Skip to main content

Advertisement

Log in

Normoxically Overexpressed Hypoxia Inducible Factor 1-Alpha is Involved in Arsenic Trioxide Resistance Acquisition in Hepatocellular Carcinoma

  • Translational Research and Biomarkers
  • Published:
Annals of Surgical Oncology Aims and scope Submit manuscript

Abstract

Background

The aim of this study was to examine the underlying signaling mechanisms of arsenic trioxide (ATO)-mediated anticancer effects and the responsible biomarker(s) for the acquired resistance in human heptatocellular carcinoma (HCC).

Materials and Methods

The therapeutic effects of ATO were examined using 2 characteristically distinct HCC cell lines, Hep-J5 (overexpressing HIF-1α/GRP78) and SK-Hep-1 (the matched control). ATO-mediated proliferation inhibition, oxidative stress, and apoptosis were analyzed using flowcytometric analysis and western blotting. The role of HIF-1α and GRP78 in HCC resistance to ATO treatment was determined using RNA silencing and inhibitor approaches.

Results

SK-Hep-1 cells, lacking both HIF-1α and GRP78 expressions were responsive to ATO-induced apoptosis via an oxidative-nitrosative mechanism. Intracellular glutathione depletion and lipid peroxidation have been identified as the early cascade of events preceding apoptosis via cytochrome c release and the severe drop of mitochondrial membrane potential (MMP). Conversely, Hep-J5 cells, with normoxic coexpression of HIF-1α and GRP78, were resistant to ATO-induced apoptosis. GRP78-silenced Hep-J5 cells remained resistant to ATO treatment. In contrast, ATO resistance in Hep-J5 cells was overcome by the addition of YC-1, a HIF-1α inhibitor.

Conclusions

HIF-1α was identified as the major positive modifier for ATO resistance acquisition in HCC, and it represents a prime molecular target for overcoming ATO resistance.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, et al. Cancer statistics, 2008. CA Cancer J Clin. 2008;58:71–96.

    Article  PubMed  Google Scholar 

  2. Anthony PP. Hepatocellular carcinoma: an overview. Histopathology. 2001;39:109–18.

    Article  PubMed  CAS  Google Scholar 

  3. Badvie S. Hepatocellular carcinoma. Postgrad Med J. 2000;76:4–11.

    Article  PubMed  CAS  Google Scholar 

  4. Erler JT, Cawthorne CJ, Williams KJ, Koritzinsky M, Wouters BG, Wilson C, et al. Hypoxia-mediated down-regulation of Bid and Bax in tumors occurs via hypoxia-inducible factor 1-dependent and -independent mechanisms and contributes to drug resistance. Mol Cell Biol. 2004;24:2875–89.

    Article  PubMed  CAS  Google Scholar 

  5. Moeller BJ, Dewhirst MW. Raising the bar: how HIF-1 helps determine tumor radiosensitivity. Cell Cycle. 2004;3:1107–10.

    Article  PubMed  CAS  Google Scholar 

  6. Song J, Qu Z, Guo X, Zhao Q, Zhao X, Gao L, et al. Hypoxia-induced autophagy contributes to the chemoresistance of hepatocellular carcinoma cells. Autophagy. 2009;5:1131–44.

    Article  PubMed  CAS  Google Scholar 

  7. Wouters BG, Koritzinsky M. Hypoxia signalling through mTOR and the unfolded protein response in cancer. Nat Rev Cancer. 2008;8:851–64.

    Article  PubMed  CAS  Google Scholar 

  8. Chiou JF, Tai CJ, Huang MT, Wei PL, Wang YH, An J, et al. Glucose-regulated protein 78 is a novel contributor to acquisition of resistance to sorafenib in hepatocellular carcinoma. Ann Surg Oncol. 2010;17:603–12.

    Article  PubMed  Google Scholar 

  9. Lee AS. The glucose-regulated proteins: stress induction and clinical applications. Trends Biochem Sci. 2001;26:504–10.

    Article  PubMed  CAS  Google Scholar 

  10. Pyrko P, Schonthal AH, Hofman FM, Chen TC, Lee AS. The unfolded protein response regulator GRP78/BiP as a novel target for increasing chemosensitivity in malignant gliomas. Cancer Res. 2007;67:9809–16.

    Article  PubMed  CAS  Google Scholar 

  11. Daneshmand S, Quek ML, Lin E, Lee C, Cote RJ, Hawes D, et al. Glucose-regulated protein GRP78 is up-regulated in prostate cancer and correlates with recurrence and survival. Hum Pathol. 2007;38:1547–52.

    Article  PubMed  CAS  Google Scholar 

  12. Su R, Li Z, Li H, Song H, Bao C, Wei J, et al. Grp78 promotes the invasion of hepatocellular carcinoma. BMC Cancer. 2010;10:20.

    Article  PubMed  Google Scholar 

  13. Chang YJ, Chiu CC, Wu CH, An J, Wu CC, Liu TZ, et al. Glucose-regulated protein 78 (GRP78) silencing enhances cell migration but does not influence cell proliferation in hepatocellular carcinoma. Ann Surg Oncol. 2010;17:1703–9.

    Article  PubMed  Google Scholar 

  14. Berenson JR, Matous J, Swift RA, Mapes R, Morrison B, Yeh HS. A phase I/II study of arsenic trioxide/bortezomib/ascorbic acid combination therapy for the treatment of relapsed or refractory multiple myeloma. Clin Cancer Res. 2007;13:1762–8.

    Article  PubMed  CAS  Google Scholar 

  15. Pettersson HM, Pietras A, Munksgaard PM, Karlsson J, Johansson L, Shoshan MC, et al. Arsenic trioxide is highly cytotoxic to small cell lung carcinoma cells. Mol Cancer Ther. 2009;8:160–70.

    Article  PubMed  CAS  Google Scholar 

  16. Pettersson HM, Karlsson J, Pietras A, Ora I, Pahlman S. Arsenic trioxide and neuroblastoma cytotoxicity. J Bioenerg Biomembr. 2007;39:35–41.

    Article  PubMed  CAS  Google Scholar 

  17. Karlsson J, Pietras A, Beckman S, Pettersson HM, Larsson C, Pahlman S. Arsenic trioxide-induced neuroblastoma cell death is accompanied by proteolytic activation of nuclear Bax. Oncogene. 2007;26:6150–9.

    Article  PubMed  CAS  Google Scholar 

  18. Miller WH, Jr., Schipper HM, Lee JS, Singer J, Waxman S. Mechanisms of action of arsenic trioxide. Cancer Res. 2002;62:3893–903.

    PubMed  CAS  Google Scholar 

  19. Chen CY, Liu TZ, Liu YW, Tseng WC, Liu RH, Lu FJ, et al. 6-shogaol (alkanone from ginger) induces apoptotic cell death of human hepatoma p53 mutant Mahlavu subline via an oxidative stress-mediated caspase-dependent mechanism. J Agric Food Chem. 2007;55:948–54.

    Article  PubMed  CAS  Google Scholar 

  20. Hsieh TJ, Liu TZ, Lu FJ, Hsieh PY, Chen CH. Actinodaphnine induces apoptosis through increased nitric oxide, reactive oxygen species and down-regulation of NF-kappaB signaling in human hepatoma Mahlavu cells. Food Chem Toxicol. 2006;44:344–54.

    Article  PubMed  CAS  Google Scholar 

  21. Fei M, Lu M, Wang Y, Zhao Y, He S, Gao S, et al. Arsenic trioxide-induced growth arrest of human hepatocellular carcinoma cells involving FOXO3a expression and localization. Med Oncol. 2009;26:178–85.

    Article  PubMed  CAS  Google Scholar 

  22. Liu XM, Xiong XF, Song Y, Tang RJ, Liang XQ, Cao EH. Possible roles of a tumor suppressor gene PIG11 in hepatocarcinogenesis and As2O3-induced apoptosis in liver cancer cells. J Gastroenterol. 2009;44:460–9.

    Article  PubMed  CAS  Google Scholar 

  23. Zhao XS, Song PL, Sun B, Jiang HC, Liu TF. Arsenic trioxide inhibits metastatic potential of mouse hepatoma H22 cells in vitro and in vivo. Hepatobiliary Pancreat Dis Int. 2009;8:510–7.

    PubMed  CAS  Google Scholar 

  24. Rao RV, Peel A, Logvinova A, Del RG, Hermel E, Yokota T, et al. Coupling endoplasmic reticulum stress to the cell death program: role of the ER chaperone GRP78. FEBS Lett. 2002;514:122–8.

    PubMed  CAS  Google Scholar 

  25. Reddy RK, Mao C, Baumeister P, Austin RC, Kaufman RJ, Lee AS. Endoplasmic reticulum chaperone protein GRP78 protects cells from apoptosis induced by topoisomerase inhibitors: role of ATP binding site in suppression of caspase-7 activation. J Biol Chem. 2003;278:20915–24.

    Article  PubMed  CAS  Google Scholar 

  26. Kroemer G, De TH. Arsenic trioxide, a novel mitochondriotoxic anticancer agent? J Natl Cancer Inst. 1999;91:743–5.

    Article  PubMed  CAS  Google Scholar 

  27. Lu J, Chew EH, Holmgren A. Targeting thioredoxin reductase is a basis for cancer therapy by arsenic trioxide. Proc Natl Acad Sci USA. 2007;104:12288–93.

    Article  PubMed  CAS  Google Scholar 

  28. Papandreou I, Cairns RA, Fontana L, Lim AL, Denko NC. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. 2006;3:187–97.

    Article  PubMed  CAS  Google Scholar 

  29. Chandel NS, Schumacker PT. Cellular oxygen sensing by mitochondria: old questions, new insight. J Appl Physiol. 2000;88:1880–9.

    Article  PubMed  CAS  Google Scholar 

  30. Fukuda R, Zhang H, Kim JW, Shimoda L, Dang CV, Semenza GL. HIF-1 regulates cytochrome oxidase subunits to optimize efficiency of respiration in hypoxic cells. Cell. 2007;129:111–22.

    Article  PubMed  CAS  Google Scholar 

  31. Kim JW, Tchernyshyov I, Semenza GL, Dang CV. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 2006;3:177–85.

    Article  PubMed  Google Scholar 

  32. Zhang H, Gao P, Fukuda R, Kumar G, Krishnamachary B, Zeller KI, et al. HIF-1 inhibits mitochondrial biogenesis and cellular respiration in VHL-deficient renal cell carcinoma by repression of C-MYC activity. Cancer Cell. 2007;11:407–20.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgment

This work is supported by National Science Council (NSC 99-2628-B-038-010-MY3) and TMU young investigator grant (TMU97-AE-1-B08) to ATHW and the Translational Research Laboratory, Cancer Center, Taipei Medical University Hospital. The other funding agency is Center of Excellence for Cancer Research, Taipei Medical University (TMU-CECR, DOH99-TD-C-111-008).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Jeng-Fong Chiou MD, PhD or Alexander T. H. Wu PhD.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Fig. 1

GRP78 is not the major contributor to ATO resistance in J5 cells. Both control (vc-J5) and GRP78-silenced (si-J5) J5 cells were subjected to 30 μM ATO treatment for 48 and 72 h. Both groups remained resistant to ATO treatment. Experiments were performed in triplicate (EPS 1247 kb)

Supplementary Fig. 2

HIF-1α and GRP78 expression profile in 5 selected HCC cell lines. Western blotting analysis was performed on 5 HCC cell lines namely, HepG2 (G2), Hep3B (3B), HepJ5 (J5), Mahlavu (Ma), and Sk-Hep1. J5 cells uniquely demonstrated a significantly elevated expression in both HIF-1α and GRP78 under normoxic condition when compared with the other cell lines (EPS 1153 kb)

Supplementary Fig. 3

Control TUNEL assay. a SK cells were treated with HIF-1α inhibitor (YC-1) as a control. YC-1 treatment had minimal cytotoxic effect (approximately 5.7% cells underwent apoptosis) on SK cells without the addition of ATO treatment. b J5 cells which received YC-1 lone treatment did not show significant apoptotic signal (approximately 4.9% apoptotic population) as demonstrated by TUNEL assay (EPS 879 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Tung, JN., Cheng, YW., Hsu, CH. et al. Normoxically Overexpressed Hypoxia Inducible Factor 1-Alpha is Involved in Arsenic Trioxide Resistance Acquisition in Hepatocellular Carcinoma. Ann Surg Oncol 18, 1492–1500 (2011). https://doi.org/10.1245/s10434-010-1444-y

Download citation

  • Received:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1245/s10434-010-1444-y

Keywords

Navigation