Exogenous HMGB1 Promotes the Proliferation and Metastasis of Pancreatic Cancer Cells

Li Zhu1, Shuai Ren1, Marcus J. Daniels2, Wenli Qiu1, Song Lian3, Tao You4, Dongqing Wang3, Zhongqiu Wang1
1Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, China
2Department of Radiology, Johns Hopkins University School of Medicine, United States
3Department of Radiology, Affiliated Hospital of Jiangsu University, China
4Department of Radiotherapy, Affiliated Hospital of Jiangsu University, China

Tóm tắt

Background: Exogenous HMGB1 plays a vital role in tumor recurrence, and HMGB1 is ubiquitous in the tumor microenvironment. However, the mechanism of action is still unclear. We investigated the role of exogenous HMGB1 in tumor proliferation and metastasis using human SW1990 and PANC-1 cells after radiotherapy and explored the possible molecular mechanism.Materials and Methods: Residual PANC-1 cells and SW1990 cells were isolated after radiotherapy. The supernatant after radiotherapy was collected. The relative expression of HMGB1 was evaluated by Enzyme Linked Immunosorbent Assay (ELISA). Electron microscope (EMS) was used to collect the images of pancreatic cancer cells pre and post radiotherapy treatment. The proliferation of pancreatic cancer cells which were treated with different radiation doses was measured by Carboxy Fluorescein Succinimidyl Ester (CFSE). The migration rates of pancreatic cancer cells were measured by wound healing assays. Subsequently, the expression of related proteins was detected by Western Blot. In vivo, the subcutaneous pancreatic tumor models of nude mice were established, and therapeutic capabilities were tested.Results: HMGB1 was detected in the supernatant of pancreatic cancer cells after radiotherapy. The results of CFSE showed that exogenous HMGB1 promotes the proliferation and metastasis of pancreatic cancer cells. The western blot results showed activation of p-GSK 3β and up-regulation of N-CA, Bcl-2, and Ki67 in response to HMGB1 stimulation, while E-CA expression was down-regulated in pancreatic cancer cells in response to HMGB1 stimulation. In vivo, ethyl pyruvate (EP, HMGB1 inhibitor) inhibits the growth of tumors and HMGB1 promotes the proliferation of tumors after radiation.Conclusion: Radiotherapy induces HMGB1 release into the extracellular space. Exogenous HMGB1 promotes the proliferation and metastasis of PANC-1 cells and SW1990 cells by activation of p-GSK 3β which is mediated by Wnt pathway.

Từ khóa


Tài liệu tham khảo

Zeng, 2019, An overactive neddylation pathway serves as a therapeutic target and MLN4924 enhances the anticancer activity of cisplatin in pancreatic cancer, Oncol Lett., 18, 2724, 10.3892/ol.2019.10596

Marrett, 2008, Canadian cancer statistics at a glance: mesothelioma, CMAJ., 178, 677, 10.1503/cmaj.080129

Liu, 2016, Oridonin inhibits pancreatic cancer cell migration and epithelial-mesenchymal transition by suppressing Wnt/β-catenin signaling pathway, Cancer Cell Int., 16, 57, 10.1186/s12935-016-0336-z

McGuigan, 2018, Pancreatic cancer: a review of clinical diagnosis, epidemiology, treatment and outcomes, World J Gastroenterol., 24, 4846, 10.3748/wjg.v24.i43.4846

Jiang, 2020, Dying tumor cell-derived exosomal miR-194-5p potentiates survival and repopulation of tumor repopulating cells upon radiotherapy in pancreatic cancer, Mol Cancer., 19, 68, 10.1186/s12943-020-01178-6

Pereira, 2020, iNOS regulates the therapeutic response of pancreatic cancer cells to radiotherapy, Cancer Res., 80, 1681, 10.1158/0008-5472.CAN-19-2991

Che, 2020, Pancreatic cancer resistance conferred by stellate cells: looking for new preclinical models, Exp Hematol Oncol., 9, 18, 10.1186/s40164-020-00176-0

Wang, 2018, SMAD4 gene mutation renders pancreatic cancer resistance to radiotherapy through promotion of autophagy, Clin Cancer Res., 24, 3176, 10.1158/1078-0432.CCR-17-3435

Wang, 2021, lncRNA ANRIL aggravates the chemoresistance of pancreatic cancer cells to gemcitabine by targeting inhibition of miR-181a and targeting HMGB1-induced autophagy, Aging., 13, 19272, 10.18632/aging.203251

Goodwin, 1973, Isolation and characterisation of two calf-thymus chromatin non-histone proteins with high contents of acidic and basic amino acids, Eur J Biochem., 40, 215, 10.1111/j.1432-1033.1973.tb03188.x

Yang, 2012, High-mobility group nucleosome-binding protein 1 acts as an alarmin and is critical for lipopolysaccharide-induced immune responses, J Exp Med., 209, 157, 10.1084/jem.20101354

Bianchi, 1992, The DNA binding site of HMG1 protein is composed of two similar segments (HMG boxes), both of which have counterparts in other eukaryotic regulatory proteins, EMBO J., 11, 1055, 10.1002/j.1460-2075.1992.tb05144.x

Kang, 2014, HMGB1 in health and disease, Mol Aspects Med., 40, 1, 10.1016/j.mam.2014.05.001

Romani, 1979, Serological analysis of species specificity in the high mobility group chromosomal proteins, J Biol Chem., 254, 2918, 10.1016/S0021-9258(17)30161-8

Tang, 2012, Tumor immunity times out: TIM-3 and HMGB1, Nat Immunol., 13, 808, 10.1038/ni.2396

Ahmed, 2020, Targeting immunogenic cell death in cancer, Mol Oncol., 14, 2994, 10.1002/1878-0261.12851

Hayashi, 2020, Tipping the immunostimulatory and inhibitory DAMP balance to harness immunogenic cell death, Nat Commun., 11, 6299, 10.1038/s41467-020-19970-9

Min, 2021, Th2 cytokines-DUOX2-ROS-HMGB1 translocation axis is important in the pathogenesis of allergic rhinitis, Clin Sci., 135, 483, 10.1042/CS20201212

Xue, 2021, HMGB1 as a therapeutic target in disease, J Cell Physiol., 236, 3406, 10.1002/jcp.30125

Tang, 2010, The redox protein HMGB1 regulates cell death and survival in cancer treatment, Autophagy., 6, 1181, 10.4161/auto.6.8.13367

Bonaldi, 2003, Monocytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion, EMBO J., 22, 5551, 10.1093/emboj/cdg516

Wang, 2014, Wnt and the Wnt signaling pathway in bone development and disease, Front Biosci., 19, 379, 10.2741/4214

Rahman, 2019, Molecular cloning and transcriptional and functional analysis of glycogen synthase kinase-3β in Haemaphysalis longicornis (Acari, Ixodidae), Parasite., 26, 39, 10.1051/parasite/2019038

Li, 2018, Oridonin inhibits migration, invasion, adhesion and TGF-β1-induced epithelial-mesenchymal transition of melanoma cells by inhibiting the activity of PI3K/Akt/GSK-3β signaling pathway, Oncol Lett., 15, 1362, 10.3892/ol.2017.7421

Zhang, 2018, GSK3β inhibits epithelial-mesenchymal transition via the Wnt/β-catenin and PI3K/Akt pathways, Int J Ophthalmol., 11, 1120, 10.18240/ijo.2018.07.08

Saraswati, 2018, Glycogen synthase kinase-3 and its inhibitors: potential target for various therapeutic conditions, Eur J Med Chem., 144, 843, 10.1016/j.ejmech.2017.11.103

Li, 2014, Extracellular lumican inhibits pancreatic cancer cell growth and is associated with prolonged survival after surgery, Clin Cancer Res., 20, 6529, 10.1158/1078-0432.CCR-14-0970

Ferlay, 2019, Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods, Int J Cancer., 144, 1941, 10.1002/ijc.31937

Nichols, 2019, Optimizing neoadjuvant radiotherapy for resectable and borderline resectable pancreatic cancer using protons, World J Gastrointest Surg., 11, 303, 10.4240/wjgs.v11.i7.303

Conlon, 1996, Long-term survival after curative resection for pancreatic ductal adenocarcinoma. Clinicopathologic analysis of 5-year survivors, Ann Surg., 223, 273, 10.1097/00000658-199603000-00007

Richard, 2017, Post-translational modifications of high mobility group box 1 and cancer, Am J Transl Res., 9, 5181

Kang, 2014, The HMGB1/RAGE inflammatory pathway promotes pancreatic tumor growth by regulating mitochondrial bioenergetics, Oncogene., 33, 567, 10.1038/onc.2012.631

He, 2017, The dual role and therapeutic potential of high-mobility group box 1 in cancer, Oncotarget., 8, 64534, 10.18632/oncotarget.17885

Zhou, 2011, Development and characterization of a potent immunoconjugate targeting the Fn14 receptor on solid tumor cells, Mol Cancer Ther., 10, 1276, 10.1158/1535-7163.MCT-11-0161

Thiery, 2003, Epithelial-mesenchymal transitions in development and pathologies, Curr Opin Cell Biol., 15, 740, 10.1016/j.ceb.2003.10.006

Rodriguez-Aznar, 2019, EMT and stemness-key players in pancreatic cancer stem cells, Cancers., 11, 1136, 10.3390/cancers11081136

Diepenbruck, 2016, Epithelial-mesenchymal transition (EMT) and metastasis: yes, no, maybe?, Curr Opin Cell Biol., 43, 7, 10.1016/j.ceb.2016.06.002

Chaffer, 2016, EMT, cell plasticity and metastasis, Cancer Metastasis Rev., 35, 645, 10.1007/s10555-016-9648-7

Cheng, 2007, Twist transcriptionally up-regulates AKT2 in breast cancer cells leading to increased migration, invasion, and resistance to paclitaxel, Cancer Res, 67, 1979, 10.1158/0008-5472.CAN-06-1479

Tiwari, 2012, EMT as the ultimate survival mechanism of cancer cells, Semin Cancer Biol., 22, 194, 10.1016/j.semcancer.2012.02.013

Clevers, 2006, Wnt/beta-catenin signaling in development and disease, Cell., 127, 469, 10.1016/j.cell.2006.10.018

Bullions, 1998, The role of beta-catenin in cell adhesion, signal transduction, and cancer, Curr Opin Oncol., 10, 81, 10.1097/00001622-199801000-00013

Lochhead, 2006, Chaperone-dependent GSK3beta transitional intermediate mediates activation-loop autophosphorylation, Mol Cell., 24, 627, 10.1016/j.molcel.2006.10.009

Li, 2019, HMGB1-Induced p62 overexpression promotes snail-mediated epithelial-mesenchymal transition in glioblastoma cells via the degradation of GSK-3β, Theranostics, 9, 1909, 10.7150/thno.30578

Li, 2020, Fluorine assembly nanocluster breaks the shackles of immunosuppression to turn the cold tumor hot, Proc Natl Acad Sci U S A., 117, 32962, 10.1073/pnas.2011297117

Li, 2020, Targeting pulmonary tumor microenvironment with CXCR4-inhibiting nanocomplex to enhance anti-PD-L1 immunotherapy, Sci Adv, 6, eaaz9240, 10.1126/sciadv.aaz9240