Skip to main content
Top
Published in: BMC Medicine 1/2022

Open Access 01-12-2022 | Ionizing Radiation | Research article

NUPR1 contributes to radiation resistance by maintaining ROS homeostasis via AhR/CYP signal axis in hepatocellular carcinoma

Authors: Yizhi Zhan, Zhanqiao Zhang, Yuechen Liu, Yuan Fang, Yuwen Xie, Yilin Zheng, Guoxin Li, Li Liang, Yi Ding

Published in: BMC Medicine | Issue 1/2022

Login to get access

Abstract

Background

Radiotherapy (RT) is one of the major therapeutic approaches to hepatocellular carcinoma (HCC). Ionizing radiation (IR) inducing the generation of reactive oxygen species (ROS) leads to a promising antitumor effect. However, the dysregulation of the redox system often causes radioresistance and impairs the efficacy of RT. Increasing evidence indicates that nuclear protein 1 (NUPR1) plays a critical role in redox reactions. In this study, we aim to explore the role of NUPR1 in maintaining ROS homeostasis and radioresistance in HCC.

Methods

The radioresistant role of NUPR1 was determined by colony formation assay, comet assay in vitro, and xenograft tumor models in vivo. Probes for ROS, apoptosis assay, and lipid peroxidation assay were used to investigate the functional effect of NUPR1 on ROS homeostasis and oxidative stress. RNA sequencing and co-immunoprecipitation assay were performed to clarify the mechanism of NUPR1 inhibiting the AhR/CYP signal axis. Finally, we analyzed clinical specimens to assess the predictive value of NUPR1 and AhR in the radiotherapeutic efficacy of HCC.

Results

We demonstrated that NUPR1 was upregulated in HCC tissues and verified that NUPR1 increased the radioresistance of HCC in vitro and in vivo. NUPR1 alleviated the generation of ROS and suppressed oxidative stress, including apoptosis and lipid peroxidation by downregulating cytochrome P450 (CYP) upon IR. ROS scavenger N-acetyl-L-cysteine (NAC) and CYP inhibitor alizarin restored the viability of NUPR1-knockdown cells during IR. Mechanistically, the interaction between NUPR1 and aryl hydrocarbon receptor (AhR) promoted the degradation and decreased nuclear translation of AhR via the autophagy-lysosome pathway, followed by being incapable of CYP’s transcription. Furthermore, genetically and pharmacologically activating AhR abrogated the radioresistant role of NUPR1. Clinical data suggested that NUPR1 and AhR could serve as novel biomarkers for predicting the radiation response of HCC.

Conclusions

Our findings revealed the role of NUPR1 in regulating ROS homeostasis and oxidative stress via the AhR/CYP signal axis upon IR. Strategies targeting the NUPR1/AhR/CYP pathway may have important clinical applications for improving the radiotherapeutic efficacy of HCC.
Appendix
Available only for authorised users
Literature
1.
go back to reference Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.
2.
go back to reference Ursino S, Greco C, Cartei F, Colosimo C, Stefanelli A, Cacopardo B, et al. Radiotherapy and hepatocellular carcinoma: update and review of the literature. Eur Rev Med Pharmacol Sci. 2012;16(11):1599–604.PubMed Ursino S, Greco C, Cartei F, Colosimo C, Stefanelli A, Cacopardo B, et al. Radiotherapy and hepatocellular carcinoma: update and review of the literature. Eur Rev Med Pharmacol Sci. 2012;16(11):1599–604.PubMed
3.
go back to reference Klein J, Dawson LA. Hepatocellular carcinoma radiation therapy: review of evidence and future opportunities. Int J Radiat Oncol Biol Phys. 2013;87(1):22–32.PubMedCrossRef Klein J, Dawson LA. Hepatocellular carcinoma radiation therapy: review of evidence and future opportunities. Int J Radiat Oncol Biol Phys. 2013;87(1):22–32.PubMedCrossRef
4.
go back to reference Wahl DR, Stenmark MH, Tao Y, Pollom EL, Caoili EM, Lawrence TS, et al. Outcomes After Stereotactic Body Radiotherapy or Radiofrequency Ablation for Hepatocellular Carcinoma. J Clin Oncol. 2016;34(5):452–9.PubMedCrossRef Wahl DR, Stenmark MH, Tao Y, Pollom EL, Caoili EM, Lawrence TS, et al. Outcomes After Stereotactic Body Radiotherapy or Radiofrequency Ablation for Hepatocellular Carcinoma. J Clin Oncol. 2016;34(5):452–9.PubMedCrossRef
5.
go back to reference Yoon HI, Seong J. Multimodality treatment involving radiotherapy for advanced liver-confined hepatocellular carcinoma. Oncology. 2014;87(Suppl 1):90–8.PubMedCrossRef Yoon HI, Seong J. Multimodality treatment involving radiotherapy for advanced liver-confined hepatocellular carcinoma. Oncology. 2014;87(Suppl 1):90–8.PubMedCrossRef
6.
go back to reference Fang Y, Zhan Y, Xie Y, Du S, Chen Y, Zeng Z, et al. Integration of glucose and cardiolipin anabolism confers radiation resistance of HCC. Hepatology. 2022;75(6):1386–401.PubMedCrossRef Fang Y, Zhan Y, Xie Y, Du S, Chen Y, Zeng Z, et al. Integration of glucose and cardiolipin anabolism confers radiation resistance of HCC. Hepatology. 2022;75(6):1386–401.PubMedCrossRef
7.
go back to reference Zou Z, Chang H, Li H, Wang S. Induction of reactive oxygen species: an emerging approach for cancer therapy. Apoptosis. 2017;22(11):1321–35.PubMedCrossRef Zou Z, Chang H, Li H, Wang S. Induction of reactive oxygen species: an emerging approach for cancer therapy. Apoptosis. 2017;22(11):1321–35.PubMedCrossRef
8.
go back to reference Kim W, Lee S, Seo D, Kim D, Kim K, Kim E, et al. Cellular Stress Responses in Radiotherapy. Cells. 2019;8:9.CrossRef Kim W, Lee S, Seo D, Kim D, Kim K, Kim E, et al. Cellular Stress Responses in Radiotherapy. Cells. 2019;8:9.CrossRef
9.
10.
go back to reference Kim W, Youn H, Kang C, Youn B. Inflammation-induced radioresistance is mediated by ROS-dependent inactivation of protein phosphatase 1 in non-small cell lung cancer cells. Apoptosis. 2015;20(9):1242–52.PubMedCrossRef Kim W, Youn H, Kang C, Youn B. Inflammation-induced radioresistance is mediated by ROS-dependent inactivation of protein phosphatase 1 in non-small cell lung cancer cells. Apoptosis. 2015;20(9):1242–52.PubMedCrossRef
11.
go back to reference Lu L, Dong J, Wang L, Xia Q, Zhang D, Kim H, et al. Activation of STAT3 and Bcl-2 and reduction of reactive oxygen species (ROS) promote radioresistance in breast cancer and overcome of radioresistance with niclosamide. Oncogene. 2018;37(39):5292–304.PubMedCrossRef Lu L, Dong J, Wang L, Xia Q, Zhang D, Kim H, et al. Activation of STAT3 and Bcl-2 and reduction of reactive oxygen species (ROS) promote radioresistance in breast cancer and overcome of radioresistance with niclosamide. Oncogene. 2018;37(39):5292–304.PubMedCrossRef
12.
go back to reference Liu R, Li W, Tao B, Wang X, Yang Z, Zhang Y, et al. Tyrosine phosphorylation activates 6-phosphogluconate dehydrogenase and promotes tumor growth and radiation resistance. Nat Commun. 2019;10(1):991.PubMedPubMedCentralCrossRef Liu R, Li W, Tao B, Wang X, Yang Z, Zhang Y, et al. Tyrosine phosphorylation activates 6-phosphogluconate dehydrogenase and promotes tumor growth and radiation resistance. Nat Commun. 2019;10(1):991.PubMedPubMedCentralCrossRef
13.
go back to reference Goruppi S, Iovanna JL. Stress-inducible protein p8 is involved in several physiological and pathological processes. J Biol Chem. 2010;285(3):1577–81.PubMedCrossRef Goruppi S, Iovanna JL. Stress-inducible protein p8 is involved in several physiological and pathological processes. J Biol Chem. 2010;285(3):1577–81.PubMedCrossRef
15.
go back to reference Clark DW, Mitra A, Fillmore RA, Jiang WG, Samant RS, Fodstad O, et al. NUPR1 interacts with p53, transcriptionally regulates p21 and rescues breast epithelial cells from doxorubicin-induced genotoxic stress. Curr Cancer Drug Targets. 2008;8(5):421–30.PubMedCrossRef Clark DW, Mitra A, Fillmore RA, Jiang WG, Samant RS, Fodstad O, et al. NUPR1 interacts with p53, transcriptionally regulates p21 and rescues breast epithelial cells from doxorubicin-induced genotoxic stress. Curr Cancer Drug Targets. 2008;8(5):421–30.PubMedCrossRef
16.
go back to reference Emma MR, Iovanna JL, Bachvarov D, Puleio R, Loria GR, Augello G, et al. NUPR1, a new target in liver cancer: implication in controlling cell growth, migration, invasion and sorafenib resistance. Cell Death Dis. 2016;7(6):e2269.PubMedPubMedCentralCrossRef Emma MR, Iovanna JL, Bachvarov D, Puleio R, Loria GR, Augello G, et al. NUPR1, a new target in liver cancer: implication in controlling cell growth, migration, invasion and sorafenib resistance. Cell Death Dis. 2016;7(6):e2269.PubMedPubMedCentralCrossRef
17.
go back to reference Li A, Li X, Chen X, Zeng C, Wang Z, Li Z, et al. NUPR1 Silencing Induces Autophagy-Mediated Apoptosis in Multiple Myeloma Cells Through the PI3K/AKT/mTOR Pathway. DNA Cell Biol. 2020;39(3):368–78.PubMedCrossRef Li A, Li X, Chen X, Zeng C, Wang Z, Li Z, et al. NUPR1 Silencing Induces Autophagy-Mediated Apoptosis in Multiple Myeloma Cells Through the PI3K/AKT/mTOR Pathway. DNA Cell Biol. 2020;39(3):368–78.PubMedCrossRef
18.
go back to reference Mu Y, Yan X, Li D, Zhao D, Wang L, Wang X, et al. NUPR1 maintains autolysosomal efflux by activating SNAP25 transcription in cancer cells. Autophagy. 2018;14(4):654–70.PubMedCrossRef Mu Y, Yan X, Li D, Zhao D, Wang L, Wang X, et al. NUPR1 maintains autolysosomal efflux by activating SNAP25 transcription in cancer cells. Autophagy. 2018;14(4):654–70.PubMedCrossRef
19.
go back to reference Santofimia-Castano P, Lan W, Bintz J, Gayet O, Carrier A, Lomberk G, et al. Inactivation of NUPR1 promotes cell death by coupling ER-stress responses with necrosis. Sci Rep. 2018;8(1):16999.PubMedPubMedCentralCrossRef Santofimia-Castano P, Lan W, Bintz J, Gayet O, Carrier A, Lomberk G, et al. Inactivation of NUPR1 promotes cell death by coupling ER-stress responses with necrosis. Sci Rep. 2018;8(1):16999.PubMedPubMedCentralCrossRef
20.
go back to reference Weis S, Bielow T, Sommerer I, Iovanna J, Malicet C, Mössner J, et al. P8 deficiency increases cellular ROS and induces HO-1. Arch Biochem Biophys. 2015;565:89–94.PubMedCrossRef Weis S, Bielow T, Sommerer I, Iovanna J, Malicet C, Mössner J, et al. P8 deficiency increases cellular ROS and induces HO-1. Arch Biochem Biophys. 2015;565:89–94.PubMedCrossRef
21.
go back to reference Hamidi T, Cano CE, Grasso D, Garcia MN, Sandi MJ, Calvo EL, et al. Nupr1-aurora kinase A pathway provides protection against metabolic stress-mediated autophagic-associated cell death. Clin Cancer Res. 2012;18(19):5234–46.PubMedCrossRef Hamidi T, Cano CE, Grasso D, Garcia MN, Sandi MJ, Calvo EL, et al. Nupr1-aurora kinase A pathway provides protection against metabolic stress-mediated autophagic-associated cell death. Clin Cancer Res. 2012;18(19):5234–46.PubMedCrossRef
23.
go back to reference Gironella M, Malicet C, Cano C, Sandi MJ, Hamidi T, Tauil RM, et al. p8/nupr1 regulates DNA-repair activity after double-strand gamma irradiation-induced DNA damage. J Cell Physiol. 2009;221(3):594–602.PubMedCrossRef Gironella M, Malicet C, Cano C, Sandi MJ, Hamidi T, Tauil RM, et al. p8/nupr1 regulates DNA-repair activity after double-strand gamma irradiation-induced DNA damage. J Cell Physiol. 2009;221(3):594–602.PubMedCrossRef
24.
go back to reference Lan W, Santofimia-Castano P, Swayden M, Xia Y, Zhou Z, Audebert S, et al. ZZW-115-dependent inhibition of NUPR1 nuclear translocation sensitizes cancer cells to genotoxic agents. JCI Insight. 2020;5(18):e138117. Lan W, Santofimia-Castano P, Swayden M, Xia Y, Zhou Z, Audebert S, et al. ZZW-115-dependent inhibition of NUPR1 nuclear translocation sensitizes cancer cells to genotoxic agents. JCI Insight. 2020;5(18):e138117.
25.
go back to reference Veith A, Moorthy B. Role of Cytochrome P450s in the Generation and Metabolism of Reactive Oxygen Species. Curr Opin Toxicol. 2018;7:44–51.PubMedCrossRef Veith A, Moorthy B. Role of Cytochrome P450s in the Generation and Metabolism of Reactive Oxygen Species. Curr Opin Toxicol. 2018;7:44–51.PubMedCrossRef
26.
go back to reference Takahashi E, Fujita K, Kamataki T, Arimoto-Kobayashi S, Okamoto K, Negishi T. Inhibition of human cytochrome P450 1B1, 1A1 and 1A2 by antigenotoxic compounds, purpurin and alizarin. Mutat Res. 2002;508(1-2):147–56.PubMedCrossRef Takahashi E, Fujita K, Kamataki T, Arimoto-Kobayashi S, Okamoto K, Negishi T. Inhibition of human cytochrome P450 1B1, 1A1 and 1A2 by antigenotoxic compounds, purpurin and alizarin. Mutat Res. 2002;508(1-2):147–56.PubMedCrossRef
27.
go back to reference Nguyen LP, Bradfield CA. The search for endogenous activators of the aryl hydrocarbon receptor. Chem Res Toxicol. 2008;21(1):102–16.PubMedCrossRef Nguyen LP, Bradfield CA. The search for endogenous activators of the aryl hydrocarbon receptor. Chem Res Toxicol. 2008;21(1):102–16.PubMedCrossRef
28.
go back to reference Lindsey S, Papoutsakis ET. The evolving role of the aryl hydrocarbon receptor (AHR) in the normophysiology of hematopoiesis. Stem Cell Rev Rep. 2012;8(4):1223–35.PubMedCrossRef Lindsey S, Papoutsakis ET. The evolving role of the aryl hydrocarbon receptor (AHR) in the normophysiology of hematopoiesis. Stem Cell Rev Rep. 2012;8(4):1223–35.PubMedCrossRef
29.
go back to reference Augello G, Emma MR, Azzolina A, Puleio R, Condorelli L, Cusimano A, et al. The NUPR1/p73 axis contributes to sorafenib resistance in hepatocellular carcinoma. Cancer Lett. 2021;519:250–62.PubMedCrossRef Augello G, Emma MR, Azzolina A, Puleio R, Condorelli L, Cusimano A, et al. The NUPR1/p73 axis contributes to sorafenib resistance in hepatocellular carcinoma. Cancer Lett. 2021;519:250–62.PubMedCrossRef
30.
go back to reference Yu L, Chen Y, Tooze SA. Autophagy pathway: Cellular and molecular mechanisms. Autophagy. 2018;14(2):207–15.PubMedCrossRef Yu L, Chen Y, Tooze SA. Autophagy pathway: Cellular and molecular mechanisms. Autophagy. 2018;14(2):207–15.PubMedCrossRef
31.
go back to reference Wei J, Wang B, Wang H, Meng L, Zhao Q, Li X, et al. Radiation-Induced Normal Tissue Damage: Oxidative Stress and Epigenetic Mechanisms. Oxidative Med Cell Longev. 2019;2019:3010342.CrossRef Wei J, Wang B, Wang H, Meng L, Zhao Q, Li X, et al. Radiation-Induced Normal Tissue Damage: Oxidative Stress and Epigenetic Mechanisms. Oxidative Med Cell Longev. 2019;2019:3010342.CrossRef
32.
go back to reference Rendic S, Guengerich FP. Summary of information on the effects of ionizing and non-ionizing radiation on cytochrome P450 and other drug metabolizing enzymes and transporters. Curr Drug Metab. 2012;13(6):787–814.PubMedPubMedCentralCrossRef Rendic S, Guengerich FP. Summary of information on the effects of ionizing and non-ionizing radiation on cytochrome P450 and other drug metabolizing enzymes and transporters. Curr Drug Metab. 2012;13(6):787–814.PubMedPubMedCentralCrossRef
33.
go back to reference Chen Y, Li N, Wang J, Zhang X, Pan W, Yu L, et al. Enhancement of mitochondrial ROS accumulation and radiotherapeutic efficacy using a Gd-doped titania nanosensitizer. Theranostics. 2019;9(1):167–78.PubMedPubMedCentralCrossRef Chen Y, Li N, Wang J, Zhang X, Pan W, Yu L, et al. Enhancement of mitochondrial ROS accumulation and radiotherapeutic efficacy using a Gd-doped titania nanosensitizer. Theranostics. 2019;9(1):167–78.PubMedPubMedCentralCrossRef
34.
go back to reference Storch K, Dickreuter E, Artati A, Adamski J, Cordes N. BEMER Electromagnetic Field Therapy Reduces Cancer Cell Radioresistance by Enhanced ROS Formation and Induced DNA Damage. PLoS One. 2016;11(12):e0167931.PubMedPubMedCentralCrossRef Storch K, Dickreuter E, Artati A, Adamski J, Cordes N. BEMER Electromagnetic Field Therapy Reduces Cancer Cell Radioresistance by Enhanced ROS Formation and Induced DNA Damage. PLoS One. 2016;11(12):e0167931.PubMedPubMedCentralCrossRef
35.
go back to reference Nebert DW, Dalton TP. The role of cytochrome P450 enzymes in endogenous signalling pathways and environmental carcinogenesis. Nat Rev Cancer. 2006;6(12):947–60.PubMedCrossRef Nebert DW, Dalton TP. The role of cytochrome P450 enzymes in endogenous signalling pathways and environmental carcinogenesis. Nat Rev Cancer. 2006;6(12):947–60.PubMedCrossRef
36.
go back to reference Loida PJ, Sligar SG. Molecular recognition in cytochrome P-450: mechanism for the control of uncoupling reactions. Biochemistry. 1993;32(43):11530–8.PubMedCrossRef Loida PJ, Sligar SG. Molecular recognition in cytochrome P-450: mechanism for the control of uncoupling reactions. Biochemistry. 1993;32(43):11530–8.PubMedCrossRef
37.
go back to reference Zangar RC, Davydov DR, Verma S. Mechanisms that regulate production of reactive oxygen species by cytochrome P450. Toxicol Appl Pharmacol. 2004;199(3):316–31.PubMedCrossRef Zangar RC, Davydov DR, Verma S. Mechanisms that regulate production of reactive oxygen species by cytochrome P450. Toxicol Appl Pharmacol. 2004;199(3):316–31.PubMedCrossRef
38.
go back to reference Zou Y, Li H, Graham ET, Deik AA, Eaton JK, Wang W, et al. Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat Chem Biol. 2020;16(3):302–9.PubMedPubMedCentralCrossRef Zou Y, Li H, Graham ET, Deik AA, Eaton JK, Wang W, et al. Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat Chem Biol. 2020;16(3):302–9.PubMedPubMedCentralCrossRef
39.
go back to reference Son B, Kwon T, Lee S, Han I, Kim W, Youn H, et al. CYP2E1 regulates the development of radiation-induced pulmonary fibrosis via ER stress- and ROS-dependent mechanisms. Am J Phys Lung Cell Mol Phys. 2017;313(5):L916–29. Son B, Kwon T, Lee S, Han I, Kim W, Youn H, et al. CYP2E1 regulates the development of radiation-induced pulmonary fibrosis via ER stress- and ROS-dependent mechanisms. Am J Phys Lung Cell Mol Phys. 2017;313(5):L916–29.
40.
go back to reference Novikov O, Wang Z, Stanford EA, Parks AJ, Ramirez-Cardenas A, Landesman E, et al. An Aryl Hydrocarbon Receptor-Mediated Amplification Loop That Enforces Cell Migration in ER-/PR-/Her2- Human Breast Cancer Cells. Mol Pharmacol. 2016;90(5):674–88.PubMedPubMedCentralCrossRef Novikov O, Wang Z, Stanford EA, Parks AJ, Ramirez-Cardenas A, Landesman E, et al. An Aryl Hydrocarbon Receptor-Mediated Amplification Loop That Enforces Cell Migration in ER-/PR-/Her2- Human Breast Cancer Cells. Mol Pharmacol. 2016;90(5):674–88.PubMedPubMedCentralCrossRef
41.
go back to reference Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S, et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature. 2011;478(7368):197–203.PubMedCrossRef Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S, et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature. 2011;478(7368):197–203.PubMedCrossRef
42.
go back to reference Sadik A, Somarribas Patterson LF, Ozturk S, Mohapatra SR, Panitz V, Secker PF, et al. IL4I1 Is a Metabolic Immune Checkpoint that Activates the AHR and Promotes Tumor Progression. Cell. 2020;182(5):1252–1270 e1234.PubMedCrossRef Sadik A, Somarribas Patterson LF, Ozturk S, Mohapatra SR, Panitz V, Secker PF, et al. IL4I1 Is a Metabolic Immune Checkpoint that Activates the AHR and Promotes Tumor Progression. Cell. 2020;182(5):1252–1270 e1234.PubMedCrossRef
43.
go back to reference Smirnova A, Wincent E, Vikstrom Bergander L, Alsberg T, Bergman J, Rannug A, et al. Evidence for New Light-Independent Pathways for Generation of the Endogenous Aryl Hydrocarbon Receptor Agonist FICZ. Chem Res Toxicol. 2016;29(1):75–86.PubMedCrossRef Smirnova A, Wincent E, Vikstrom Bergander L, Alsberg T, Bergman J, Rannug A, et al. Evidence for New Light-Independent Pathways for Generation of the Endogenous Aryl Hydrocarbon Receptor Agonist FICZ. Chem Res Toxicol. 2016;29(1):75–86.PubMedCrossRef
44.
go back to reference Vogel CFA, Van Winkle LS, Esser C, Haarmann-Stemmann T. The aryl hydrocarbon receptor as a target of environmental stressors - Implications for pollution mediated stress and inflammatory responses. Redox Biol. 2020;34:101530.PubMedPubMedCentralCrossRef Vogel CFA, Van Winkle LS, Esser C, Haarmann-Stemmann T. The aryl hydrocarbon receptor as a target of environmental stressors - Implications for pollution mediated stress and inflammatory responses. Redox Biol. 2020;34:101530.PubMedPubMedCentralCrossRef
45.
go back to reference Paris A, Tardif N, Galibert MD, Corre S. AhR and Cancer: From Gene Profiling to Targeted Therapy. Int J Mol Sci. 2021;22(2):752. Paris A, Tardif N, Galibert MD, Corre S. AhR and Cancer: From Gene Profiling to Targeted Therapy. Int J Mol Sci. 2021;22(2):752.
46.
go back to reference Pappas B, Yang Y, Wang Y, Kim K, Chung HJ, Cheung M, et al. p23 protects the human aryl hydrocarbon receptor from degradation via a heat shock protein 90-independent mechanism. Biochem Pharmacol. 2018;152:34–44.PubMedPubMedCentralCrossRef Pappas B, Yang Y, Wang Y, Kim K, Chung HJ, Cheung M, et al. p23 protects the human aryl hydrocarbon receptor from degradation via a heat shock protein 90-independent mechanism. Biochem Pharmacol. 2018;152:34–44.PubMedPubMedCentralCrossRef
47.
go back to reference Shivanna B, Chu C, Moorthy B. The Aryl Hydrocarbon Receptor (AHR): A Novel Therapeutic Target for Pulmonary Diseases? Int J Mol Sci. 2022;23(3):1516. Shivanna B, Chu C, Moorthy B. The Aryl Hydrocarbon Receptor (AHR): A Novel Therapeutic Target for Pulmonary Diseases? Int J Mol Sci. 2022;23(3):1516.
Metadata
Title
NUPR1 contributes to radiation resistance by maintaining ROS homeostasis via AhR/CYP signal axis in hepatocellular carcinoma
Authors
Yizhi Zhan
Zhanqiao Zhang
Yuechen Liu
Yuan Fang
Yuwen Xie
Yilin Zheng
Guoxin Li
Li Liang
Yi Ding
Publication date
01-12-2022
Publisher
BioMed Central
Published in
BMC Medicine / Issue 1/2022
Electronic ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-022-02554-3

Other articles of this Issue 1/2022

BMC Medicine 1/2022 Go to the issue