Skip to main content
Top
Published in: Cancer Cell International 1/2017

Open Access 01-12-2017 | Primary research

Dual roles of extracellular signal-regulated kinase (ERK) in quinoline compound BPIQ-induced apoptosis and anti-migration of human non-small cell lung cancer cells

Authors: Yao Fong, Chang-Yi Wu, Kuo-Feng Chang, Bing-Hung Chen, Wan-Ju Chou, Chih-Hua Tseng, Yen-Chun Chen, Hui-Min David Wang, Yeh-Long Chen, Chien-Chih Chiu

Published in: Cancer Cell International | Issue 1/2017

Login to get access

Abstract

Background

2,9-Bis[2-(pyrrolidin-1-yl)ethoxy]-6-{4-[2-(pyrrolidin-1-yl)ethoxy] phenyl}-11H-indeno[1,2-c]quinoline-11-one (BPIQ), is a synthetic quinoline analog. A previous study showed the anti-cancer potential of BPIQ through modulating mitochondrial-mediated apoptosis. However, the effect of BPIQ on cell migration, an index of cancer metastasis, has not yet been examined. Furthermore, among signal pathways involved in stresses, the members of the mitogen-activated protein kinase (MAPK) family are crucial for regulating the survival and migration of cells. In this study, the aim was to explore further the role of MAPK members, including JNK, p38 and extracellular signal-regulated kinase (ERK) in BPIQ-induced apoptosis and anti-migration of human non-small cell lung cancer (NSCLC) cells.

Methods

Western Blot assay was performed for detecting the activation of MAPK members in NSCLC H1299 cells following BPIQ administration. Cellular proliferation was determined using a trypan blue exclusion assay. Cellular apoptosis was detected using flow cytometer-based Annexin V/propidium iodide dual staining. Cellular migration was determined using wound-healing assay and Boyden’s chamber assay. Zymography assay was performed for examining MMP-2 and -9 activities. The assessment of MAPK inhibition was performed for further validating the role of JNK, p38, and ERK in BPIQ-induced growth inhibition, apoptosis, and migration of NSCLC cells.

Results

Western Blot assay showed that BPIQ treatment upregulates the phosphorylated levels of both MAPK proteins JNK and ERK. However, only ERK inhibitor rescues BPIQ-induced growth inhibition of NSCLC H1299 cells. The results of Annexin V assay further confirmed the pro-apoptotic role of ERK in BPIQ-induced cell death of H1299 cells. The results of wound healing and Boyden chamber assays showed that sub-IC50 (sub-lethal) concentrations of BPIQ cause a significant inhibition of migration in H1299 cells accompanied with downregulating the activity of MMP-2 and -9, the motility index of cancer cells. Inhibition of ERK significantly enhances BPIQ-induced anti-migration of H1299 cells.

Conclusions

Our results suggest ERK may play dual roles in BPIQ-induced apoptosis and anti-migration, and it would be worthwhile further developing strategies for treating chemoresistant lung cancers through modulating ERK activity.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Tseng RC, Lee CC, Hsu HS, Tzao C, Wang YC. Distinct HIC1-SIRT1-p53 loop deregulation in lung squamous carcinoma and adenocarcinoma patients. Neoplasia. 2009;11(8):763–70.CrossRefPubMedPubMedCentral Tseng RC, Lee CC, Hsu HS, Tzao C, Wang YC. Distinct HIC1-SIRT1-p53 loop deregulation in lung squamous carcinoma and adenocarcinoma patients. Neoplasia. 2009;11(8):763–70.CrossRefPubMedPubMedCentral
3.
go back to reference Wagner TD, Yang GY. The role of chemotherapy and radiation in the treatment of locally advanced non-small cell lung cancer (NSCLC). Curr Drug Targets. 2010;11(1):67–73.CrossRefPubMed Wagner TD, Yang GY. The role of chemotherapy and radiation in the treatment of locally advanced non-small cell lung cancer (NSCLC). Curr Drug Targets. 2010;11(1):67–73.CrossRefPubMed
4.
go back to reference Pirker R, Minar W. Chemotherapy of advanced non-small cell lung cancer. Front Radiat Ther Oncol. 2010;42:157–63.CrossRefPubMed Pirker R, Minar W. Chemotherapy of advanced non-small cell lung cancer. Front Radiat Ther Oncol. 2010;42:157–63.CrossRefPubMed
5.
go back to reference O’Rourke N, Roque IFM, Farre Bernado N, Macbeth F. Concurrent chemoradiotherapy in non-small cell lung cancer. Cochrane Database Syst Rev. 2010;6:CD002140. O’Rourke N, Roque IFM, Farre Bernado N, Macbeth F. Concurrent chemoradiotherapy in non-small cell lung cancer. Cochrane Database Syst Rev. 2010;6:CD002140.
6.
go back to reference Ettinger DS, Akerley W, Bepler G, Blum MG, Chang A, Cheney RT, Chirieac LR, D’Amico TA, Demmy TL, Ganti AK, et al. Non-small cell lung cancer. J Natl Compr Canc Netw. 2010;8(7):740–801.PubMed Ettinger DS, Akerley W, Bepler G, Blum MG, Chang A, Cheney RT, Chirieac LR, D’Amico TA, Demmy TL, Ganti AK, et al. Non-small cell lung cancer. J Natl Compr Canc Netw. 2010;8(7):740–801.PubMed
7.
go back to reference Elbaz HA, Stueckle TA, Wang HY, O’Doherty GA, Lowry DT, Sargent LM, Wang L, Dinu CZ, Rojanasakul Y. Digitoxin and a synthetic monosaccharide analog inhibit cell viability in lung cancer cells. Toxicol Appl Pharmacol. 2012;258(1):51–60.CrossRefPubMed Elbaz HA, Stueckle TA, Wang HY, O’Doherty GA, Lowry DT, Sargent LM, Wang L, Dinu CZ, Rojanasakul Y. Digitoxin and a synthetic monosaccharide analog inhibit cell viability in lung cancer cells. Toxicol Appl Pharmacol. 2012;258(1):51–60.CrossRefPubMed
8.
go back to reference Schulze-Topphoff U, Shetty A, Varrin-Doyer M, Molnarfi N, Sagan SA, Sobel RA, Nelson PA, Zamvil SS. Laquinimod, a quinoline-3-carboxamide, induces type II myeloid cells that modulate central nervous system autoimmunity. PLoS ONE. 2012;7(3):e33797.CrossRefPubMedPubMedCentral Schulze-Topphoff U, Shetty A, Varrin-Doyer M, Molnarfi N, Sagan SA, Sobel RA, Nelson PA, Zamvil SS. Laquinimod, a quinoline-3-carboxamide, induces type II myeloid cells that modulate central nervous system autoimmunity. PLoS ONE. 2012;7(3):e33797.CrossRefPubMedPubMedCentral
9.
go back to reference Kumar S, Bawa S, Gupta H. Biological activities of quinoline derivatives. Mini Rev Med Chem. 2009;9(14):1648–54.CrossRefPubMed Kumar S, Bawa S, Gupta H. Biological activities of quinoline derivatives. Mini Rev Med Chem. 2009;9(14):1648–54.CrossRefPubMed
10.
go back to reference Tseng CH, Tzeng CC, Yang CL, Lu PJ, Chen HL, Li HY, Chuang YC, Yang CN, Chen YL. Synthesis and antiproliferative evaluation of certain indeno[1,2-c]quinoline derivatives. Part 2. J Med Chem. 2010;53(16):6164–79.CrossRefPubMed Tseng CH, Tzeng CC, Yang CL, Lu PJ, Chen HL, Li HY, Chuang YC, Yang CN, Chen YL. Synthesis and antiproliferative evaluation of certain indeno[1,2-c]quinoline derivatives. Part 2. J Med Chem. 2010;53(16):6164–79.CrossRefPubMed
11.
12.
go back to reference Tseng CH, Chen YL, Lu PJ, Yang CN, Tzeng CC. Synthesis and antiproliferative evaluation of certain indeno[1,2-c]quinoline derivatives. Bioorg Med Chem. 2008;16(6):3153–62.CrossRefPubMed Tseng CH, Chen YL, Lu PJ, Yang CN, Tzeng CC. Synthesis and antiproliferative evaluation of certain indeno[1,2-c]quinoline derivatives. Bioorg Med Chem. 2008;16(6):3153–62.CrossRefPubMed
13.
go back to reference Shenoy S, Vasania VS, Gopal M, Mehta A. 8-Methyl-4-(3-diethylaminopropylamino) pyrimido [4′,5′;4,5] thieno (2,3-b) quinoline (MDPTQ), a quinoline derivate that causes ROS-mediated apoptosis in leukemia cell lines. Toxicol Appl Pharmacol. 2007;222(1):80–8.CrossRefPubMed Shenoy S, Vasania VS, Gopal M, Mehta A. 8-Methyl-4-(3-diethylaminopropylamino) pyrimido [4′,5′;4,5] thieno (2,3-b) quinoline (MDPTQ), a quinoline derivate that causes ROS-mediated apoptosis in leukemia cell lines. Toxicol Appl Pharmacol. 2007;222(1):80–8.CrossRefPubMed
14.
go back to reference Koizumi N, Hatano E, Nitta T, Tada M, Harada N, Taura K, Ikai I, Shimahara Y. Blocking of PI3 K/Akt pathway enhances apoptosis induced by SN-38, an active form of CPT-11, in human hepatoma cells. Int J Oncol. 2005;26(5):1301–6.PubMed Koizumi N, Hatano E, Nitta T, Tada M, Harada N, Taura K, Ikai I, Shimahara Y. Blocking of PI3 K/Akt pathway enhances apoptosis induced by SN-38, an active form of CPT-11, in human hepatoma cells. Int J Oncol. 2005;26(5):1301–6.PubMed
15.
go back to reference Bruchim I, Ben-Harim Z, Piura E, Haran G, Fishman A. Analysis of two topotecan treatment schedules in patients with recurrent ovarian cancer. J Chemother. 2016;28:129–34.CrossRefPubMed Bruchim I, Ben-Harim Z, Piura E, Haran G, Fishman A. Analysis of two topotecan treatment schedules in patients with recurrent ovarian cancer. J Chemother. 2016;28:129–34.CrossRefPubMed
16.
go back to reference Park SH, Cho EK, Kim Y, Kyung SY, An CH, Lee SP, Park JW, Jeong SH, Lee JI, Choi SJ, et al. Salvage treatment with topotecan in patients with irinotecan-refractory small cell lung cancer. Cancer Chemother Pharmacol. 2008;62(6):1009–14.CrossRefPubMed Park SH, Cho EK, Kim Y, Kyung SY, An CH, Lee SP, Park JW, Jeong SH, Lee JI, Choi SJ, et al. Salvage treatment with topotecan in patients with irinotecan-refractory small cell lung cancer. Cancer Chemother Pharmacol. 2008;62(6):1009–14.CrossRefPubMed
18.
go back to reference Pommier Y. Topoisomerase I inhibitors: camptothecins and beyond. Nat Rev Cancer. 2006;6(10):789–802.CrossRefPubMed Pommier Y. Topoisomerase I inhibitors: camptothecins and beyond. Nat Rev Cancer. 2006;6(10):789–802.CrossRefPubMed
19.
go back to reference Chiu CC, Chou HL, Chen BH, Chang KF, Tseng CH, Fong Y, Fu TF, Chang HW, Wu CY, Tsai EM, et al. BPIQ, a novel synthetic quinoline derivative, inhibits growth and induces mitochondrial apoptosis of lung cancer cells in vitro and in zebrafish xenograft model. BMC Cancer. 2015;15:962.CrossRefPubMedPubMedCentral Chiu CC, Chou HL, Chen BH, Chang KF, Tseng CH, Fong Y, Fu TF, Chang HW, Wu CY, Tsai EM, et al. BPIQ, a novel synthetic quinoline derivative, inhibits growth and induces mitochondrial apoptosis of lung cancer cells in vitro and in zebrafish xenograft model. BMC Cancer. 2015;15:962.CrossRefPubMedPubMedCentral
20.
go back to reference Cheng KC, Hung CT, Chen KJ, Wu WC, Suen JL, Chang CH, Lu CY, Tseng CH, Chen YL, Chiu CC. Quinoline-based compound BPIQ exerts anti-proliferative effects on human retinoblastoma cells via modulating intracellular reactive oxygen species. Arch Immunol Ther Exp (Warsz). 2016;64(2):139–47.CrossRef Cheng KC, Hung CT, Chen KJ, Wu WC, Suen JL, Chang CH, Lu CY, Tseng CH, Chen YL, Chiu CC. Quinoline-based compound BPIQ exerts anti-proliferative effects on human retinoblastoma cells via modulating intracellular reactive oxygen species. Arch Immunol Ther Exp (Warsz). 2016;64(2):139–47.CrossRef
21.
go back to reference Coulthard LR, White DE, Jones DL, McDermott MF, Burchill SA. p38(MAPK): stress responses from molecular mechanisms to therapeutics. Trends Mol Med. 2009;15(8):369–79.CrossRefPubMedPubMedCentral Coulthard LR, White DE, Jones DL, McDermott MF, Burchill SA. p38(MAPK): stress responses from molecular mechanisms to therapeutics. Trends Mol Med. 2009;15(8):369–79.CrossRefPubMedPubMedCentral
22.
go back to reference Michaelidis B, Hatzikamari M, Antoniou V, Anestis A, Lazou A. Stress activated protein kinases, JNKs and p38 MAPK, are differentially activated in ganglia and heart of land snail Helix lucorum (L.) during seasonal hibernation and arousal. Comp Biochem Physiol A: Mol Integr Physiol. 2009;153(2):149–53.CrossRef Michaelidis B, Hatzikamari M, Antoniou V, Anestis A, Lazou A. Stress activated protein kinases, JNKs and p38 MAPK, are differentially activated in ganglia and heart of land snail Helix lucorum (L.) during seasonal hibernation and arousal. Comp Biochem Physiol A: Mol Integr Physiol. 2009;153(2):149–53.CrossRef
23.
go back to reference Chen K, Chu BZ, Liu F, Li B, Gao CM, Li LL, Sun QS, Shen ZF, Jiang YY. New benzimidazole acridine derivative induces human colon cancer cell apoptosis in vitro via the ROS-JNK signaling pathway. Acta Pharmacol Sin. 2015;36(9):1074–84.CrossRefPubMedPubMedCentral Chen K, Chu BZ, Liu F, Li B, Gao CM, Li LL, Sun QS, Shen ZF, Jiang YY. New benzimidazole acridine derivative induces human colon cancer cell apoptosis in vitro via the ROS-JNK signaling pathway. Acta Pharmacol Sin. 2015;36(9):1074–84.CrossRefPubMedPubMedCentral
24.
go back to reference Schreck I, Grico N, Hansjosten I, Marquardt C, Bormann S, Seidel A, Kvietkova DL, Pieniazek D, Segerback D, Diabate S, et al. The nucleotide excision repair protein XPC is essential for bulky DNA adducts to promote interleukin-6 expression via the activation of p38-SAPK. Oncogene. 2016;35(7):908–18.CrossRefPubMed Schreck I, Grico N, Hansjosten I, Marquardt C, Bormann S, Seidel A, Kvietkova DL, Pieniazek D, Segerback D, Diabate S, et al. The nucleotide excision repair protein XPC is essential for bulky DNA adducts to promote interleukin-6 expression via the activation of p38-SAPK. Oncogene. 2016;35(7):908–18.CrossRefPubMed
25.
go back to reference Wei Y, An Z, Zou Z, Sumpter R, Su M, Zang X, Sinha S, Gaestel M, Levine B. The stress-responsive kinases MAPKAPK2/MAPKAPK3 activate starvation-induced autophagy through Beclin 1 phosphorylation. eLife. 2015;4:05289. doi:10.7554/eLife.05289. Wei Y, An Z, Zou Z, Sumpter R, Su M, Zang X, Sinha S, Gaestel M, Levine B. The stress-responsive kinases MAPKAPK2/MAPKAPK3 activate starvation-induced autophagy through Beclin 1 phosphorylation. eLife. 2015;4:05289. doi:10.​7554/​eLife.​05289.
26.
go back to reference Choi MS, Jeong HJ, Kang TH, Shin HM, Oh ST, Choi Y, Jeon S. Meso-dihydroguaiaretic acid induces apoptosis and inhibits cell migration via p38 activation and EGFR/Src/intergrin beta3 downregulation in breast cancer cells. Life Sci. 2015;141:81–9.CrossRefPubMed Choi MS, Jeong HJ, Kang TH, Shin HM, Oh ST, Choi Y, Jeon S. Meso-dihydroguaiaretic acid induces apoptosis and inhibits cell migration via p38 activation and EGFR/Src/intergrin beta3 downregulation in breast cancer cells. Life Sci. 2015;141:81–9.CrossRefPubMed
27.
go back to reference Ong JY, Yong PV, Lim YM, Ho AS. 2-Methoxy-1,4-naphthoquinone (MNQ) induces apoptosis of A549 lung adenocarcinoma cells via oxidation-triggered JNK and p38 MAPK signaling pathways. Life Sci. 2015;135:158–64.CrossRefPubMed Ong JY, Yong PV, Lim YM, Ho AS. 2-Methoxy-1,4-naphthoquinone (MNQ) induces apoptosis of A549 lung adenocarcinoma cells via oxidation-triggered JNK and p38 MAPK signaling pathways. Life Sci. 2015;135:158–64.CrossRefPubMed
29.
go back to reference Dhillon AS, Hagan S, Rath O, Kolch W. MAP kinase signalling pathways in cancer. Oncogene. 2007;26(22):3279–90.CrossRefPubMed Dhillon AS, Hagan S, Rath O, Kolch W. MAP kinase signalling pathways in cancer. Oncogene. 2007;26(22):3279–90.CrossRefPubMed
30.
go back to reference McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Wong EW, Chang F, Lehmann B, Terrian DM, Milella M, Tafuri A, et al. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta. 2007;1773(8):1263–84.CrossRefPubMed McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Wong EW, Chang F, Lehmann B, Terrian DM, Milella M, Tafuri A, et al. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta. 2007;1773(8):1263–84.CrossRefPubMed
31.
go back to reference Yamakawa K, Yokohira M, Nakano Y, Kishi S, Kanie S, Imaida K. Activation of MEK1/2-ERK1/2 signaling during NNK-induced lung carcinogenesis in female A/J mice. Cancer Med. 2016. doi:10.1002/cam4.652. Yamakawa K, Yokohira M, Nakano Y, Kishi S, Kanie S, Imaida K. Activation of MEK1/2-ERK1/2 signaling during NNK-induced lung carcinogenesis in female A/J mice. Cancer Med. 2016. doi:10.​1002/​cam4.​652.
32.
go back to reference Qu Y, Wu X, Yin Y, Yang Y, Ma D, Li H. Antitumor activity of selective MEK1/2 inhibitor AZD6244 in combination with PI3K/mTOR inhibitor BEZ235 in gefitinib-resistant NSCLC xenograft models. J Exp Clin Cancer Res. 2014;33:52.CrossRefPubMedPubMedCentral Qu Y, Wu X, Yin Y, Yang Y, Ma D, Li H. Antitumor activity of selective MEK1/2 inhibitor AZD6244 in combination with PI3K/mTOR inhibitor BEZ235 in gefitinib-resistant NSCLC xenograft models. J Exp Clin Cancer Res. 2014;33:52.CrossRefPubMedPubMedCentral
33.
go back to reference Cagnol S, Chambard JC. ERK and cell death: mechanisms of ERK-induced cell death—apoptosis, autophagy and senescence. FEBS J. 2010;277(1):2–21.CrossRefPubMed Cagnol S, Chambard JC. ERK and cell death: mechanisms of ERK-induced cell death—apoptosis, autophagy and senescence. FEBS J. 2010;277(1):2–21.CrossRefPubMed
34.
go back to reference Yahiro K, Hirayama T, Moss J, Noda M. Helicobacter pylori VacA toxin causes cell death by inducing accumulation of cytoplasmic connexin 43. Cell Death Dis. 2015;6:e1971.CrossRefPubMedPubMedCentral Yahiro K, Hirayama T, Moss J, Noda M. Helicobacter pylori VacA toxin causes cell death by inducing accumulation of cytoplasmic connexin 43. Cell Death Dis. 2015;6:e1971.CrossRefPubMedPubMedCentral
35.
go back to reference Kole L, Sarkar M, Deb A, Giri B. Pioglitazone, an anti-diabetic drug requires sustained MAPK activation for its anti-tumor activity in MCF7 breast cancer cells, independent of PPAR-gamma pathway. Pharmacol Rep. 2016;68(1):144–54.CrossRefPubMed Kole L, Sarkar M, Deb A, Giri B. Pioglitazone, an anti-diabetic drug requires sustained MAPK activation for its anti-tumor activity in MCF7 breast cancer cells, independent of PPAR-gamma pathway. Pharmacol Rep. 2016;68(1):144–54.CrossRefPubMed
36.
go back to reference Liu Y, Yang Y, Ye YC, Shi QF, Chai K, Tashiro S, Onodera S, Ikejima T. Activation of ERK-p53 and ERK-mediated phosphorylation of Bcl-2 are involved in autophagic cell death induced by the c-Met inhibitor SU11274 in human lung cancer A549 cells. J Pharmacol Sci. 2012;118(4):423–32.CrossRefPubMed Liu Y, Yang Y, Ye YC, Shi QF, Chai K, Tashiro S, Onodera S, Ikejima T. Activation of ERK-p53 and ERK-mediated phosphorylation of Bcl-2 are involved in autophagic cell death induced by the c-Met inhibitor SU11274 in human lung cancer A549 cells. J Pharmacol Sci. 2012;118(4):423–32.CrossRefPubMed
37.
go back to reference Kim YH, Lee DH, Jeong JH, Guo ZS, Lee YJ. Quercetin augments TRAIL-induced apoptotic death: involvement of the ERK signal transduction pathway. Biochem Pharmacol. 2008;75(10):1946–58.CrossRefPubMedPubMedCentral Kim YH, Lee DH, Jeong JH, Guo ZS, Lee YJ. Quercetin augments TRAIL-induced apoptotic death: involvement of the ERK signal transduction pathway. Biochem Pharmacol. 2008;75(10):1946–58.CrossRefPubMedPubMedCentral
38.
go back to reference Rieber M, Rieber MS. Signalling responses linked to betulinic acid-induced apoptosis are antagonized by MEK inhibitor U0126 in adherent or 3D spheroid melanoma irrespective of p53 status. Int J Cancer. 2006;118(5):1135–43.CrossRefPubMed Rieber M, Rieber MS. Signalling responses linked to betulinic acid-induced apoptosis are antagonized by MEK inhibitor U0126 in adherent or 3D spheroid melanoma irrespective of p53 status. Int J Cancer. 2006;118(5):1135–43.CrossRefPubMed
39.
go back to reference Tewari R, Sharma V, Koul N, Sen E. Involvement of miltefosine-mediated ERK activation in glioma cell apoptosis through Fas regulation. J Neurochem. 2008;107(3):616–27.CrossRefPubMed Tewari R, Sharma V, Koul N, Sen E. Involvement of miltefosine-mediated ERK activation in glioma cell apoptosis through Fas regulation. J Neurochem. 2008;107(3):616–27.CrossRefPubMed
40.
go back to reference Li NY, Weber CE, Wai PY, Cuevas BD, Zhang J, Kuo PC, Mi Z. An MAPK-dependent pathway induces epithelial-mesenchymal transition via Twist activation in human breast cancer cell lines. Surgery. 2013;154(2):404–10.CrossRefPubMed Li NY, Weber CE, Wai PY, Cuevas BD, Zhang J, Kuo PC, Mi Z. An MAPK-dependent pathway induces epithelial-mesenchymal transition via Twist activation in human breast cancer cell lines. Surgery. 2013;154(2):404–10.CrossRefPubMed
41.
go back to reference Baquero P, Jimenez-Mora E, Santos A, Lasa M, Chiloeches A. TGFbeta induces epithelial-mesenchymal transition of thyroid cancer cells by both the BRAF/MEK/ERK and Src/FAK pathways. Mol Carcinog. 2015. doi:10.1002/mc.22415.PubMed Baquero P, Jimenez-Mora E, Santos A, Lasa M, Chiloeches A. TGFbeta induces epithelial-mesenchymal transition of thyroid cancer cells by both the BRAF/MEK/ERK and Src/FAK pathways. Mol Carcinog. 2015. doi:10.​1002/​mc.​22415.PubMed
42.
go back to reference Sun X, Lin L, Chen Y, Liu T, Liu R, Wang Z, Mou K, Xu J, Li B, Song H. Nitidine chloride inhibits ovarian cancer cell migration and invasion by suppressing MMP-2/9 production via the ERK signaling pathway. Mol Med Rep. 2016;13(4):3161–8.PubMed Sun X, Lin L, Chen Y, Liu T, Liu R, Wang Z, Mou K, Xu J, Li B, Song H. Nitidine chloride inhibits ovarian cancer cell migration and invasion by suppressing MMP-2/9 production via the ERK signaling pathway. Mol Med Rep. 2016;13(4):3161–8.PubMed
43.
go back to reference Tsai PC, Chu CL, Chiu CC, Chang LS, Lin SR. Inhibition of Src activation with cardiotoxin III blocks migration and invasion of MDA-MB-231 cells. Toxicon. 2013;74:56–67.CrossRefPubMed Tsai PC, Chu CL, Chiu CC, Chang LS, Lin SR. Inhibition of Src activation with cardiotoxin III blocks migration and invasion of MDA-MB-231 cells. Toxicon. 2013;74:56–67.CrossRefPubMed
44.
go back to reference Tsai PC, Hsieh CY, Chiu CC, Wang CK, Chang LS, Lin SR. Cardiotoxin III suppresses MDA-MB-231 cell metastasis through the inhibition of EGF/EGFR-mediated signaling pathway. Toxicon. 2012;60(5):734–43.CrossRefPubMed Tsai PC, Hsieh CY, Chiu CC, Wang CK, Chang LS, Lin SR. Cardiotoxin III suppresses MDA-MB-231 cell metastasis through the inhibition of EGF/EGFR-mediated signaling pathway. Toxicon. 2012;60(5):734–43.CrossRefPubMed
45.
go back to reference Wirth M, Berthold E, Grashoff M, Pfutzner H, Schubert U, Hauser H. Detection of mycoplasma contaminations by the polymerase chain reaction. Cytotechnology. 1994;16(2):67–77.CrossRefPubMed Wirth M, Berthold E, Grashoff M, Pfutzner H, Schubert U, Hauser H. Detection of mycoplasma contaminations by the polymerase chain reaction. Cytotechnology. 1994;16(2):67–77.CrossRefPubMed
46.
go back to reference Chiu CC, Chen JY, Lin KL, Huang CJ, Lee JC, Chen BH, Chen WY, Lo YH, Chen YL, Tseng CH, et al. p38 MAPK and NF-kappaB pathways are involved in naphtho[1,2-b] furan-4,5-dione induced anti-proliferation and apoptosis of human hepatoma cells. Cancer Lett. 2010;295(1):92–9.CrossRefPubMed Chiu CC, Chen JY, Lin KL, Huang CJ, Lee JC, Chen BH, Chen WY, Lo YH, Chen YL, Tseng CH, et al. p38 MAPK and NF-kappaB pathways are involved in naphtho[1,2-b] furan-4,5-dione induced anti-proliferation and apoptosis of human hepatoma cells. Cancer Lett. 2010;295(1):92–9.CrossRefPubMed
47.
go back to reference Geback T, Schulz MM, Koumoutsakos P, Detmar M. TScratch: a novel and simple software tool for automated analysis of monolayer wound healing assays. Biotechniques. 2009;46(4):265–74.PubMed Geback T, Schulz MM, Koumoutsakos P, Detmar M. TScratch: a novel and simple software tool for automated analysis of monolayer wound healing assays. Biotechniques. 2009;46(4):265–74.PubMed
48.
go back to reference Chiu CC, Liu PL, Huang KJ, Wang HM, Chang KF, Chou CK, Chang FR, Chong IW, Fang K, Chen JS, et al. Goniothalamin inhibits growth of human lung cancer cells through DNA damage, apoptosis, and reduced migration ability. J Agric Food Chem. 2011;59(8):4288–93.CrossRefPubMed Chiu CC, Liu PL, Huang KJ, Wang HM, Chang KF, Chou CK, Chang FR, Chong IW, Fang K, Chen JS, et al. Goniothalamin inhibits growth of human lung cancer cells through DNA damage, apoptosis, and reduced migration ability. J Agric Food Chem. 2011;59(8):4288–93.CrossRefPubMed
49.
go back to reference Li L, Davie JR. The role of Sp1 and Sp3 in normal and cancer cell biology. Ann Anat. 2010;192(5):275–83.CrossRefPubMed Li L, Davie JR. The role of Sp1 and Sp3 in normal and cancer cell biology. Ann Anat. 2010;192(5):275–83.CrossRefPubMed
50.
go back to reference Qin H, Sun Y, Benveniste EN. The transcription factors Sp1, Sp3, and AP-2 are required for constitutive matrix metalloproteinase-2 gene expression in astroglioma cells. J Biol Chem. 1999;274(41):29130–7.CrossRefPubMed Qin H, Sun Y, Benveniste EN. The transcription factors Sp1, Sp3, and AP-2 are required for constitutive matrix metalloproteinase-2 gene expression in astroglioma cells. J Biol Chem. 1999;274(41):29130–7.CrossRefPubMed
51.
go back to reference Bae IH, Park MJ, Yoon SH, Kang SW, Lee SS, Choi KM, Um HD. Bcl-w promotes gastric cancer cell invasion by inducing matrix metalloproteinase-2 expression via phosphoinositide 3-kinase, Akt, and Sp1. Cancer Res. 2006;66(10):4991–5.CrossRefPubMed Bae IH, Park MJ, Yoon SH, Kang SW, Lee SS, Choi KM, Um HD. Bcl-w promotes gastric cancer cell invasion by inducing matrix metalloproteinase-2 expression via phosphoinositide 3-kinase, Akt, and Sp1. Cancer Res. 2006;66(10):4991–5.CrossRefPubMed
52.
go back to reference Zhao S, Wu J, Zheng F, Tang Q, Yang L, Li L, Wu W, Hann SS. Beta-elemene inhibited expression of DNA methyltransferase 1 through activation of ERK1/2 and AMPKalpha signalling pathways in human lung cancer cells: the role of Sp1. J Cell Mol Med. 2015;19(3):630–41.CrossRefPubMedPubMedCentral Zhao S, Wu J, Zheng F, Tang Q, Yang L, Li L, Wu W, Hann SS. Beta-elemene inhibited expression of DNA methyltransferase 1 through activation of ERK1/2 and AMPKalpha signalling pathways in human lung cancer cells: the role of Sp1. J Cell Mol Med. 2015;19(3):630–41.CrossRefPubMedPubMedCentral
53.
go back to reference Nam EH, Lee Y, Zhao XF, Park YK, Lee JW, Kim S. ZEB2-Sp1 cooperation induces invasion by upregulating cadherin-11 and integrin alpha5 expression. Carcinogenesis. 2014;35(2):302–14.CrossRefPubMed Nam EH, Lee Y, Zhao XF, Park YK, Lee JW, Kim S. ZEB2-Sp1 cooperation induces invasion by upregulating cadherin-11 and integrin alpha5 expression. Carcinogenesis. 2014;35(2):302–14.CrossRefPubMed
54.
go back to reference Hung WC, Chang HC. Indole-3-carbinol inhibits Sp1-induced matrix metalloproteinase-2 expression to attenuate migration and invasion of breast cancer cells. J Agric Food Chem. 2009;57(1):76–82.CrossRefPubMed Hung WC, Chang HC. Indole-3-carbinol inhibits Sp1-induced matrix metalloproteinase-2 expression to attenuate migration and invasion of breast cancer cells. J Agric Food Chem. 2009;57(1):76–82.CrossRefPubMed
55.
go back to reference Xin D, Rendon BE, Zhao M, Winner M, McGhee Coleman A, Mitchell RA. The MIF homologue D-dopachrome tautomerase promotes COX-2 expression through beta-catenin-dependent and -independent mechanisms. Mol Cancer Res. 2010;8(12):1601–9.CrossRefPubMedPubMedCentral Xin D, Rendon BE, Zhao M, Winner M, McGhee Coleman A, Mitchell RA. The MIF homologue D-dopachrome tautomerase promotes COX-2 expression through beta-catenin-dependent and -independent mechanisms. Mol Cancer Res. 2010;8(12):1601–9.CrossRefPubMedPubMedCentral
56.
go back to reference Yue X, Lan F, Yang W, Yang Y, Han L, Zhang A, Liu J, Zeng H, Jiang T, Pu P, et al. Interruption of beta-catenin suppresses the EGFR pathway by blocking multiple oncogenic targets in human glioma cells. Brain Res. 2010;1366:27–37.CrossRefPubMed Yue X, Lan F, Yang W, Yang Y, Han L, Zhang A, Liu J, Zeng H, Jiang T, Pu P, et al. Interruption of beta-catenin suppresses the EGFR pathway by blocking multiple oncogenic targets in human glioma cells. Brain Res. 2010;1366:27–37.CrossRefPubMed
57.
go back to reference Estrada Y, Dong J, Ossowski L. Positive crosstalk between ERK and p38 in melanoma stimulates migration and in vivo proliferation. Pigment Cell Melanoma Res. 2009;22(1):66–76.CrossRefPubMed Estrada Y, Dong J, Ossowski L. Positive crosstalk between ERK and p38 in melanoma stimulates migration and in vivo proliferation. Pigment Cell Melanoma Res. 2009;22(1):66–76.CrossRefPubMed
58.
go back to reference Hwang YP, Yun HJ, Choi JH, Han EH, Kim HG, Song GY, Kwon KI, Jeong TC, Jeong HG. Suppression of EGF-induced tumor cell migration and matrix metalloproteinase-9 expression by capsaicin via the inhibition of EGFR-mediated FAK/Akt, PKC/Raf/ERK, p38 MAPK, and AP-1 signaling. Mol Nutr Food Res. 2011;55(4):594–605.CrossRefPubMed Hwang YP, Yun HJ, Choi JH, Han EH, Kim HG, Song GY, Kwon KI, Jeong TC, Jeong HG. Suppression of EGF-induced tumor cell migration and matrix metalloproteinase-9 expression by capsaicin via the inhibition of EGFR-mediated FAK/Akt, PKC/Raf/ERK, p38 MAPK, and AP-1 signaling. Mol Nutr Food Res. 2011;55(4):594–605.CrossRefPubMed
59.
go back to reference Watson JL, Greenshields A, Hill R, Hilchie A, Lee PW, Giacomantonio CA, Hoskin DW. Curcumin-induced apoptosis in ovarian carcinoma cells is p53-independent and involves p38 mitogen-activated protein kinase activation and downregulation of Bcl-2 and survivin expression and Akt signaling. Mol Carcinog. 2010;49(1):13–24.PubMed Watson JL, Greenshields A, Hill R, Hilchie A, Lee PW, Giacomantonio CA, Hoskin DW. Curcumin-induced apoptosis in ovarian carcinoma cells is p53-independent and involves p38 mitogen-activated protein kinase activation and downregulation of Bcl-2 and survivin expression and Akt signaling. Mol Carcinog. 2010;49(1):13–24.PubMed
60.
go back to reference Rosini P, De Chiara G, Lucibello M, Garaci E, Cozzolino F, Torcia M. NGF withdrawal induces apoptosis in CESS B cell line through p38 MAPK activation and Bcl-2 phosphorylation. Biochem Biophys Res Commun. 2000;278(3):753–9.CrossRefPubMed Rosini P, De Chiara G, Lucibello M, Garaci E, Cozzolino F, Torcia M. NGF withdrawal induces apoptosis in CESS B cell line through p38 MAPK activation and Bcl-2 phosphorylation. Biochem Biophys Res Commun. 2000;278(3):753–9.CrossRefPubMed
61.
go back to reference Bruzzese F, Pucci B, Milone MR, Ciardiello C, Franco R, Chianese MI, Rocco M, Di Gennaro E, Leone A, Luciano A, et al. Panobinostat synergizes with zoledronic acid in prostate cancer and multiple myeloma models by increasing ROS and modulating mevalonate and p38-MAPK pathways. Cell Death Dis. 2013;4:e878.CrossRefPubMedPubMedCentral Bruzzese F, Pucci B, Milone MR, Ciardiello C, Franco R, Chianese MI, Rocco M, Di Gennaro E, Leone A, Luciano A, et al. Panobinostat synergizes with zoledronic acid in prostate cancer and multiple myeloma models by increasing ROS and modulating mevalonate and p38-MAPK pathways. Cell Death Dis. 2013;4:e878.CrossRefPubMedPubMedCentral
62.
go back to reference Milone MR, Pucci B, Bruzzese F, Carbone C, Piro G, Costantini S, Capone F, Leone A, Di Gennaro E, Caraglia M, et al. Acquired resistance to zoledronic acid and the parallel acquisition of an aggressive phenotype are mediated by p38-MAP kinase activation in prostate cancer cells. Cell Death Dis. 2013;4:e641.CrossRefPubMedPubMedCentral Milone MR, Pucci B, Bruzzese F, Carbone C, Piro G, Costantini S, Capone F, Leone A, Di Gennaro E, Caraglia M, et al. Acquired resistance to zoledronic acid and the parallel acquisition of an aggressive phenotype are mediated by p38-MAP kinase activation in prostate cancer cells. Cell Death Dis. 2013;4:e641.CrossRefPubMedPubMedCentral
63.
go back to reference Caraglia M, Marra M, Leonetti C, Meo G, D’Alessandro AM, Baldi A, Santini D, Tonini G, Bertieri R, Zupi G, et al. R115777 (Zarnestra((R)))/Zoledronic acid (Zometa((R))) cooperation on inhibition of prostate cancer proliferation is paralleled by Erk/Akt inactivation and reduced Bcl-2 and bad phosphorylation. J Cell Physiol. 2007;211(2):533–43.CrossRefPubMed Caraglia M, Marra M, Leonetti C, Meo G, D’Alessandro AM, Baldi A, Santini D, Tonini G, Bertieri R, Zupi G, et al. R115777 (Zarnestra((R)))/Zoledronic acid (Zometa((R))) cooperation on inhibition of prostate cancer proliferation is paralleled by Erk/Akt inactivation and reduced Bcl-2 and bad phosphorylation. J Cell Physiol. 2007;211(2):533–43.CrossRefPubMed
64.
go back to reference Pelaia G, Gallelli L, Renda T, Fratto D, Falcone D, Caraglia M, Busceti MT, Terracciano R, Vatrella A, Maselli R, et al. Effects of statins and farnesyl transferase inhibitors on ERK phosphorylation, apoptosis and cell viability in non-small lung cancer cells. Cell Proliferat. 2012;45(6):557–65.CrossRef Pelaia G, Gallelli L, Renda T, Fratto D, Falcone D, Caraglia M, Busceti MT, Terracciano R, Vatrella A, Maselli R, et al. Effects of statins and farnesyl transferase inhibitors on ERK phosphorylation, apoptosis and cell viability in non-small lung cancer cells. Cell Proliferat. 2012;45(6):557–65.CrossRef
65.
go back to reference Wang X, Martindale JL, Holbrook NJ. Requirement for ERK activation in cisplatin-induced apoptosis. J Biol Chem. 2000;275(50):39435–43.CrossRefPubMed Wang X, Martindale JL, Holbrook NJ. Requirement for ERK activation in cisplatin-induced apoptosis. J Biol Chem. 2000;275(50):39435–43.CrossRefPubMed
66.
go back to reference Bacus SS, Gudkov AV, Lowe M, Lyass L, Yung Y, Komarov AP, Keyomarsi K, Yarden Y, Seger R. Taxol-induced apoptosis depends on MAP kinase pathways (ERK and p38) and is independent of p53. Oncogene. 2001;20(2):147–55.CrossRefPubMed Bacus SS, Gudkov AV, Lowe M, Lyass L, Yung Y, Komarov AP, Keyomarsi K, Yarden Y, Seger R. Taxol-induced apoptosis depends on MAP kinase pathways (ERK and p38) and is independent of p53. Oncogene. 2001;20(2):147–55.CrossRefPubMed
67.
go back to reference Do MT, Na M, Kim HG, Khanal T, Choi JH, Jin SW, Oh SH, Hwang IH, Chung YC, Kim HS, et al. Ilimaquinone induces death receptor expression and sensitizes human colon cancer cells to TRAIL-induced apoptosis through activation of ROS-ERK/p38 MAPK-CHOP signaling pathways. Food Chem Toxicol. 2014;71:51–9.CrossRefPubMed Do MT, Na M, Kim HG, Khanal T, Choi JH, Jin SW, Oh SH, Hwang IH, Chung YC, Kim HS, et al. Ilimaquinone induces death receptor expression and sensitizes human colon cancer cells to TRAIL-induced apoptosis through activation of ROS-ERK/p38 MAPK-CHOP signaling pathways. Food Chem Toxicol. 2014;71:51–9.CrossRefPubMed
68.
go back to reference Yadav V, Varshney P, Sultana S, Yadav J, Saini N. Moxifloxacin and ciprofloxacin induces S-phase arrest and augments apoptotic effects of cisplatin in human pancreatic cancer cells via ERK activation. BMC Cancer. 2015;15:581.CrossRefPubMedPubMedCentral Yadav V, Varshney P, Sultana S, Yadav J, Saini N. Moxifloxacin and ciprofloxacin induces S-phase arrest and augments apoptotic effects of cisplatin in human pancreatic cancer cells via ERK activation. BMC Cancer. 2015;15:581.CrossRefPubMedPubMedCentral
69.
go back to reference Wang B, Feng Y, Song X, Liu Q, Ning Y, Ou X, Yang J, Zhang X, Wen F. Involvement of ERK, Bcl-2 family and caspase 3 in recombinant human activin A-induced apoptosis in A549. Toxicology. 2009;258(2–3):176–83.CrossRefPubMed Wang B, Feng Y, Song X, Liu Q, Ning Y, Ou X, Yang J, Zhang X, Wen F. Involvement of ERK, Bcl-2 family and caspase 3 in recombinant human activin A-induced apoptosis in A549. Toxicology. 2009;258(2–3):176–83.CrossRefPubMed
70.
go back to reference Randhawa H, Kibble K, Zeng H, Moyer MP, Reindl KM. Activation of ERK signaling and induction of colon cancer cell death by piperlongumine. Toxicol In Vitro. 2013;27(6):1626–33.CrossRefPubMedPubMedCentral Randhawa H, Kibble K, Zeng H, Moyer MP, Reindl KM. Activation of ERK signaling and induction of colon cancer cell death by piperlongumine. Toxicol In Vitro. 2013;27(6):1626–33.CrossRefPubMedPubMedCentral
71.
go back to reference Bangaru ML, Chen S, Woodliff J, Kansra S. Curcumin (diferuloylmethane) induces apoptosis and blocks migration of human medulloblastoma cells. Anticancer Res. 2010;30(2):499–504.PubMed Bangaru ML, Chen S, Woodliff J, Kansra S. Curcumin (diferuloylmethane) induces apoptosis and blocks migration of human medulloblastoma cells. Anticancer Res. 2010;30(2):499–504.PubMed
72.
go back to reference Lai TY, Chen LM, Lin JY, Tzang BS, Lin JA, Tsai CH, Lin YM, Huang CY, Liu CJ, Hsu HH. 17beta-estradiol inhibits prostaglandin E2-induced COX-2 expressions and cell migration by suppressing Akt and ERK1/2 signaling pathways in human LoVo colon cancer cells. Mol Cell Biochem. 2010;342(1–2):63–70.CrossRefPubMed Lai TY, Chen LM, Lin JY, Tzang BS, Lin JA, Tsai CH, Lin YM, Huang CY, Liu CJ, Hsu HH. 17beta-estradiol inhibits prostaglandin E2-induced COX-2 expressions and cell migration by suppressing Akt and ERK1/2 signaling pathways in human LoVo colon cancer cells. Mol Cell Biochem. 2010;342(1–2):63–70.CrossRefPubMed
73.
go back to reference Itoh T, Tanioka M, Yoshida H, Yoshioka T, Nishimoto H, Itohara S. Reduced angiogenesis and tumor progression in gelatinase A-deficient mice. Cancer Res. 1998;58(5):1048–51.PubMed Itoh T, Tanioka M, Yoshida H, Yoshioka T, Nishimoto H, Itohara S. Reduced angiogenesis and tumor progression in gelatinase A-deficient mice. Cancer Res. 1998;58(5):1048–51.PubMed
74.
go back to reference Fojas de Borja P, Collins NK, Du P, Azizkhan-Clifford J, Mudryj M. Cyclin A-CDK phosphorylates Sp1 and enhances Sp1-mediated transcription. EMBO J. 2001;20(20):5737–47.CrossRefPubMed Fojas de Borja P, Collins NK, Du P, Azizkhan-Clifford J, Mudryj M. Cyclin A-CDK phosphorylates Sp1 and enhances Sp1-mediated transcription. EMBO J. 2001;20(20):5737–47.CrossRefPubMed
76.
go back to reference Tsai JR, Chong IW, Chen YH, Hwang JJ, Yin WH, Chen HL, Chou SH, Chiu CC, Liu PL. Magnolol induces apoptosis via caspase-independent pathways in non-small cell lung cancer cells. Arch Pharm Res. 2014;37(4):548–57.CrossRefPubMed Tsai JR, Chong IW, Chen YH, Hwang JJ, Yin WH, Chen HL, Chou SH, Chiu CC, Liu PL. Magnolol induces apoptosis via caspase-independent pathways in non-small cell lung cancer cells. Arch Pharm Res. 2014;37(4):548–57.CrossRefPubMed
77.
go back to reference Vlotides G, Tanyeri A, Spampatti M, Zitzmann K, Chourdakis M, Spttl C, Maurer J, Nolting S, Goke B, Auernhammer CJ. Anticancer effects of metformin on neuroendocrine tumor cells in vitro. Hormones. 2014;13(4):498–508.PubMed Vlotides G, Tanyeri A, Spampatti M, Zitzmann K, Chourdakis M, Spttl C, Maurer J, Nolting S, Goke B, Auernhammer CJ. Anticancer effects of metformin on neuroendocrine tumor cells in vitro. Hormones. 2014;13(4):498–508.PubMed
Metadata
Title
Dual roles of extracellular signal-regulated kinase (ERK) in quinoline compound BPIQ-induced apoptosis and anti-migration of human non-small cell lung cancer cells
Authors
Yao Fong
Chang-Yi Wu
Kuo-Feng Chang
Bing-Hung Chen
Wan-Ju Chou
Chih-Hua Tseng
Yen-Chun Chen
Hui-Min David Wang
Yeh-Long Chen
Chien-Chih Chiu
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2017
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-017-0403-0

Other articles of this Issue 1/2017

Cancer Cell International 1/2017 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine