DOI QR코드

DOI QR Code

The Effect of Lipopolysaccharide on Noxa Expression Is Mediated through IRF1, 3, and 7

  • Piya, Sujan (Department of Leukemia Section of Molecular Hematology and Therapy, The University of Texas) ;
  • Kim, Tae-Hyoung (Department of Biochemistry, Chosun University School of Medicine)
  • Received : 2017.08.25
  • Accepted : 2018.01.04
  • Published : 2018.03.28

Abstract

Lipopolysaccharide (LPS), a component of the cell wall of gram-negative bacteria, elicits the secretion of cytokines, such as interferons, that stimulate the host defense system. Previously, we demonstrated that interferons induce interferon regulatory factors (IRFs) 1, 3, and 7, which regulate the transcription of Noxa and alter the expression profiles of Bcl-2 family proteins in tumors. However, the immediate consequences of LPS stimulation on Noxa and BH3 expression in tumor cells remain uncharacterized. In this study, we determined that LPS induced Noxa expression in CT26 cells. Furthermore, studies in HCT116 parental and HCT116 p53-deficient cells revealed that LPS-mediated Noxa was independent of p53. Meanwhile, IRF1, 3, and 7 in CT26, HCT116 parental, and HT116 p53-deficient cells were upregulated by LPS stimulation, suggesting that LPS induces the expression of these IRFs in a p53-independent manner. The responsiveness of IRF1, 3, 4, and 7 binding to the Noxa promoter region to LPS indicated that IRF1, 3, and 7 activated Noxa expression, whereas IRF4 repressed Noxa expression. Together, these results suggest that LPS directly affects Noxa expression in tumor cells through IRFs, implicating that it may contribute to LPS-induced tumor regression.

Keywords

References

  1. Freudenberg MA, Tchaptchet S, Keck S, Fejer G, Huber M, Schutze N, et al. 2008. Lipopolysaccharide sensing an important factor in the innate immune response to gram-negative bacterial infections: benefits and hazards of LPS hypersensitivity. Immunobiology 213: 193-203. https://doi.org/10.1016/j.imbio.2007.11.008
  2. Won EK, Zahner MC, Grant EA, Gore P, Chicoine MR. 2003. Analysis of the antitumoral mechanisms of lipopolysaccharide against glioblastoma multiforme. Anticancer Drugs 14: 457-466. https://doi.org/10.1097/00001813-200307000-00012
  3. Chicoine MR, Won EK, Zahner MC. 2001. Intratumoral injection of lipopolysaccharide causes regression of subcutaneously implanted mouse glioblastoma multiforme. Neurosurgery 48: 607-614; discussion 614-615. https://doi.org/10.1097/00006123-200103000-00032
  4. Inagawa H, Nishizawa T, Honda T, Nakamoto T, Takagi K, Soma G. 1998. Mechanisms by which chemotherapeutic agents augment the antitumor effects of tumor necrosis factor: involvement of the pattern shift of cytokines from Th2 to Th1 in tumor lesions. Anticancer Res. 18: 3957-3964.
  5. Goto S, Sakai S, Kera J, Suma Y, Soma GI, Takeuchi S. 1996. Intradermal administration of lipopolysaccharide in treatment of human cancer. Cancer Immunol. Immunother. 42: 255-261. https://doi.org/10.1007/s002620050279
  6. Dighe AS, Richards E, Old LJ, Schreiber RD. 1994. Enhanced in vivo growth and resistance to rejection of tumor cells expressing dominant negative IFN gamma receptors. Immunity 1: 447-456. https://doi.org/10.1016/1074-7613(94)90087-6
  7. Deguine J, Bousso P. 2013. Dynamics of NK cell interactions in vivo. Immunol. Rev. 251: 154-159. https://doi.org/10.1111/imr.12015
  8. Stojanovic A, Cerwenka A. 2011. Natural killer cells and solid tumors. J. Innate Immun. 3: 355-364.
  9. Alfano M, Graziano F, Genovese L, Poli G. 2013. Macrophage polarization at the crossroad between HIV-1 infection and cancer development. Arterioscler. Thromb. Vasc. Biol. 33: 1145-1152. https://doi.org/10.1161/ATVBAHA.112.300171
  10. Raza H, John A, Shafarin J. 2016. Potentiation of LPS-induced apoptotic cell death in human hepatoma HepG2 cells by aspirin via ROS and mitochondrial dysfunction: protection by N-acetyl cysteine. PLoS One 11: e0159750. https://doi.org/10.1371/journal.pone.0159750
  11. Zhao Y, Shao F. 2016. Diverse mechanisms for inflammasome sensing of cytosolic bacteria and bacterial virulence. Curr. Opin. Microbiol. 29: 37-42. https://doi.org/10.1016/j.mib.2015.10.003
  12. Kayagaki N, Stowe IB, Lee BL, O'Rourke K, Anderson K, Warming S, et al. 2015. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 526: 666-671.
  13. Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, et al. 2015. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 526: 660-665. https://doi.org/10.1038/nature15514
  14. Shi J, Gao W, Shao F. 2017. Pyroptosis: gasdermin-mediated programmed necrotic cell death. Trends Biochem. Sci. 42: 245-254. https://doi.org/10.1016/j.tibs.2016.10.004
  15. Birkinshaw RW, Czabotar PE. 2017. The BCL-2 family of proteins and mitochondrial outer membrane permeabilisation. Semin. Cell Dev. Biol. 72: 152-162. https://doi.org/10.1016/j.semcdb.2017.04.001
  16. Garner TP, Lopez A, Reyna DE, Spitz AZ, Gavathiotis E. 2017. Progress in targeting the BCL-2 family of proteins. Curr. Opin. Chem. Biol. 39: 133-142.
  17. Chipuk JE, Moldoveanu T, Llambi F, Parsons MJ, Green DR. 2010. The BCL-2 family reunion. Mol. Cell 37: 299-310. https://doi.org/10.1016/j.molcel.2010.01.025
  18. Walensky LD, Pitter K, Morash J, Oh KJ, Barbuto S, Fisher J, et al. 2006 . A stapled BID BH3 helix directly binds and activates BAX. Mol. Cell 24: 199-210. https://doi.org/10.1016/j.molcel.2006.08.020
  19. Kim H, Tu HC, Ren D, Takeuchi O, Jeffers JR, Zambetti GP, et al. 2009. Stepwise activation of BAX and BAK by tBID, BIM, and PUMA initiates mitochondrial apoptosis. Mol. Cell 36: 487-499. https://doi.org/10.1016/j.molcel.2009.09.030
  20. Guikema JE, Amiot M, Eldering E. 2017. Exploiting the pro-apoptotic function of NOXA as a therapeutic modality in cancer. Expert Opin. Ther. Targets 21: 767-779. https://doi.org/10.1080/14728222.2017.1349754
  21. Dai H, Smith A, Meng XW, Schneider PA, Pang YP, Kaufmann SH. 2011. Transient binding of an activator BH3 domain to the Bak BH3-binding groove initiates Bak oligomerization. J. Cell Biol. 194: 39-48.
  22. Piya S, Moon AR, Song PI, Hiscott J, Lin R, Seol DW, et al. 2011. Suppression of IRF4 by IRF1, 3, and 7 in Noxa expression is a necessary event for IFN-gamma-mediated tumor elimination. Mol. Cancer Res. 9: 1356-1365. https://doi.org/10.1158/1541-7786.MCR-11-0185
  23. Oda E, Ohki R, Murasawa H, Nemoto J, Shibue T, Yamashita T, et al. 2000. Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science 288: 1053-1058. https://doi.org/10.1126/science.288.5468.1053
  24. Seo YW, Shin JN, Ko KH, Cha JH, Park JY, Lee BR, et al. 2003. The molecular mechanism of Noxa-induced mitochondrial dysfunction in p53-mediated cell death. J. Biol. Chem. 278: 48292-48299.
  25. Kuzmich NN, Sivak KV, Chubarev VN, Porozov YB, Savateeva-Lyubimova TN, Peri F. 2017. TLR4 signaling pathway modulators as potential therapeutics in inflammation and sepsis. Vaccines (Basel) 5: E34. https://doi.org/10.3390/vaccines5040034
  26. Hennessy EJ. 2016. Selective inhibitors of Bcl-2 and Bcl-xL: balancing antitumor activity with on-target toxicity. Bioorg. Med. Chem. Lett. 26: 2105-2114. https://doi.org/10.1016/j.bmcl.2016.03.032
  27. Bhola PD, Letai A. 2016. Mitochondria - judges and executioners of cell death sentences. Mol. Cell 61: 695-704. https://doi.org/10.1016/j.molcel.2016.02.019
  28. Brunschwig A. 1939. The efficacy of "Coley's toxin" in the treatment of sarcoma: an experimental study. Ann. Surg. 109: 109-113.
  29. Nauts HC, Fowler GA, Bogatko FH. 1953. A review of the influence of bacterial infection and of bacterial products (Coley's toxins) on malignant tumors in man; a critical analysis of 30 inoperable cases treated by Coley's mixed toxins, in which diagnosis was confirmed by microscopic examination selected for special study. Acta Med. Scand. Suppl. 276: 1-103.
  30. Maletzki C, Klier U, Obst W, Kreikemeyer B, Linnebacher M. 2012. Reevaluating the concept of treating experimental tumors with a mixed bacterial vaccine: Coley's toxin. Clin. Dev. Immunol. 2012: 230625.
  31. Freudenberg MA, Kumazawa Y, Meding S, Langhorne J, Galanos C. 1991. Gamma interferon production in endotoxin-responder and -nonresponder mice during infection. Infect. Immun. 59: 3484-3491.
  32. Freudenberg MA, Merlin T, Gumenscheimer M, Kalis C, Landmann R, Galanos C. 2001. Role of lipopolysaccharide susceptibility in the innate immune response to Salmonella typhimurium infection: LPS, a primary target for recognition of gram-negative bacteria. Microbes Infect. 3: 1213-1222. https://doi.org/10.1016/S1286-4579(01)01481-2
  33. Nansen A, Randrup Thomsen A. 2001. Viral infection causes rapid sensitization to lipopolysaccharide: central role of IFN-alpha beta. J. Immunol. 166: 982-988.
  34. Chen L, Willis SN, Wei A, Smith BJ, Fletcher JI, Hinds MG, et al. 2005. Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function. Mol. Cell 17: 393-403. https://doi.org/10.1016/j.molcel.2004.12.030
  35. Kuwana T, Bouchier-Hayes L, Chipuk JE, Bonzon C, Sullivan BA, Green DR, et al. 2005. BH3 domains of BH3-only proteins differentially regulate Bax-mediated mitochondrial membrane permeabilization both directly and indirectly. Mol. Cell 17: 525-535. https://doi.org/10.1016/j.molcel.2005.02.003
  36. Porta C, Hadj-Slimane R, Nejmeddine M, Pampin M, Tovey MG, Espert L, et al. 2005. Interferons alpha and gamma induce p53-dependent and p53-independent apoptosis, respectively. Oncogene 24: 605-615. https://doi.org/10.1038/sj.onc.1208204
  37. Kim JY, Ahn HJ, Ryu JH, Suk K , Park JH. 2004. BH3-only protein Noxa is a mediator of hypoxic cell death induced by hypoxia-inducible factor 1alpha. J. Exp. Med. 199: 113-124. https://doi.org/10.1084/jem.20030613
  38. Lallemand C, Blanchard B, Palmieri M, Lebon P, May E, Tovey MG. 2007. Single-stranded RNA viruses inactivate the transcriptional activity of p53 but induce NOXA-dependent apoptosis via post-translational modifications of IRF-1, IRF-3 and CREB. Oncogene 26: 328-338. https://doi.org/10.1038/sj.onc.1209795
  39. Qin JZ, Ziffra J, Stennett L, Bodner B, Bonish BK, Chaturvedi V, et al. 2005. Proteasome inhibitors trigger NOXA-mediated apoptosis in melanoma and myeloma cells. Cancer Res. 65: 6282-6293.
  40. Nikiforov MA, Riblett M, Tang WH, Gratchouck V, Zhuang D, Fernandez Y, et al. 2007. Tumor cell-selective regulation of NOXA by c-MYC in response to proteasome inhibition. Proc. Natl. Acad. Sci. USA 104: 19488-19493. https://doi.org/10.1073/pnas.0708380104
  41. Flinterman M, Guelen L, Ezzati-Nik S, Killick R, Melino G, Tominaga K, et al. 2005. E1A activates transcription of p73 and Noxa to induce apoptosis. J. Biol. Chem. 280: 5945-5959.
  42. Obexer P, Geiger K, Ambros PF, Meister B, Ausserlechner MJ. 2007. FKHRL1-mediated expression of Noxa and Bim induces apoptosis via the mitochondria in neuroblastoma cells. Cell Death Differ. 14: 534-547. https://doi.org/10.1038/sj.cdd.4402017
  43. Desai S, Maurin M, Smith MA, Bolick SC, Dessureault S, Tao J, et al. 2010. PRDM1 is required for mantle cell lymphoma response to bortezomib. Mol. Cancer Res. 8: 907-918. https://doi.org/10.1158/1541-7786.MCR-10-0131
  44. Valis K, Prochazka L, Boura E, Chladova J, Obsil T, Rohlena J, et al. 2011. Hippo/Mst1 stimulates transcription of the proapoptotic mediator NOXA in a FoxO1-dependent manner. Cancer Res. 71: 946-954. https://doi.org/10.1158/0008-5472.CAN-10-2203
  45. Marshall AD, Picchione F, Geltink RI, Grosveld GC. 2013. PAX3-FOXO1 induces up-regulation of Noxa sensitizing alveolar rhabdomyosarcoma cells to apoptosis. Neoplasia 15: 738-748. https://doi.org/10.1593/neo.121888

Cited by

  1. Low-dose lipopolysaccharide protects nerve cells against spinal cord injury via regulating the PI3K-AKT-Nrf2 signaling pathway vol.99, pp.5, 2018, https://doi.org/10.1139/bcb-2020-0641