Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01-12-2018 | Research

Radiotherapy-induced cell death activates paracrine HMGB1-TLR2 signaling and accelerates pancreatic carcinoma metastasis

Authors: Xuelian Chen, Lirong Zhang, Yujie Jiang, Lian Song, Yanfang Liu, Fang Cheng, Xin Fan, Xiongfeng Cao, Aihua Gong, Dongqing Wang, Haitao Zhu

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

Dying cells after irradiation could promote the repopulation of surviving cancer cells leading to tumor recurrence. We aim to define the role of dying cells in promoting pancreatic cancer cells metastasis following radiotherapy.

Methods

Using the transwell system as the in vitro co-culture model, a small number of untreated pancreatic cancer cells were seeded in the upper chamber, while a larger number of lethally treated pancreatic cancer cells were seeded in the lower chamber. A series of experiments were conducted to investigate the role of dying-cell-derived HMGB1 on the invasion of pancreatic cancer in vitro and cancer metastasis in vivo. We then designed shRNA knockdown and Western blot assays to detect signaling activity.

Results

We found that dying pancreatic cancer cells significantly promote the invasion of pancreatic cancer cells in vitro and cancer metastasis in vivo. HMGB1 gene knockdown attenuated the migration-stimulating effect of irradiated, dying cells on living pancreatic cancer cells. Finally, we showed that dying-cell-derived HMGB1 functions in a paracrine manner to affect cancer-cell migration dependent on acquiring an epithelial-mesenchymal transition (EMT) phenotype and PI3K/pAkt activation. This process is mediated by the receptor for TLR2.

Conclusion

Our study indicates that, during radiotherapy, dying pancreatic cancer cells activate paracrine signaling events that promote the mobility of surviving tumor cells. We suggest a strategy to inhibit HMGB1 for preventing pancreatic carcinoma relapse and metastasis.
Appendix
Available only for authorised users
Literature
1.
go back to reference Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21.CrossRefPubMed Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21.CrossRefPubMed
2.
3.
go back to reference Seifert L, Werba G, Tiwari S, Giao Ly NN, Nguy S, Alothman S, Alqunaibit D, Avanzi A, Daley D, Barilla R, et al. Radiation therapy induces macrophages to suppress T-cell responses against pancreatic tumors in mice. Gastroenterology. 2016;150(7):1659–72. e5CrossRefPubMedPubMedCentral Seifert L, Werba G, Tiwari S, Giao Ly NN, Nguy S, Alothman S, Alqunaibit D, Avanzi A, Daley D, Barilla R, et al. Radiation therapy induces macrophages to suppress T-cell responses against pancreatic tumors in mice. Gastroenterology. 2016;150(7):1659–72. e5CrossRefPubMedPubMedCentral
4.
go back to reference Fokas E, O'Neill E, Gordon-Weeks A, Mukherjee S, McKenna WG, Muschel RJ. Pancreatic ductal adenocarcinoma: from genetics to biology to radiobiology to oncoimmunology and all the way back to the clinic. Biochim Biophys Acta. 2015;1855(1):61–82.PubMed Fokas E, O'Neill E, Gordon-Weeks A, Mukherjee S, McKenna WG, Muschel RJ. Pancreatic ductal adenocarcinoma: from genetics to biology to radiobiology to oncoimmunology and all the way back to the clinic. Biochim Biophys Acta. 2015;1855(1):61–82.PubMed
5.
go back to reference Du YX, Liu ZW, You L, Wu WM, Zhao YP. Advances in understanding the molecular mechanism of pancreatic cancer metastasis. Hepatobiliary Pancreat Dis Int. 2016;15(4):361–70.CrossRefPubMed Du YX, Liu ZW, You L, Wu WM, Zhao YP. Advances in understanding the molecular mechanism of pancreatic cancer metastasis. Hepatobiliary Pancreat Dis Int. 2016;15(4):361–70.CrossRefPubMed
6.
go back to reference Liu X, He Y, Li F, Huang Q, Kato TA, Hall RP, Li CY. Redefining the roles of apoptotic factors in carcinogenesis. Mol Cell Oncol. 2016;3(3):e1054550.CrossRefPubMed Liu X, He Y, Li F, Huang Q, Kato TA, Hall RP, Li CY. Redefining the roles of apoptotic factors in carcinogenesis. Mol Cell Oncol. 2016;3(3):e1054550.CrossRefPubMed
7.
go back to reference Huang Q, Li F, Liu X, Li W, Shi W, Liu FF, O'Sullivan B, He Z, Peng Y, Tan AC, et al. Caspase 3-mediated stimulation of tumor cell repopulation during cancer radiotherapy. Nat Med. 2011;17(7):860–6.CrossRefPubMedPubMedCentral Huang Q, Li F, Liu X, Li W, Shi W, Liu FF, O'Sullivan B, He Z, Peng Y, Tan AC, et al. Caspase 3-mediated stimulation of tumor cell repopulation during cancer radiotherapy. Nat Med. 2011;17(7):860–6.CrossRefPubMedPubMedCentral
8.
go back to reference Donato AL, Huang Q, Liu X, Li F, Zimmerman MA, Li CY. Caspase 3 promotes surviving melanoma tumor cell growth after cytotoxic therapy. J Invest Dermatol. 2014;134(6):1686–92.CrossRefPubMedPubMedCentral Donato AL, Huang Q, Liu X, Li F, Zimmerman MA, Li CY. Caspase 3 promotes surviving melanoma tumor cell growth after cytotoxic therapy. J Invest Dermatol. 2014;134(6):1686–92.CrossRefPubMedPubMedCentral
9.
go back to reference Lee SY, Jeong EK, Ju MK, Jeon HM, Kim MY, Kim CH, Park HG, Han SI, Kang HS. Induction of metastasis, cancer stem cell phenotype, and oncogenic metabolism in cancer cells by ionizing radiation. Mol Cancer. 2017;16(1):10.CrossRefPubMedPubMedCentral Lee SY, Jeong EK, Ju MK, Jeon HM, Kim MY, Kim CH, Park HG, Han SI, Kang HS. Induction of metastasis, cancer stem cell phenotype, and oncogenic metabolism in cancer cells by ionizing radiation. Mol Cancer. 2017;16(1):10.CrossRefPubMedPubMedCentral
10.
go back to reference De Bacco F, Luraghi P, Medico E, Reato G, Girolami F, Perera T, Gabriele P, Comoglio PM, Boccaccio C. Induction of MET by ionizing radiation and its role in radioresistance and invasive growth of cancer. J Natl Cancer Inst. 2011;103(8):645–61.CrossRefPubMed De Bacco F, Luraghi P, Medico E, Reato G, Girolami F, Perera T, Gabriele P, Comoglio PM, Boccaccio C. Induction of MET by ionizing radiation and its role in radioresistance and invasive growth of cancer. J Natl Cancer Inst. 2011;103(8):645–61.CrossRefPubMed
11.
go back to reference Zhang J, Zhang L, Zhang S, Yu Q, Xiong F, Huang K, Wang CY, Yang P. HMGB1, an innate alarmin, plays a critical role in chronic inflammation of adipose tissue in obesity. Mol Cell Endocrinol. 2017;454:103–11.CrossRefPubMed Zhang J, Zhang L, Zhang S, Yu Q, Xiong F, Huang K, Wang CY, Yang P. HMGB1, an innate alarmin, plays a critical role in chronic inflammation of adipose tissue in obesity. Mol Cell Endocrinol. 2017;454:103–11.CrossRefPubMed
13.
go back to reference Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, et al. HMGB1 in health and disease. Mol Asp Med. 2014;40:1–116.CrossRef Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, et al. HMGB1 in health and disease. Mol Asp Med. 2014;40:1–116.CrossRef
14.
go back to reference Cebrian MJ, Bauden M, Andersson R, Holdenrieder S, Ansari D. Paradoxical role of HMGB1 in pancreatic cancer: tumor suppressor or tumor promoter? Anticancer Res. 2016;36(9):4381–9.CrossRefPubMed Cebrian MJ, Bauden M, Andersson R, Holdenrieder S, Ansari D. Paradoxical role of HMGB1 in pancreatic cancer: tumor suppressor or tumor promoter? Anticancer Res. 2016;36(9):4381–9.CrossRefPubMed
15.
go back to reference Herzog C, Lorenz A, Gillmann HJ, Chowdhury A, Larmann J, Harendza T, Echtermeyer F, Muller M, Schmitz M, Stypmann J, et al. Thrombomodulin's lectin-like domain reduces myocardial damage by interfering with HMGB1-mediated TLR2 signalling. Cardiovasc Res. 2014;101(3):400–10.CrossRefPubMed Herzog C, Lorenz A, Gillmann HJ, Chowdhury A, Larmann J, Harendza T, Echtermeyer F, Muller M, Schmitz M, Stypmann J, et al. Thrombomodulin's lectin-like domain reduces myocardial damage by interfering with HMGB1-mediated TLR2 signalling. Cardiovasc Res. 2014;101(3):400–10.CrossRefPubMed
16.
go back to reference Kim S, Kim SY, Pribis JP, Lotze M, Mollen KP, Shapiro R, Loughran P, Scott MJ, Billiar TR. Signaling of high mobility group box 1 (HMGB1) through toll-like receptor 4 in macrophages requires CD14. Mol Med. 2013;19:88–98.CrossRefPubMedPubMedCentral Kim S, Kim SY, Pribis JP, Lotze M, Mollen KP, Shapiro R, Loughran P, Scott MJ, Billiar TR. Signaling of high mobility group box 1 (HMGB1) through toll-like receptor 4 in macrophages requires CD14. Mol Med. 2013;19:88–98.CrossRefPubMedPubMedCentral
17.
go back to reference Nestl A, Von Stein OD, Zatloukal K, Thies WG, Herrlich P, Hofmann M, Sleeman JP. Gene expression patterns associated with the metastatic phenotype in rodent and human tumors. Cancer Res. 2001;61(4):1569–77.PubMed Nestl A, Von Stein OD, Zatloukal K, Thies WG, Herrlich P, Hofmann M, Sleeman JP. Gene expression patterns associated with the metastatic phenotype in rodent and human tumors. Cancer Res. 2001;61(4):1569–77.PubMed
18.
go back to reference Evans A, Lennard TW, Davies BR. High-mobility group protein 1(Y): metastasis-associated or metastasis-inducing? J Surg Oncol. 2004;88(2):86–99.CrossRefPubMed Evans A, Lennard TW, Davies BR. High-mobility group protein 1(Y): metastasis-associated or metastasis-inducing? J Surg Oncol. 2004;88(2):86–99.CrossRefPubMed
19.
go back to reference Krebs AM, Mitschke J, Lasierra Losada M, Schmalhofer O, Boerries M, Busch H, Boettcher M, Mougiakakos D, Reichardt W, Bronsert P, et al. The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nat Cell Biol. 2017;19(5):518–29.CrossRefPubMed Krebs AM, Mitschke J, Lasierra Losada M, Schmalhofer O, Boerries M, Busch H, Boettcher M, Mougiakakos D, Reichardt W, Bronsert P, et al. The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nat Cell Biol. 2017;19(5):518–29.CrossRefPubMed
21.
go back to reference Zhu L, Li X, Chen Y, Fang J, Ge Z. High-mobility group box 1: a novel inducer of the epithelial-mesenchymal transition in colorectal carcinoma. Cancer Lett. 2015;357(2):527–34.CrossRefPubMed Zhu L, Li X, Chen Y, Fang J, Ge Z. High-mobility group box 1: a novel inducer of the epithelial-mesenchymal transition in colorectal carcinoma. Cancer Lett. 2015;357(2):527–34.CrossRefPubMed
22.
go back to reference Xu YF, Ge FJ, Han B, Yang XQ, Su H, Zhao AC, Zhao MH, Yang YB, Yang J. High-mobility group box 1 expression and lymph node metastasis in intrahepatic cholangiocarcinoma. World J Gastroenterol. 2015;21(11):3256–65.CrossRefPubMedPubMedCentral Xu YF, Ge FJ, Han B, Yang XQ, Su H, Zhao AC, Zhao MH, Yang YB, Yang J. High-mobility group box 1 expression and lymph node metastasis in intrahepatic cholangiocarcinoma. World J Gastroenterol. 2015;21(11):3256–65.CrossRefPubMedPubMedCentral
23.
go back to reference Xie X, Zhu H, Yang H, Huang W, Wu Y, Wang Y, Luo Y, Wang D, Shao G. Solamargine triggers hepatoma cell death through apoptosis. Oncol Lett. 2015;10(1):168–74.CrossRefPubMedPubMedCentral Xie X, Zhu H, Yang H, Huang W, Wu Y, Wang Y, Luo Y, Wang D, Shao G. Solamargine triggers hepatoma cell death through apoptosis. Oncol Lett. 2015;10(1):168–74.CrossRefPubMedPubMedCentral
24.
go back to reference Zhu H, Wang D, Liu Y, Su Z, Zhang L, Chen F, Zhou Y, Wu Y, Yu M, Zhang Z, et al. Role of the hypoxia-inducible factor-1 alpha induced autophagy in the conversion of non-stem pancreatic cancer cells into CD133+ pancreatic cancer stem-like cells. Cancer Cell Int. 2013;13(1):119.CrossRefPubMedPubMedCentral Zhu H, Wang D, Liu Y, Su Z, Zhang L, Chen F, Zhou Y, Wu Y, Yu M, Zhang Z, et al. Role of the hypoxia-inducible factor-1 alpha induced autophagy in the conversion of non-stem pancreatic cancer cells into CD133+ pancreatic cancer stem-like cells. Cancer Cell Int. 2013;13(1):119.CrossRefPubMedPubMedCentral
25.
go back to reference Zhang L, Wang D, Li Y, Liu Y, Xie X, Wu Y, Zhou Y, Ren J, Zhang J, Zhu H, et al. CCL21/CCR7 axis contributed to CD133+ pancreatic cancer stem-like cell metastasis via EMT and Erk/NF-kappaB pathway. PLoS One. 2016;11(8):e0158529.CrossRefPubMedPubMedCentral Zhang L, Wang D, Li Y, Liu Y, Xie X, Wu Y, Zhou Y, Ren J, Zhang J, Zhu H, et al. CCL21/CCR7 axis contributed to CD133+ pancreatic cancer stem-like cell metastasis via EMT and Erk/NF-kappaB pathway. PLoS One. 2016;11(8):e0158529.CrossRefPubMedPubMedCentral
26.
go back to reference Feng X, Yu Y, He S, Cheng J, Gong Y, Zhang Z, Yang X, Xu B, Liu X, Li CY, et al. Dying glioma cells establish a proangiogenic microenvironment through a caspase 3 dependent mechanism. Cancer Lett. 2017;385:12–20.CrossRefPubMed Feng X, Yu Y, He S, Cheng J, Gong Y, Zhang Z, Yang X, Xu B, Liu X, Li CY, et al. Dying glioma cells establish a proangiogenic microenvironment through a caspase 3 dependent mechanism. Cancer Lett. 2017;385:12–20.CrossRefPubMed
29.
go back to reference Luo Y, Chihara Y, Fujimoto K, Sasahira T, Kuwada M, Fujiwara R, Fujii K, Ohmori H, Kuniyasu H. High mobility group box 1 released from necrotic cells enhances regrowth and metastasis of cancer cells that have survived chemotherapy. Eur J Cancer. 2013;49(3):741–51.CrossRefPubMed Luo Y, Chihara Y, Fujimoto K, Sasahira T, Kuwada M, Fujiwara R, Fujii K, Ohmori H, Kuniyasu H. High mobility group box 1 released from necrotic cells enhances regrowth and metastasis of cancer cells that have survived chemotherapy. Eur J Cancer. 2013;49(3):741–51.CrossRefPubMed
30.
go back to reference Kang R, Xie Y, Zhang Q, Hou W, Jiang Q, Zhu S, Liu J, Zeng D, Wang H, Bartlett DL, et al. Intracellular HMGB1 as a novel tumor suppressor of pancreatic cancer. Cell Res. 2017;27(7):916–32.CrossRefPubMedPubMedCentral Kang R, Xie Y, Zhang Q, Hou W, Jiang Q, Zhu S, Liu J, Zeng D, Wang H, Bartlett DL, et al. Intracellular HMGB1 as a novel tumor suppressor of pancreatic cancer. Cell Res. 2017;27(7):916–32.CrossRefPubMedPubMedCentral
31.
go back to reference Zhang Z, Wang M, Zhou L, Feng X, Cheng J, Yu Y, Gong Y, Zhu Y, Li C, Tian L, et al. Increased HMGB1 and cleaved caspase-3 stimulate the proliferation of tumor cells and are correlated with the poor prognosis in colorectal cancer. J Exp Clin Cancer Res. 2015;34:51.CrossRefPubMedPubMedCentral Zhang Z, Wang M, Zhou L, Feng X, Cheng J, Yu Y, Gong Y, Zhu Y, Li C, Tian L, et al. Increased HMGB1 and cleaved caspase-3 stimulate the proliferation of tumor cells and are correlated with the poor prognosis in colorectal cancer. J Exp Clin Cancer Res. 2015;34:51.CrossRefPubMedPubMedCentral
32.
go back to reference Chen RC, Yi PP, Zhou RR, Xiao MF, Huang ZB, Tang DL, Huang Y, Fan XG. The role of HMGB1-RAGE axis in migration and invasion of hepatocellular carcinoma cell lines. Mol Cell Biochem. 2014;390(1–2):271–80.CrossRefPubMedPubMedCentral Chen RC, Yi PP, Zhou RR, Xiao MF, Huang ZB, Tang DL, Huang Y, Fan XG. The role of HMGB1-RAGE axis in migration and invasion of hepatocellular carcinoma cell lines. Mol Cell Biochem. 2014;390(1–2):271–80.CrossRefPubMedPubMedCentral
33.
go back to reference Takada M, Hirata K, Ajiki T, Suzuki Y, Kuroda Y. Expression of receptor for advanced glycation end products (RAGE) and MMP-9 in human pancreatic cancer cells. Hepato-Gastroenterology. 2004;51(58):928–30.PubMed Takada M, Hirata K, Ajiki T, Suzuki Y, Kuroda Y. Expression of receptor for advanced glycation end products (RAGE) and MMP-9 in human pancreatic cancer cells. Hepato-Gastroenterology. 2004;51(58):928–30.PubMed
34.
go back to reference Santoni M, Andrikou K, Sotte V, Bittoni A, Lanese A, Pellei C, Piva F, Conti A, Nabissi M, Santoni G, et al. Toll like receptors and pancreatic diseases: from a pathogenetic mechanism to a therapeutic target. Cancer Treat Rev. 2015;41(7):569–76.CrossRefPubMed Santoni M, Andrikou K, Sotte V, Bittoni A, Lanese A, Pellei C, Piva F, Conti A, Nabissi M, Santoni G, et al. Toll like receptors and pancreatic diseases: from a pathogenetic mechanism to a therapeutic target. Cancer Treat Rev. 2015;41(7):569–76.CrossRefPubMed
35.
go back to reference Carl C, Flindt A, Hartmann J, Dahlke M, Rades D, Dunst J, Lehnert H, Gieseler F, Ungefroren H. Ionizing radiation induces a motile phenotype in human carcinoma cells in vitro through hyperactivation of the TGF-beta signaling pathway. Cell Mol Life Sci. 2016;73(2):427–43.CrossRefPubMed Carl C, Flindt A, Hartmann J, Dahlke M, Rades D, Dunst J, Lehnert H, Gieseler F, Ungefroren H. Ionizing radiation induces a motile phenotype in human carcinoma cells in vitro through hyperactivation of the TGF-beta signaling pathway. Cell Mol Life Sci. 2016;73(2):427–43.CrossRefPubMed
36.
go back to reference Nagarajan D, Melo T, Deng Z, Almeida C, Zhao W. ERK/GSK3beta/snail signaling mediates radiation-induced alveolar epithelial-to-mesenchymal transition. Free Radic Biol Med. 2012;52(6):983–92.CrossRefPubMed Nagarajan D, Melo T, Deng Z, Almeida C, Zhao W. ERK/GSK3beta/snail signaling mediates radiation-induced alveolar epithelial-to-mesenchymal transition. Free Radic Biol Med. 2012;52(6):983–92.CrossRefPubMed
37.
go back to reference Chen W, Wu S, Zhang G, Wang W, Shi Y. Effect of AKT inhibition on epithelial-mesenchymal transition and ZEB1-potentiated radiotherapy in nasopharyngeal carcinoma. Oncol Lett. 2013;6(5):1234–40.CrossRefPubMedPubMedCentral Chen W, Wu S, Zhang G, Wang W, Shi Y. Effect of AKT inhibition on epithelial-mesenchymal transition and ZEB1-potentiated radiotherapy in nasopharyngeal carcinoma. Oncol Lett. 2013;6(5):1234–40.CrossRefPubMedPubMedCentral
Metadata
Title
Radiotherapy-induced cell death activates paracrine HMGB1-TLR2 signaling and accelerates pancreatic carcinoma metastasis
Authors
Xuelian Chen
Lirong Zhang
Yujie Jiang
Lian Song
Yanfang Liu
Fang Cheng
Xin Fan
Xiongfeng Cao
Aihua Gong
Dongqing Wang
Haitao Zhu
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2018
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0726-2

Other articles of this Issue 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 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