Skip to main content
Top
Published in: Cancer Cell International 1/2021

Open Access 01-12-2021 | Leukemia | Primary Research

Extracellular HMGB1 interacts with RAGE and promotes chemoresistance in acute leukemia cells

Authors: Weixin Lai, Xinyu Li, Qian Kong, Han Chen, Yunyao Li, Lu-Hong Xu, Jianpei Fang

Published in: Cancer Cell International | Issue 1/2021

Login to get access

Abstract

Background

Nowadays, acute leukemia (AL) among children has favorable outcome, yet some of them get refractory or relapse mainly due to drug resistance. High-mobility group box 1 (HMGB1) has been proven to have a important role in drug resistance via upregulation of autophagy after chemotherapy treatment in acute leukemia. However, the mechanism how extracellular HMGB1 acts on AL cells and leads to chemoresistance remains elusive.

Method

CCK8 was used to examine the toxicity of chemotherapeutic drug. Elisa was performed to detect the release of HMGB1. Western blot and mRFP-GFP-LC3 adenoviral particles as well as transmission electron microscopy were used to detect the autophagy flux. Western blot and flow cytometry were applied to evaluate the apoptosis. qPCR and western blot were conducted to detect the expression of drug efflux protein. Lentivirus infection was applied to knock down RAGE. In addition, T-ALL NOD/SCID mice xenograft model was used to observe the effect of inhibiting HMGB1/RAGE axis.

Results

We found that extracellular HMGB1 do upregulate autophagy and in the meantime downregulate apoptosis, primarily through interaction with receptor for advanced glycation end products (RAGE). Suppression of RAGE by RNA interference alleviated the level of autophagy and enhanced apoptosis. What’s more, HMGB1/RAGE induced autophagy was associated with the activation of ERK1/2 and decreased phosphorylation of mammalian target of rapamycin (mTOR), while HMGB1/RAGE limited apoptosis in a Bcl-2-regulated way mediated by P53. On the other hand, we found that HMGB1/RAGE activated the NF-κB pathway and promoted the expression of P-glycation protein (P-gp) as well as multidrug resistance-associated protein (MRP), both are ATP-binding cassette transporters. In vivo experiment, we found that blocking HMGB1/RAGE axis do have a mild pathological condition and a better survival in T-ALL mice.

Conclusion

HMGB1/RAGE have a important role in drug resistance after chemotherapy treatment, mainly by regulating autophagy and apoptosis as well as promoting the expression of drug efflux protein such as P-gp and MRP. HMGB1/RAGE might be a promising target to cure AL, especially for those met with relapse and refractory.
Literature
2.
go back to reference Karrman K, Johansson B. Pediatric T-cell acute lymphoblastic leukemia. Genes Chromosom Cancer. 2017;56(2):89–116.PubMedCrossRef Karrman K, Johansson B. Pediatric T-cell acute lymphoblastic leukemia. Genes Chromosom Cancer. 2017;56(2):89–116.PubMedCrossRef
3.
go back to reference Hoffman AE, Schoonmade LJ, Kaspers GJ. Pediatric relapsed acute myeloid leukemia: a systematic review. Expert Rev Anticancer Ther. 2021;21(1):45–52.PubMedCrossRef Hoffman AE, Schoonmade LJ, Kaspers GJ. Pediatric relapsed acute myeloid leukemia: a systematic review. Expert Rev Anticancer Ther. 2021;21(1):45–52.PubMedCrossRef
4.
go back to reference Shen S, Cai J, Chen J, et al. Long-term results of the risk-stratified treatment of childhood acute lymphoblastic leukemia in China. Hematol Oncol. 2018;36(4):679–88.PubMedCrossRef Shen S, Cai J, Chen J, et al. Long-term results of the risk-stratified treatment of childhood acute lymphoblastic leukemia in China. Hematol Oncol. 2018;36(4):679–88.PubMedCrossRef
5.
go back to reference Hunger SP, Lu X, Devidas M, et al. Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the Children’s Oncology Group. J Clin Oncol. 2012;30(14):1663–9.PubMedPubMedCentralCrossRef Hunger SP, Lu X, Devidas M, et al. Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the Children’s Oncology Group. J Clin Oncol. 2012;30(14):1663–9.PubMedPubMedCentralCrossRef
6.
go back to reference Sarmento-Ribeiro AB, Scorilas A, Gonçalves AC, et al. The emergence of drug resistance to targeted cancer therapies: clinical evidence. Drug Resist Updates. 2019;47:100646.CrossRef Sarmento-Ribeiro AB, Scorilas A, Gonçalves AC, et al. The emergence of drug resistance to targeted cancer therapies: clinical evidence. Drug Resist Updates. 2019;47:100646.CrossRef
7.
go back to reference Sehgal AR, Konig H, Johnson DE, et al. You eat what you are: autophagy inhibition as a therapeutic strategy in leukemia. Leukemia. 2015;29(3):517–25.PubMedCrossRef Sehgal AR, Konig H, Johnson DE, et al. You eat what you are: autophagy inhibition as a therapeutic strategy in leukemia. Leukemia. 2015;29(3):517–25.PubMedCrossRef
8.
go back to reference Sever ON, Demir OG. Autophagy: cell death or survive mechanism. J Oncol Sci. 2017;3(2):37–44.CrossRef Sever ON, Demir OG. Autophagy: cell death or survive mechanism. J Oncol Sci. 2017;3(2):37–44.CrossRef
10.
go back to reference Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.CrossRef Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.CrossRef
11.
go back to reference Evangelisti C, Evangelisti C, Chiarini F, et al. Autophagy in acute leukemias: A double-edged sword with important therapeutic implications. Biochim Biophys Acta BBA Mol Cell Res. 2015;1853(1):14–26.CrossRef Evangelisti C, Evangelisti C, Chiarini F, et al. Autophagy in acute leukemias: A double-edged sword with important therapeutic implications. Biochim Biophys Acta BBA Mol Cell Res. 2015;1853(1):14–26.CrossRef
12.
go back to reference Nikoletopoulou V, Markaki M, Palikaras K, et al. Crosstalk between apoptosis, necrosis and autophagy. Biochim Biophys Acta BBA Mol Cell Res. 2013;1833(12):3448–59.CrossRef Nikoletopoulou V, Markaki M, Palikaras K, et al. Crosstalk between apoptosis, necrosis and autophagy. Biochim Biophys Acta BBA Mol Cell Res. 2013;1833(12):3448–59.CrossRef
13.
go back to reference Xu J, Patel NH, Gewirtz DA. Triangular relationship between p53, autophagy, and chemotherapy resistance. Int J Mol Sci. 2020;21(23):8991.PubMedCentralCrossRef Xu J, Patel NH, Gewirtz DA. Triangular relationship between p53, autophagy, and chemotherapy resistance. Int J Mol Sci. 2020;21(23):8991.PubMedCentralCrossRef
14.
go back to reference Yang L, Yu Y, Kang R, et al. Up-regulated autophagy by endogenous high mobility group box-1 promotes chemoresistance in leukemia cells. Leuk Lymphoma. 2012;53(2):315–22.PubMedCrossRef Yang L, Yu Y, Kang R, et al. Up-regulated autophagy by endogenous high mobility group box-1 promotes chemoresistance in leukemia cells. Leuk Lymphoma. 2012;53(2):315–22.PubMedCrossRef
15.
go back to reference Zhao M, Yang M, Yang L, et al. HMGB1 regulates autophagy through increasing transcriptional activities of JNK and ERK in human myeloid leukemia cells. BMB Rep. 2011;44(9):601–6.PubMedCrossRef Zhao M, Yang M, Yang L, et al. HMGB1 regulates autophagy through increasing transcriptional activities of JNK and ERK in human myeloid leukemia cells. BMB Rep. 2011;44(9):601–6.PubMedCrossRef
16.
go back to reference Huang Z, Zhong Z, Zhang L, et al. Down-regulation of HMGB1 expression by shRNA constructs inhibits the bioactivity of urothelial carcinoma cell lines via the NF-κB pathway. Sci Rep. 2015;5(1):12807.PubMedPubMedCentralCrossRef Huang Z, Zhong Z, Zhang L, et al. Down-regulation of HMGB1 expression by shRNA constructs inhibits the bioactivity of urothelial carcinoma cell lines via the NF-κB pathway. Sci Rep. 2015;5(1):12807.PubMedPubMedCentralCrossRef
17.
go back to reference Liu Y, Chen P, Xu L, et al. Extracellular HMGB1 prevents necroptosis in acute myeloid leukemia cells. Biomed Pharmacother. 2019;112:108714.PubMedCrossRef Liu Y, Chen P, Xu L, et al. Extracellular HMGB1 prevents necroptosis in acute myeloid leukemia cells. Biomed Pharmacother. 2019;112:108714.PubMedCrossRef
18.
go back to reference Huang C, Chiang S, Chen WT, et al. HMGB1 promotes ERK-mediated mitochondrial Drp1 phosphorylation for chemoresistance through RAGE in colorectal cancer. Cell Death Dis. 2018;9(10):1004.PubMedPubMedCentralCrossRef Huang C, Chiang S, Chen WT, et al. HMGB1 promotes ERK-mediated mitochondrial Drp1 phosphorylation for chemoresistance through RAGE in colorectal cancer. Cell Death Dis. 2018;9(10):1004.PubMedPubMedCentralCrossRef
19.
go back to reference Yin H, Yang X, Gu W, et al. HMGB1-mediated autophagy attenuates gemcitabine-induced apoptosis in bladder cancer cells involving JNK and ERK activation. Oncotarget. 2017;8(42):71642–56.PubMedPubMedCentralCrossRef Yin H, Yang X, Gu W, et al. HMGB1-mediated autophagy attenuates gemcitabine-induced apoptosis in bladder cancer cells involving JNK and ERK activation. Oncotarget. 2017;8(42):71642–56.PubMedPubMedCentralCrossRef
20.
go back to reference Yuan S, Liu Z, Xu Z, et al. High mobility group box 1 (HMGB1): a pivotal regulator of hematopoietic malignancies. J Hematol Oncol. 2020;13(1):1–91.CrossRef Yuan S, Liu Z, Xu Z, et al. High mobility group box 1 (HMGB1): a pivotal regulator of hematopoietic malignancies. J Hematol Oncol. 2020;13(1):1–91.CrossRef
21.
go back to reference Farahat M, Sharaf M, Attia T. The serum high-mobility group box 1 level and RAGE expression in childhood acute lymphoblastic leukemic patients’. Egypt J Haematol. 2015;40(2):60.CrossRef Farahat M, Sharaf M, Attia T. The serum high-mobility group box 1 level and RAGE expression in childhood acute lymphoblastic leukemic patients’. Egypt J Haematol. 2015;40(2):60.CrossRef
22.
go back to reference Li Y, Xie J, Li X, et al. Poly (ADP-ribosylation) of HMGB1 facilitates its acetylation and promotes HMGB1 translocation-associated chemotherapy-induced autophagy in leukaemia cells. Oncol Lett. 2020;19(1):368–78.PubMed Li Y, Xie J, Li X, et al. Poly (ADP-ribosylation) of HMGB1 facilitates its acetylation and promotes HMGB1 translocation-associated chemotherapy-induced autophagy in leukaemia cells. Oncol Lett. 2020;19(1):368–78.PubMed
23.
go back to reference Kang R, Livesey KM, Zeh HR, et al. HMGB1 as an autophagy sensor in oxidative stress. Autophagy. 2011;7(8):904–6.PubMedCrossRef Kang R, Livesey KM, Zeh HR, et al. HMGB1 as an autophagy sensor in oxidative stress. Autophagy. 2011;7(8):904–6.PubMedCrossRef
24.
go back to reference Kong Q, Xu L, Xu W, et al. HMGB1 translocation is involved in the transformation of autophagy complexes and promotes chemoresistance in leukaemia. Int J Oncol. 2015;47(1):161–70.PubMedCrossRef Kong Q, Xu L, Xu W, et al. HMGB1 translocation is involved in the transformation of autophagy complexes and promotes chemoresistance in leukaemia. Int J Oncol. 2015;47(1):161–70.PubMedCrossRef
25.
go back to reference Andersson U, Yang H, Harris H. High-mobility group box 1 protein (HMGB1) operates as an alarmin outside as well as inside cells. Semin Immunol. 2018;38:40–8.PubMedCrossRef Andersson U, Yang H, Harris H. High-mobility group box 1 protein (HMGB1) operates as an alarmin outside as well as inside cells. Semin Immunol. 2018;38:40–8.PubMedCrossRef
26.
27.
go back to reference Jangde N, Ray R, Rai V. RAGE and its ligands: from pathogenesis to therapeutics. Crit Rev Biochem Mol Biol. 2020;55(6):555–75.PubMedCrossRef Jangde N, Ray R, Rai V. RAGE and its ligands: from pathogenesis to therapeutics. Crit Rev Biochem Mol Biol. 2020;55(6):555–75.PubMedCrossRef
28.
go back to reference Bucciarelli LG, Wendt T, Rong L, et al. RAGE is a multiligand receptor of the immunoglobulin superfamily: implications for homeostasis and chronic disease. Cell Mol Life Sci. 2002;59(7):1117–28.PubMedCrossRef Bucciarelli LG, Wendt T, Rong L, et al. RAGE is a multiligand receptor of the immunoglobulin superfamily: implications for homeostasis and chronic disease. Cell Mol Life Sci. 2002;59(7):1117–28.PubMedCrossRef
29.
go back to reference El-Far AH, Sroga G, Al Jaouni SK, et al. Role and mechanisms of RAGE-ligand complexes and RAGE-inhibitors in cancer progression. Int J Mol Sci. 2020;21(10):3613.PubMedCentralCrossRef El-Far AH, Sroga G, Al Jaouni SK, et al. Role and mechanisms of RAGE-ligand complexes and RAGE-inhibitors in cancer progression. Int J Mol Sci. 2020;21(10):3613.PubMedCentralCrossRef
30.
go back to reference Kang R, Tang D, Livesey KM, et al. The receptor for advanced glycation end-products (RAGE) protects pancreatic tumor cells against oxidative injury. Antioxid Redox Signal. 2011;15(8):2175–84.PubMedPubMedCentralCrossRef Kang R, Tang D, Livesey KM, et al. The receptor for advanced glycation end-products (RAGE) protects pancreatic tumor cells against oxidative injury. Antioxid Redox Signal. 2011;15(8):2175–84.PubMedPubMedCentralCrossRef
31.
go back to reference Fuentes MK, Nigavekar SS, Arumugam T, et al. RAGE activation by S100P in colon cancer stimulates growth, migration, and cell signaling pathways. Dis Colon Rectum. 2007;50(8):1230–40.PubMedCrossRef Fuentes MK, Nigavekar SS, Arumugam T, et al. RAGE activation by S100P in colon cancer stimulates growth, migration, and cell signaling pathways. Dis Colon Rectum. 2007;50(8):1230–40.PubMedCrossRef
32.
go back to reference Kang R, Tang D, Schapiro NE, et al. The receptor for advanced glycation end products (RAGE) sustains autophagy and limits apoptosis, promoting pancreatic tumor cell survival. Cell Death Differ. 2010;17(4):666–76.PubMedCrossRef Kang R, Tang D, Schapiro NE, et al. The receptor for advanced glycation end products (RAGE) sustains autophagy and limits apoptosis, promoting pancreatic tumor cell survival. Cell Death Differ. 2010;17(4):666–76.PubMedCrossRef
33.
go back to reference Bierhaus A, Humpert PM, Morcos M, et al. Understanding RAGE, the receptor for advanced glycation end products. J Mol Med. 2005;83(11):876–86.PubMedCrossRef Bierhaus A, Humpert PM, Morcos M, et al. Understanding RAGE, the receptor for advanced glycation end products. J Mol Med. 2005;83(11):876–86.PubMedCrossRef
34.
go back to reference Fletcher JI, Haber M, Henderson MJ, et al. ABC transporters in cancer: more than just drug efflux pumps. Nat Rev Cancer. 2010;10(2):147–56.PubMedCrossRef Fletcher JI, Haber M, Henderson MJ, et al. ABC transporters in cancer: more than just drug efflux pumps. Nat Rev Cancer. 2010;10(2):147–56.PubMedCrossRef
35.
go back to reference Fazlina N, Maha A, Zarina AL, et al. Assessment of P-gp and MRP1 activities using MultiDrugQuant Assay Kit: a preliminary study of correlation between protein expressions and its functional activities in newly diagnosed acute leukaemia patients. Malays J Pathol. 2008;30(2):87–93.PubMed Fazlina N, Maha A, Zarina AL, et al. Assessment of P-gp and MRP1 activities using MultiDrugQuant Assay Kit: a preliminary study of correlation between protein expressions and its functional activities in newly diagnosed acute leukaemia patients. Malays J Pathol. 2008;30(2):87–93.PubMed
36.
go back to reference Morrish E, Copeland A, Moujalled DM, et al. Clinical MDR1 inhibitors enhance Smac-mimetic bioavailability to kill murine LSCs and improve survival in AML models. Blood Adv. 2020;4(20):5062–77.PubMedPubMedCentralCrossRef Morrish E, Copeland A, Moujalled DM, et al. Clinical MDR1 inhibitors enhance Smac-mimetic bioavailability to kill murine LSCs and improve survival in AML models. Blood Adv. 2020;4(20):5062–77.PubMedPubMedCentralCrossRef
37.
go back to reference Legrand O, Simonin G, Beauchamp-Nicoud A, et al. Simultaneous activity of MRP1 and Pgp is correlated with in vitro resistance to daunorubicin and with in vivo resistance in adult acute myeloid leukemia. Blood. 1999;94(3):1046–56.PubMedCrossRef Legrand O, Simonin G, Beauchamp-Nicoud A, et al. Simultaneous activity of MRP1 and Pgp is correlated with in vitro resistance to daunorubicin and with in vivo resistance in adult acute myeloid leukemia. Blood. 1999;94(3):1046–56.PubMedCrossRef
38.
go back to reference Yin Y, Li W, Deng M, et al. Extracellular high mobility group box chromosomal protein 1 promotes drug resistance by increasing the expression of P-glycoprotein expression in gastric adenocarcinoma cells. Mol Med Rep. 2014;9(4):1439–43.PubMedCrossRef Yin Y, Li W, Deng M, et al. Extracellular high mobility group box chromosomal protein 1 promotes drug resistance by increasing the expression of P-glycoprotein expression in gastric adenocarcinoma cells. Mol Med Rep. 2014;9(4):1439–43.PubMedCrossRef
39.
go back to reference Zhao BX, Sun YB, Wang SQ, et al. Grape seed procyanidin reversal of p-glycoprotein associated multi-drug resistance via down-regulation of NF-kappaB and MAPK/ERK mediated YB-1 activity in A2780/T cells. PLoS ONE. 2013;8(8):e71071.PubMedPubMedCentralCrossRef Zhao BX, Sun YB, Wang SQ, et al. Grape seed procyanidin reversal of p-glycoprotein associated multi-drug resistance via down-regulation of NF-kappaB and MAPK/ERK mediated YB-1 activity in A2780/T cells. PLoS ONE. 2013;8(8):e71071.PubMedPubMedCentralCrossRef
40.
go back to reference Chen C, Lu L, Yan S, et al. Autophagy and doxorubicin resistance in cancer. Anticancer Drugs. 2018;29(1):1–9.PubMedCrossRef Chen C, Lu L, Yan S, et al. Autophagy and doxorubicin resistance in cancer. Anticancer Drugs. 2018;29(1):1–9.PubMedCrossRef
41.
go back to reference Muller S, Scaffidi P, Degryse B, et al. New EMBO members’ review: the double life of HMGB1 chromatin protein: architectural factor and extracellular signal. EMBO J. 2001;20(16):4337–40.PubMedPubMedCentralCrossRef Muller S, Scaffidi P, Degryse B, et al. New EMBO members’ review: the double life of HMGB1 chromatin protein: architectural factor and extracellular signal. EMBO J. 2001;20(16):4337–40.PubMedPubMedCentralCrossRef
43.
go back to reference Hori O, Brett J, Slattery T, et al. The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. J Biol Chem. 1995;270(43):25752.PubMedCrossRef Hori O, Brett J, Slattery T, et al. The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. J Biol Chem. 1995;270(43):25752.PubMedCrossRef
44.
go back to reference Kalea AZ, See F, Harja E, et al. Alternatively spliced RAGEv1 inhibits tumorigenesis through suppression of JNK signaling. Cancer Res. 2010;70(13):5628–38.PubMedPubMedCentralCrossRef Kalea AZ, See F, Harja E, et al. Alternatively spliced RAGEv1 inhibits tumorigenesis through suppression of JNK signaling. Cancer Res. 2010;70(13):5628–38.PubMedPubMedCentralCrossRef
45.
go back to reference Wu D, Ding Y, Wang S, et al. Increased expression of high mobility group box 1 (HMGB1) is associated with progression and poor prognosis in human nasopharyngeal carcinoma. J Pathol. 2008;216(2):167–75.PubMedCrossRef Wu D, Ding Y, Wang S, et al. Increased expression of high mobility group box 1 (HMGB1) is associated with progression and poor prognosis in human nasopharyngeal carcinoma. J Pathol. 2008;216(2):167–75.PubMedCrossRef
46.
go back to reference Kang R, Tang D, Lotze MT, et al. AGER/RAGE-mediated autophagy promotes pancreatic tumorigenesis and bioenergetics through the IL6-pSTAT3 pathway. Autophagy. 2012;8(6):989–91.PubMedPubMedCentralCrossRef Kang R, Tang D, Lotze MT, et al. AGER/RAGE-mediated autophagy promotes pancreatic tumorigenesis and bioenergetics through the IL6-pSTAT3 pathway. Autophagy. 2012;8(6):989–91.PubMedPubMedCentralCrossRef
47.
go back to reference Kang R, Livesey KM, Zeh IHJ, et al. Metabolic regulation by HMGB1-mediated autophagy and mitophagy. Autophagy. 2011;7(10):1256–8.PubMedCrossRef Kang R, Livesey KM, Zeh IHJ, et al. Metabolic regulation by HMGB1-mediated autophagy and mitophagy. Autophagy. 2011;7(10):1256–8.PubMedCrossRef
48.
49.
50.
go back to reference Liu L, Gao F, Ye Y, et al. Influence of HMGB1/MAPK/m-TOR signaling pathway on cell autophagy and chemotherapy resistance in K562 cells. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2016;41(10):1016–23.PubMed Liu L, Gao F, Ye Y, et al. Influence of HMGB1/MAPK/m-TOR signaling pathway on cell autophagy and chemotherapy resistance in K562 cells. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2016;41(10):1016–23.PubMed
51.
go back to reference Livesey KM, Kang R, Vernon P, et al. p53/HMGB1 complexes regulate autophagy and apoptosis. Canver Res. 2012;72(8):1996–2005.CrossRef Livesey KM, Kang R, Vernon P, et al. p53/HMGB1 complexes regulate autophagy and apoptosis. Canver Res. 2012;72(8):1996–2005.CrossRef
52.
go back to reference Aubrey BJ, Kelly GL, Janic A, et al. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2018;25(1):104–13.PubMedCrossRef Aubrey BJ, Kelly GL, Janic A, et al. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2018;25(1):104–13.PubMedCrossRef
53.
54.
go back to reference Kalea AZ, Reiniger N, Yang H, et al. Alternative splicing of the murine receptor for advanced glycation end-products (RAGE) gene. FASEB J. 2009;23(6):1766–74.PubMedPubMedCentralCrossRef Kalea AZ, Reiniger N, Yang H, et al. Alternative splicing of the murine receptor for advanced glycation end-products (RAGE) gene. FASEB J. 2009;23(6):1766–74.PubMedPubMedCentralCrossRef
55.
go back to reference Xie J, Méndez JD, Méndez-Valenzuela V, et al. Cellular signalling of the receptor for advanced glycation end products (RAGE). Cell Signal. 2013;25(11):2185–97.PubMedCrossRef Xie J, Méndez JD, Méndez-Valenzuela V, et al. Cellular signalling of the receptor for advanced glycation end products (RAGE). Cell Signal. 2013;25(11):2185–97.PubMedCrossRef
56.
go back to reference Yu L, Li L, Medeiros LJ, et al. NF-κB signaling pathway and its potential as a target for therapy in lymphoid neoplasms. Blood Rev. 2017;31(2):77–92.PubMedCrossRef Yu L, Li L, Medeiros LJ, et al. NF-κB signaling pathway and its potential as a target for therapy in lymphoid neoplasms. Blood Rev. 2017;31(2):77–92.PubMedCrossRef
57.
go back to reference Karin M, Greten FR. NF-κB: linking inflammation and immunity to cancer development and progression. Nat Rev Immunol. 2005;5(10):749–59.PubMedCrossRef Karin M, Greten FR. NF-κB: linking inflammation and immunity to cancer development and progression. Nat Rev Immunol. 2005;5(10):749–59.PubMedCrossRef
58.
Metadata
Title
Extracellular HMGB1 interacts with RAGE and promotes chemoresistance in acute leukemia cells
Authors
Weixin Lai
Xinyu Li
Qian Kong
Han Chen
Yunyao Li
Lu-Hong Xu
Jianpei Fang
Publication date
01-12-2021
Publisher
BioMed Central
Keyword
Leukemia
Published in
Cancer Cell International / Issue 1/2021
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-021-02387-9

Other articles of this Issue 1/2021

Cancer Cell International 1/2021 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