Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 2/2015

01-02-2015 | Original Article

N-3 polyunsaturated fatty acids inhibit IFN-γ-induced IL-18 binding protein production by prostate cancer cells

Authors: Xiaofeng Wang, Andrew Breeze, Marianna Kulka

Published in: Cancer Immunology, Immunotherapy | Issue 2/2015

Login to get access

Abstract

Prostate cancer cells can produce IL-18 binding protein (IL-18BP) in response to interferon-γ (IFN-γ), which may function to neutralize IL-18, an anti-tumor factor formerly known as IFN-γ inducing factor. The consumption of n-3 polyunsaturated fatty acids (PUFAs) has been associated with a lower risk of certain types of cancer including prostate cancer, although the precise mechanisms of this effect are poorly understood. We hypothesized that n-3 PUFAs could modify IL-18BP production by prostate cancer cells by altering IFN-γ receptor-mediated signal transduction. Here, we demonstrate that n-3 PUFA treatment significantly reduced IFN-γ-induced IL-18BP production by DU-145 and PC-3 prostate cancer cells by inhibiting IL-18BP mRNA expression and was associated with a reduction in IFN-γ receptor expression. Furthermore, IFN-γ-induced phosphorylation of Janus kinase 1 (JAK1), signal transducers and activators of transcription 1 (STAT1), extracellular signal-regulated kinases 1/2 (ERK1/2), and P38 were suppressed by n-3 PUFA treatment. By contrast, n-6 PUFA had no effect on IFN-γ receptor expression, but decreased IFN-γ-induced IL-18BP production and IFN-γ stimulation of JAK1, STAT1, ERK1/2, and JNK phosphorylation. These data indicate that both n-3 and n-6 PUFAs may be beneficial in prostate cancer by altering IFN-γ signaling, thus inhibiting IL-18BP production and thereby rendering prostate cancer cells more sensitive to IL-18-mediated immune responses.
Appendix
Available only for authorised users
Literature
3.
go back to reference Leung BP, Culshaw S, Gracie JA, Hunter D, Canetti CA, Campbell C, Cunha F, Liew FY, McInnes IB (2001) A role for IL-18 in neutrophil activation. J Immunol 167(5):2879–2886PubMedCrossRef Leung BP, Culshaw S, Gracie JA, Hunter D, Canetti CA, Campbell C, Cunha F, Liew FY, McInnes IB (2001) A role for IL-18 in neutrophil activation. J Immunol 167(5):2879–2886PubMedCrossRef
4.
go back to reference Darwich L, Coma G, Pena R, Bellido R, Blanco EJ, Este JA, Borras FE, Clotet B, Ruiz L, Rosell A, Andreo F, Parkhouse RM, Bofill M (2009) Secretion of interferon-gamma by human macrophages demonstrated at the single-cell level after costimulation with interleukin (IL)-12 plus IL-18. Immunology 126(3):386–393. doi:10.1111/j.1365-2567.2008.02905.x PubMedCentralPubMedCrossRef Darwich L, Coma G, Pena R, Bellido R, Blanco EJ, Este JA, Borras FE, Clotet B, Ruiz L, Rosell A, Andreo F, Parkhouse RM, Bofill M (2009) Secretion of interferon-gamma by human macrophages demonstrated at the single-cell level after costimulation with interleukin (IL)-12 plus IL-18. Immunology 126(3):386–393. doi:10.​1111/​j.​1365-2567.​2008.​02905.​x PubMedCentralPubMedCrossRef
5.
go back to reference Srivastava S, Pelloso D, Feng H, Voiles L, Lewis D, Haskova Z, Whitacre M, Trulli S, Chen YJ, Toso J, Jonak ZL, Chang HC, Robertson MJ (2013) Effects of interleukin-18 on natural killer cells: costimulation of activation through Fc receptors for immunoglobulin. Cancer Immunol Immunother 62(6):1073–1082. doi:10.1007/s00262-013-1403-0 PubMedCentralPubMedCrossRef Srivastava S, Pelloso D, Feng H, Voiles L, Lewis D, Haskova Z, Whitacre M, Trulli S, Chen YJ, Toso J, Jonak ZL, Chang HC, Robertson MJ (2013) Effects of interleukin-18 on natural killer cells: costimulation of activation through Fc receptors for immunoglobulin. Cancer Immunol Immunother 62(6):1073–1082. doi:10.​1007/​s00262-013-1403-0 PubMedCentralPubMedCrossRef
9.
go back to reference Schmitt MJ, Philippidou D, Reinsbach SE, Margue C, Wienecke-Baldacchino A, Nashan D, Behrmann I, Kreis S (2012) Interferon-gamma-induced activation of Signal Transducer and Activator of Transcription 1 (STAT1) up-regulates the tumor suppressing microRNA-29 family in melanoma cells. Cell Commun Signal 10(1):41. doi:10.1186/1478-811X-10-41 PubMedCentralPubMedCrossRef Schmitt MJ, Philippidou D, Reinsbach SE, Margue C, Wienecke-Baldacchino A, Nashan D, Behrmann I, Kreis S (2012) Interferon-gamma-induced activation of Signal Transducer and Activator of Transcription 1 (STAT1) up-regulates the tumor suppressing microRNA-29 family in melanoma cells. Cell Commun Signal 10(1):41. doi:10.​1186/​1478-811X-10-41 PubMedCentralPubMedCrossRef
10.
go back to reference Shin EC, Shin WC, Choi Y, Kim H, Park JH, Kim SJ (2001) Effect of interferon-gamma on the susceptibility to Fas (CD95/APO-1)-mediated cell death in human hepatoma cells. Cancer Immunol Immunother 50(1):23–30PubMedCrossRef Shin EC, Shin WC, Choi Y, Kim H, Park JH, Kim SJ (2001) Effect of interferon-gamma on the susceptibility to Fas (CD95/APO-1)-mediated cell death in human hepatoma cells. Cancer Immunol Immunother 50(1):23–30PubMedCrossRef
11.
go back to reference Street D, Kaufmann AM, Vaughan A, Fisher SG, Hunter M, Schreckenberger C, Potkul RK, Gissmann L, Qiao L (1997) Interferon-gamma enhances susceptibility of cervical cancer cells to lysis by tumor-specific cytotoxic T cells. Gynecol Oncol 65(2):265–272. doi:10.1006/gyno.1997.4667 PubMedCrossRef Street D, Kaufmann AM, Vaughan A, Fisher SG, Hunter M, Schreckenberger C, Potkul RK, Gissmann L, Qiao L (1997) Interferon-gamma enhances susceptibility of cervical cancer cells to lysis by tumor-specific cytotoxic T cells. Gynecol Oncol 65(2):265–272. doi:10.​1006/​gyno.​1997.​4667 PubMedCrossRef
13.
go back to reference Hayakawa Y, Takeda K, Yagita H, Smyth MJ, Van Kaer L, Okumura K, Saiki I (2002) IFN-gamma-mediated inhibition of tumor angiogenesis by natural killer T-cell ligand, alpha-galactosylceramide. Blood 100(5):1728–1733PubMed Hayakawa Y, Takeda K, Yagita H, Smyth MJ, Van Kaer L, Okumura K, Saiki I (2002) IFN-gamma-mediated inhibition of tumor angiogenesis by natural killer T-cell ligand, alpha-galactosylceramide. Blood 100(5):1728–1733PubMed
15.
17.
go back to reference Leitzmann MF, Stampfer MJ, Michaud DS, Augustsson K, Colditz GC, Willett WC, Giovannucci EL (2004) Dietary intake of n-3 and n-6 fatty acids and the risk of prostate cancer. Am J Clin Nutr 80(1):204–216PubMed Leitzmann MF, Stampfer MJ, Michaud DS, Augustsson K, Colditz GC, Willett WC, Giovannucci EL (2004) Dietary intake of n-3 and n-6 fatty acids and the risk of prostate cancer. Am J Clin Nutr 80(1):204–216PubMed
19.
go back to reference Irons R, Fritsche KL (2005) Omega-3 polyunsaturated fatty acids impair in vivo interferon- gamma responsiveness via diminished receptor signaling. J Infect Dis 191(3):481–486. doi:10.1086/427264 PubMedCrossRef Irons R, Fritsche KL (2005) Omega-3 polyunsaturated fatty acids impair in vivo interferon- gamma responsiveness via diminished receptor signaling. J Infect Dis 191(3):481–486. doi:10.​1086/​427264 PubMedCrossRef
22.
go back to reference Hobeika AC, Etienne W, Cruz PE, Subramaniam PS, Johnson HM (1998) IFNgamma induction of p21WAF1 in prostate cancer cells: role in cell cycle, alteration of phenotype and invasive potential. Int J Cancer 77(1):138–145PubMedCrossRef Hobeika AC, Etienne W, Cruz PE, Subramaniam PS, Johnson HM (1998) IFNgamma induction of p21WAF1 in prostate cancer cells: role in cell cycle, alteration of phenotype and invasive potential. Int J Cancer 77(1):138–145PubMedCrossRef
23.
go back to reference Kominsky SL, Hobeika AC, Lake FA, Torres BA, Johnson HM (2000) Down-regulation of neu/HER-2 by interferon-gamma in prostate cancer cells. Cancer Res 60(14):3904–3908PubMed Kominsky SL, Hobeika AC, Lake FA, Torres BA, Johnson HM (2000) Down-regulation of neu/HER-2 by interferon-gamma in prostate cancer cells. Cancer Res 60(14):3904–3908PubMed
24.
go back to reference Igney FH, Krammer PH (2002) Immune escape of tumors: apoptosis resistance and tumor counterattack. J Leukoc Biol 71(6):907–920PubMed Igney FH, Krammer PH (2002) Immune escape of tumors: apoptosis resistance and tumor counterattack. J Leukoc Biol 71(6):907–920PubMed
25.
go back to reference Selleck WA, Canfield SE, Hassen WA, Meseck M, Kuzmin AI, Eisensmith RC, Chen SH, Hall SJ (2003) IFN-gamma sensitization of prostate cancer cells to Fas-mediated death: a gene therapy approach. Mol Ther 7(2):185–192PubMedCrossRef Selleck WA, Canfield SE, Hassen WA, Meseck M, Kuzmin AI, Eisensmith RC, Chen SH, Hall SJ (2003) IFN-gamma sensitization of prostate cancer cells to Fas-mediated death: a gene therapy approach. Mol Ther 7(2):185–192PubMedCrossRef
26.
go back to reference Hastie C (2008) Interferon gamma, a possible therapeutic approach for late-stage prostate cancer? Anticancer Res 28(5B):2843–2849PubMed Hastie C (2008) Interferon gamma, a possible therapeutic approach for late-stage prostate cancer? Anticancer Res 28(5B):2843–2849PubMed
27.
go back to reference He YF, Wang XH, Zhang GM, Chen HT, Zhang H, Feng ZH (2005) Sustained low-level expression of interferon-gamma promotes tumor development: potential insights in tumor prevention and tumor immunotherapy. Cancer Immunol Immunother 54(9):891–897. doi:10.1007/s00262-004-0654-1 PubMedCrossRef He YF, Wang XH, Zhang GM, Chen HT, Zhang H, Feng ZH (2005) Sustained low-level expression of interferon-gamma promotes tumor development: potential insights in tumor prevention and tumor immunotherapy. Cancer Immunol Immunother 54(9):891–897. doi:10.​1007/​s00262-004-0654-1 PubMedCrossRef
28.
go back to reference Dinarello CA (2000) Targeting interleukin 18 with interleukin 18 binding protein. Ann Rheum Dis 59(Suppl 1):17–20CrossRef Dinarello CA (2000) Targeting interleukin 18 with interleukin 18 binding protein. Ann Rheum Dis 59(Suppl 1):17–20CrossRef
30.
go back to reference Micallef MJ, Tanimoto T, Kohno K, Ikeda M, Kurimoto M (1997) Interleukin 18 induces the sequential activation of natural killer cells and cytotoxic T lymphocytes to protect syngeneic mice from transplantation with Meth A sarcoma. Cancer Res 57(20):4557–4563PubMed Micallef MJ, Tanimoto T, Kohno K, Ikeda M, Kurimoto M (1997) Interleukin 18 induces the sequential activation of natural killer cells and cytotoxic T lymphocytes to protect syngeneic mice from transplantation with Meth A sarcoma. Cancer Res 57(20):4557–4563PubMed
31.
go back to reference Begin ME, Ells G, Das UN, Horrobin DF (1986) Differential killing of human carcinoma cells supplemented with n-3 and n-6 polyunsaturated fatty acids. J Natl Cancer Inst 77(5):1053–1062PubMed Begin ME, Ells G, Das UN, Horrobin DF (1986) Differential killing of human carcinoma cells supplemented with n-3 and n-6 polyunsaturated fatty acids. J Natl Cancer Inst 77(5):1053–1062PubMed
33.
go back to reference Yang T, Fang S, Zhang HX, Xu LX, Zhang ZQ, Yuan KT, Xue CL, Yu HL, Zhang S, Li YF, Shi HP, Zhang Y (2013) N-3 PUFAs have antiproliferative and apoptotic effects on human colorectal cancer stem-like cells in vitro. J Nutr Biochem 24(5):744–753. doi:10.1016/j.jnutbio.2012.03.023 PubMedCrossRef Yang T, Fang S, Zhang HX, Xu LX, Zhang ZQ, Yuan KT, Xue CL, Yu HL, Zhang S, Li YF, Shi HP, Zhang Y (2013) N-3 PUFAs have antiproliferative and apoptotic effects on human colorectal cancer stem-like cells in vitro. J Nutr Biochem 24(5):744–753. doi:10.​1016/​j.​jnutbio.​2012.​03.​023 PubMedCrossRef
36.
go back to reference Ramana CV, Gil MP, Schreiber RD, Stark GR (2002) Stat1-dependent and -independent pathways in IFN-gamma-dependent signaling. Trends Immunol 23(2):96–101PubMedCrossRef Ramana CV, Gil MP, Schreiber RD, Stark GR (2002) Stat1-dependent and -independent pathways in IFN-gamma-dependent signaling. Trends Immunol 23(2):96–101PubMedCrossRef
37.
go back to reference Valledor AF, Sanchez-Tillo E, Arpa L, Park JM, Caelles C, Lloberas J, Celada A (2008) Selective roles of MAPKs during the macrophage response to IFN-gamma. J Immunol 180(7):4523–4529PubMedCrossRef Valledor AF, Sanchez-Tillo E, Arpa L, Park JM, Caelles C, Lloberas J, Celada A (2008) Selective roles of MAPKs during the macrophage response to IFN-gamma. J Immunol 180(7):4523–4529PubMedCrossRef
42.
go back to reference Thomasz L, Oglio R, Rossich L, Villamar S, Perona M, Salvarredi L, Dagrosa A, Pisarev MA, Juvenal GJ (2013) 6 Iodo-delta-lactone: a derivative of arachidonic acid with antitumor effects in HT-29 colon cancer cells. Prostaglandins Leukot Essent Fatty Acids 88(4):273–280. doi:10.1016/j.plefa.2013.01.002 PubMedCrossRef Thomasz L, Oglio R, Rossich L, Villamar S, Perona M, Salvarredi L, Dagrosa A, Pisarev MA, Juvenal GJ (2013) 6 Iodo-delta-lactone: a derivative of arachidonic acid with antitumor effects in HT-29 colon cancer cells. Prostaglandins Leukot Essent Fatty Acids 88(4):273–280. doi:10.​1016/​j.​plefa.​2013.​01.​002 PubMedCrossRef
43.
go back to reference Rietjens IM, van Tilburg CA, Coenen TM, Alink GM, Konings AW (1987) Influence of polyunsaturated fatty acid supplementation and membrane fluidity on ozone and nitrogen dioxide sensitivity of rat alveolar macrophages. J Toxicol Environ Health 21(1–2):45–56. doi:10.1080/15287398709531001 PubMedCrossRef Rietjens IM, van Tilburg CA, Coenen TM, Alink GM, Konings AW (1987) Influence of polyunsaturated fatty acid supplementation and membrane fluidity on ozone and nitrogen dioxide sensitivity of rat alveolar macrophages. J Toxicol Environ Health 21(1–2):45–56. doi:10.​1080/​1528739870953100​1 PubMedCrossRef
44.
go back to reference Brown M, Anderson KM, Patel H, Hopfinger AJ, Harris JE (1992) Eicosatetraynoic and arachidonic acid-induced changes in cell membrane fluidity consonant with differences in computer-aided design-structures. Biochim Biophys Acta 1105(2):285–290PubMedCrossRef Brown M, Anderson KM, Patel H, Hopfinger AJ, Harris JE (1992) Eicosatetraynoic and arachidonic acid-induced changes in cell membrane fluidity consonant with differences in computer-aided design-structures. Biochim Biophys Acta 1105(2):285–290PubMedCrossRef
Metadata
Title
N-3 polyunsaturated fatty acids inhibit IFN-γ-induced IL-18 binding protein production by prostate cancer cells
Authors
Xiaofeng Wang
Andrew Breeze
Marianna Kulka
Publication date
01-02-2015
Publisher
Springer Berlin Heidelberg
Published in
Cancer Immunology, Immunotherapy / Issue 2/2015
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-014-1630-z

Other articles of this Issue 2/2015

Cancer Immunology, Immunotherapy 2/2015 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