, Jin L, Xie N, Luo P, Gao W, Tu P, Ai X. Establishment and large-scale validation of a three-dimensional tumor model on an array chip for anticancer drug evaluation. Front Pharmacol 2022; 13:1032975. [PMID: 36313330 PMCID: PMC9596801 DOI: 10.3389/fphar.2022.1032975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/28/2022] [Indexed: 11/29/2022] Open
Abstract
Two-dimensional (2D) tumor model has always poorly predicted drug response of animal model due to the lack of recapitulation of tumor microenvironment. Establishing a biomimetic, controllable, and cost-effective three-dimensional (3D) model and large-scale validation of its in vivo predictivity has shown promise in bridging the gap between the 2D tumor model and animal model. Here, we established a matrigel-based 3D micro-tumor model on an array chip for large-scale anticancer drug evaluation. Compared with the 2D tumor model, the 3D tumor model on the chip showed spheroid morphology, slower proliferation kinetics, and comparable reproducibility. Next, the results of the chemotherapeutic evaluation from 18 drugs against 27 cancer cell lines showed 17.6% of drug resistance on the 3D tumor model. Moreover, the evaluation results of targeted drugs showed expected sensitivity and higher specificity on the 3D tumor model compared with the 2D model. Finally, the evaluation results on the 3D tumor model were more consistent with the in vivo cell-derived xenograft model, and excluded 95% false-positive results from the 2D model. Overall, the matrigel-based 3D micro-tumor model on the array chip provides a promising tool to accelerate anticancer drug discovery.
Collapse
Affiliation(s)
- Rong-Rong Xiao
- R&D Department, Beijing Daxiang Biotech Co., Ltd., Beijing, China
| | - Lei Jin
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co., Ltd., Shanghai, China
| | - Nan Xie
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co., Ltd., Shanghai, China
| | - Piaopiao Luo
- R&D Department, Beijing Daxiang Biotech Co., Ltd., Beijing, China
| | - Wenjie Gao
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co., Ltd., Shanghai, China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaoni Ai
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- *Correspondence: Xiaoni Ai,
| |
Collapse
|
2
|
Fu L, Feng X, Wang JJ, Xun Z, Hu JD, Zhang JJ, Zhao YW, Huang ZB, Shi DQ. Efficient synthesis and evaluation of antitumor activities of novel functionalized 1,8-naphthyridine derivatives. ACS COMBINATORIAL SCIENCE 2015; 17:24-31. [PMID: 25412896 DOI: 10.1021/co500120b] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
An efficient synthesis of novel functionalized 1,8-naphthyridine and chromeno[2,3-b]quinoline derivatives via cascade reaction of 2-chloroquinoline-3-carbaldehyde and enaminones or cyclic 1,3-dicarbonyl compounds was developed. All of the 1,8-naphthyridine derivatives synthesized were evaluated for their antiproliferative properties in vitro against cancer cells and several compounds were found to have high activities.
Collapse
Affiliation(s)
- Lei Fu
- Key Laboratory of Organic
Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering
and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Xian Feng
- Key Laboratory of Organic
Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering
and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Jian-Jun Wang
- Key Laboratory of Organic
Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering
and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Zhan Xun
- Key Laboratory of Organic
Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering
and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Jun-Die Hu
- Key Laboratory of Organic
Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering
and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Juan-Juan Zhang
- Key Laboratory of Organic
Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering
and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Yan-Wei Zhao
- Key Laboratory of Organic
Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering
and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Zhi-Bin Huang
- Key Laboratory of Organic
Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering
and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Da-Qing Shi
- Key Laboratory of Organic
Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering
and Materials Science, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
3
|
Bai XG, Xu CL, Zhao SS, He HW, Wang YC, Wang JX. Synthesis and cytotoxic evaluation of alkoxylated chalcones. Molecules 2014; 19:17256-78. [PMID: 25353380 PMCID: PMC6271338 DOI: 10.3390/molecules191117256] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/26/2014] [Accepted: 10/17/2014] [Indexed: 01/03/2023] Open
Abstract
A series of chalcones a1–20 bearing a 4-OMe groups on the A-ring were initially synthesized and their anticancer activities towards HepG2 cells evaluated. Subsequently, a series of chalcones b1–42 bearing methoxy groups at the 2' and 6'-positions of the B-ring were synthesized and their anticancer activities towards five human cancer cell lines (HepG2, HeLa, MCF-7, A549 and SW1990) and two non-tumoral human cell lines evaluated. The results showed that six compounds (b6, b8, b11, b16, b18, b22, b23 and b29) displayed promising activities, with compounds b22 and b29 in particular showing higher levels of activity than etoposide against all five cancer cell lines. Compound b29 showed a promising SI value compared with both HMLE and L02 (2.1–6.5 fold in HMLE and > 33 > 103.1 fold in L02, respectively).
Collapse
Affiliation(s)
- Xiao-Guang Bai
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Chang-Liang Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Shuang-Shuang Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Hong-Wei He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yu-Cheng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Ju-Xian Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
4
|
Yang JM, Hu Y, Li Q, Yu F, Cao J, Fang D, Huang ZB, Shi DQ. Efficient and regioselective synthesis of novel functionalized dispiropyrrolidines and their cytotoxic activities. ACS COMBINATORIAL SCIENCE 2014; 16:139-45. [PMID: 24517857 DOI: 10.1021/co400096c] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
An efficient and regioselective synthesis of novel functionalized dispiropyrrolizidine derivatives via a three-component [3 + 2] cycloaddition reaction of azomethine ylides is described. This protocol has the advantages of high efficiency, mild reaction conditions, a one-pot procedure, and convenient operation. Many of these compounds were evaluated for their antiproliferative properties in vitro against cancer cells and several compounds were found to have good activities.
Collapse
Affiliation(s)
- Jin-Ming Yang
- School
of Pharmacy, Yancheng Teachers University, Yancheng 224051, P. R. China
- Key
Laboratory of Organic Synthesis of Jiangsu Province, College of Chemistry,
Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Yu Hu
- Key
Laboratory of Organic Synthesis of Jiangsu Province, College of Chemistry,
Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Qiang Li
- Hainan Chuntch Pharmaceutical Company Limited, Haikou 570216, P. R. China
| | - Fan Yu
- School
of Pharmacy, Yancheng Teachers University, Yancheng 224051, P. R. China
| | - Jian Cao
- School
of Pharmacy, Yancheng Teachers University, Yancheng 224051, P. R. China
| | - Dong Fang
- School
of Pharmacy, Yancheng Teachers University, Yancheng 224051, P. R. China
- Key
Laboratory of Organic Synthesis of Jiangsu Province, College of Chemistry,
Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Zhi-Bin Huang
- Key
Laboratory of Organic Synthesis of Jiangsu Province, College of Chemistry,
Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Da-Qing Shi
- Key
Laboratory of Organic Synthesis of Jiangsu Province, College of Chemistry,
Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
5
|
Wang Y, Zhan Y, Xu R, Shao R, Jiang J, Wang Z. Src mediates extracellular signal-regulated kinase 1/2 activation and autophagic cell death induced by cardiac glycosides in human non-small cell lung cancer cell lines. Mol Carcinog 2014; 54 Suppl 1:E26-34. [DOI: 10.1002/mc.22147] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 02/04/2014] [Accepted: 02/17/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Yan Wang
- Biochemistry Department; Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P.R. China
- Pharmacology Department; Institute of Materia Medica; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P.R. China
| | - Yuechen Zhan
- Biochemistry Department; Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P.R. China
| | - Rong Xu
- Biochemistry Department; Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P.R. China
| | - Rongguang Shao
- Biochemistry Department; Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P.R. China
| | - Jiandong Jiang
- Biochemistry Department; Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P.R. China
- Pharmacology Department; Institute of Materia Medica; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P.R. China
| | - Zhen Wang
- Biochemistry Department; Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing P.R. China
| |
Collapse
|
6
|
Özdemir F, Akalın G, Şen M, Önder NI, Işcan A, Kutlu HM, Incesu Z. Towards novel anti-tumor strategies for hepatic cancer: ɛ-viniferin in combination with vincristine displays pharmacodynamic synergy at lower doses in HepG2 cells. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2013; 18:324-34. [PMID: 24341688 DOI: 10.1089/omi.2013.0045] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hepatocellular carcinoma is the fifth most common cancer and the third leading cause of cancer-related deaths worldwide. The efficacy of novel combination treatments are increasingly evaluated with use of integrative biology research and development (R&D) strategies and methodological triangulation. We investigated the anti-tumor effect of ɛ-viniferin alone, and the putative synergy of ɛ-viniferin with vincristine on the growth of HepG2 cells in vitro. Growth inhibition and apoptosis induction were determined by MTT assay and annexin V/propidium iodide (PI), respectively. Morphological changes and DNA fragmentation were investigated under electron microscopy and by agarose gel electrophoresis, respectively. The results collectively showed that treating cells with ɛ-viniferin and vincristine significantly inhibited cell viability at lower doses as compared to each agent applied alone. IC(50) values for ɛ-viniferin and vincristine were determined as 98.3 and 52.5 μM at 24 h, respectively. IC(50) value of ɛ-viniferin in combination with vincristine was 15.8+11.25 μM (mean/SD) at 24 h. The viability of cells treated with 17.9 μM vincristine alone for 24 h was 79.62%; it reduced to 26.53% when 25 μM ɛ-viniferin was added in combination with vincristine (p<0.05). We found that combination of drugs promoted the sensitivity of cells against to vincristine treatment. The effect of combined use was in support of a synergistic pharmacodynamic effect. Moreover, low doses of the combination regimen induced phosphatidyl re-localization, morphological changes, and DNA fragmentation, and therefore caused apoptotic death. This study thus suggests that low concentrations of ɛ-viniferin and vincristine can enhance the anti-tumor effects efficiently by inducing HepG2 cell apoptosis. Further studies in other model systems are warranted with a view to potential future applications in the clinic of such combination regimens and their putative mechanism of action in the observed synergy reported here.
Collapse
Affiliation(s)
- Filiz Özdemir
- 1 Faculty of Pharmacy, Department of Biochemistry, Anadolu University , Tepebası, Eskisehir, Turkey
| | | | | | | | | | | | | |
Collapse
|
7
|
Li B, He H, Zhang S, Zhao W, Li N, Shao R. Salmonella typhimurium strain SL7207 induces apoptosis and inhibits the growth of HepG2 hepatoma cells in vitro and in vivo. Acta Pharm Sin B 2012. [DOI: 10.1016/j.apsb.2012.10.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
8
|
Chen Y, Yu D, Zhang H, He H, Zhang C, Zhao W, Shao RG. CD133(+)EpCAM(+) phenotype possesses more characteristics of tumor initiating cells in hepatocellular carcinoma Huh7 cells. Int J Biol Sci 2012; 8:992-1004. [PMID: 22904667 PMCID: PMC3421230 DOI: 10.7150/ijbs.4454] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 07/24/2012] [Indexed: 12/26/2022] Open
Abstract
Background: EpCAM or CD133 has been used as the tumor initiating cells (TICs) marker in hepatocellular carcinoma (HCC). We investigated whether cells expressing with both EpCAM and CD133 surface marker were more representative for TICs in hepatocellular carcinoma Huh7 cells. Methods: Four different phenotypes of CD133+EpCAM+, CD133+EpCAM-, CD133-EpCAM+ and CD133-EpCAM- in Huh7 cells were sorted by flow cytometry. Then cell differentiation, self-renewal, drug-resistance, spheroid formation and the levels of stem cell-related genes were detected to compare the characteristics of TICs. The ability of tumorigenicity was measured in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice to verify TICs. Results: CD133+EpCAM+ cells have many characteristics of TICs in Huh7 cells compared with CD133+EpCAM-, CD133-EpCAM+, CD133-EpCAM- cells, including enrichment in side population cells, higher differentiation capacity, increased colony-formation ability, preferential expression of stem cell-related genes, appearance of drug-resistant to some chemotherapeutics, more spheroid formation of culture cells and stronger tumorigenicity in NOD/SCID mice. Conclusion: CD133+EpCAM+ phenotype is precisely represented TICs in Huh7 cells. It might be useful for studying biology mechanism of TICs in hepatocellular carcinoma and screening new targets for cancer therapy.
Collapse
Affiliation(s)
- Yi Chen
- Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Tiantan Xili, Beijing 100050, China
| | | | | | | | | | | | | |
Collapse
|
9
|
Chen Y, Yu D, Zhang H, He H, Zhang C, Zhao W, Shao RG. CD133(+)EpCAM(+) phenotype possesses more characteristics of tumor initiating cells in hepatocellular carcinoma Huh7 cells. Int J Biol Sci 2012. [PMID: 22904667 DOI: 10.7150/ijbs.4454ijbsv08p0992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND EpCAM or CD133 has been used as the tumor initiating cells (TICs) marker in hepatocellular carcinoma (HCC). We investigated whether cells expressing with both EpCAM and CD133 surface marker were more representative for TICs in hepatocellular carcinoma Huh7 cells. METHODS Four different phenotypes of CD133(+)EpCAM(+), CD133(+)EpCAM(-), CD133(-)EpCAM(+) and CD133(-)EpCAM(-) in Huh7 cells were sorted by flow cytometry. Then cell differentiation, self-renewal, drug-resistance, spheroid formation and the levels of stem cell-related genes were detected to compare the characteristics of TICs. The ability of tumorigenicity was measured in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice to verify TICs. RESULTS CD133(+)EpCAM(+) cells have many characteristics of TICs in Huh7 cells compared with CD133(+)EpCAM(-), CD133(-)EpCAM(+), CD133(-)EpCAM(-) cells, including enrichment in side population cells, higher differentiation capacity, increased colony-formation ability, preferential expression of stem cell-related genes, appearance of drug-resistant to some chemotherapeutics, more spheroid formation of culture cells and stronger tumorigenicity in NOD/SCID mice. CONCLUSION CD133(+)EpCAM(+) phenotype is precisely represented TICs in Huh7 cells. It might be useful for studying biology mechanism of TICs in hepatocellular carcinoma and screening new targets for cancer therapy.
Collapse
Affiliation(s)
- Yi Chen
- Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Tiantan Xili, Beijing 100050, China
| | | | | | | | | | | | | |
Collapse
|
10
|
Zhang H, Zhang S, He H, Zhao W, Chen J, Shao RG. GAP161 targets and downregulates G3BP to suppress cell growth and potentiate cisplaitin-mediated cytotoxicity to colon carcinoma HCT116 cells. Cancer Sci 2012; 103:1848-56. [PMID: 22703643 DOI: 10.1111/j.1349-7006.2012.02361.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 06/07/2012] [Accepted: 06/11/2012] [Indexed: 01/04/2023] Open
Abstract
Ras-GTPase-activating protein SH3 domain-binding proteins (G3BP) are overexpressed in various human tumors and participate in several signaling pathways involved in growth, differentiation and apoptosis. G3BP interact with RasGAP (Ras-GTPase activating protein) only in growing cells and depend on Ras activation, and participate in the Ras signal pathway. Therefore, the blockage and downregulation of G3BP may be a new strategy for cancer therapy. In this report, we demonstrate that a novel peptide GAP161 blocked the functions of G3BP and markedly suppressed HCT116 cell growth through the induction of apoptosis. The peptide bound with G3BP, which interfered with the interaction of G3BP1 with RasGAP and further suppressed Ras signaling pathways. GAP161 downregulated G3BP1 and G3BP2 proteins. Similarly, the knockdown of G3BP substantially decreased the proliferation of HCT116 cells and inhibited Ras signal pathways. Furthermore, the downregulation of G3BP could enhance cisplatin-induced apoptosis and growth inhibition of HCT116 cells. We also found that GAP161 suppressed the growth of BALB/c mice bearing colon CT26 tumors and nude mice bearing HCT116 xenografts. These results suggest that downregulation of G3BP might be useful in cancer therapy and that GAP161 is a promising new therapeutic agent for cancers.
Collapse
Affiliation(s)
- Hao Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | |
Collapse
|
11
|
RasGAP-derived peptide 38GAP potentiates the cytotoxicity of cisplatin through inhibitions of Akt, ERK and NF-κB in colon carcinoma HCT116 cells. Cancer Lett 2011; 308:62-70. [PMID: 21570766 DOI: 10.1016/j.canlet.2011.04.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 04/17/2011] [Accepted: 04/18/2011] [Indexed: 01/05/2023]
Abstract
To increase the efficacy of currently used anti-cancer genotoxins, a combination use of different drugs is a potential new therapeutical tool. Here, we reported that a synthetic RasGAP-derived peptide 38GAP with RasGAP(301-326) and TAT penetration sequences could enhance the effect of chemotherapeutic agent CDDP in human colon carcinoma HCT116 cells. Our results showed that 38GAP significantly increased the CDDP-induced apoptosis in HCT116 cells. This synergistic effect was associated with abrogation of CDDP-induced G2/M arrest by down-regulations of phospho-Cdc2 and p21, and inhibitions of phospho-AKT, phospho-ERK and NF-κB. In animal models, 38GAP combined with CDDP significantly suppressed CT26 tumor growth, while 38GAP alone showed slight inhibitory effect. Our data suggest that 38GAP in combination with chemotherapeutics will become a potential therapeutic strategy for colon cancers.
Collapse
|
12
|
The benzoylurea derivative F13 inhibits cell growth, migration and invasion through inducing expression of ERK1/2-mediated RECK in fibrosarcoma HT-1080 cells. Anticancer Drugs 2010; 21:372-80. [DOI: 10.1097/cad.0b013e3283357c44] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
13
|
Zhao M, He HW, Sun HX, Ren KH, Shao RG. Dual knockdown of N-ras and epiregulin synergistically suppressed the growth of human hepatoma cells. Biochem Biophys Res Commun 2009; 387:239-44. [PMID: 19563783 DOI: 10.1016/j.bbrc.2009.06.128] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Accepted: 06/24/2009] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is a major challenge because of its resistance to conventional cytotoxic chemotherapy and radiotherapy. Multi-targeted therapy might be a new option for HCC treatment. Our previous study showed that N-ras gene was activated in HCC and was inhibited by RNA interference. In the present study, we investigated the alternation of gene expression by microarray in N-Ras-siRNA-treated HepG2 cells. The results revealed that the EREG gene, encoding epiregulin, was dramatically up-regulated in response to silence of N-ras. We speculated that the up-regulation of epiregulin was involved in the compensatory mechanism of N-ras knockdown for cell growth. Therefore, we evaluated whether dual silence of N-ras and epiregulin display a greater suppression of cell growth. The results confirmed that dual knockdown of N-ras and epiregulin synergistically inhibited cell growth. Our results also showed that dual knockdown of N-ras and epiregulin significantly induced cell arrest at G0/G1 phase. Furthermore, Western blot assay showed that dual knockdown of N-ras and epiregulin markedly reduced the phosphorylations of ERK1/2, Akt and Rb, and inhibited the expression of cyclin D1. Our findings imply that multi-targeted silence of oncogenes might be an effective treatment for HCC.
Collapse
Affiliation(s)
- Meng Zhao
- Department of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100050, China
| | | | | | | | | |
Collapse
|