Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 8/2009

01-08-2009 | Original Article

Sodium butyrate upregulates expression of NKG2D ligand MICA/B in HeLa and HepG2 cell lines and increases their susceptibility to NK lysis

Authors: Cai Zhang, Yiping Wang, Zhixia Zhou, Jian Zhang, Zhigang Tian

Published in: Cancer Immunology, Immunotherapy | Issue 8/2009

Login to get access

Abstract

Natural killer (NK) cells are important effectors in the immune response to tumors. A number of cell-surface inhibitory and activating receptors on NK cells tightly regulate their interaction with target cell ligands. In particular, the strength of an anti-tumor immune response appears to depend critically on surface levels of one activating receptor, NKG2D. Correspondingly, expression of NKG2D ligands on target cells is a requirement for effective tumor immunosurveillance and the elimination of pathogen-infected cells. Sodium butyrate, a potent repressor of histone deacetylase (HDAC), has recently been proposed as a potential agent in cancer treatment based on its ability to modify, in several cancer cell types, the expression of a variety of genes related to cell cycle regulation and apoptosis. Here we report that, in the HeLa and HepG2 tumor cell lines, sodium butyrate upregulated the expression of the MHC class I-related chain molecules A and B (MICA and MICB) at both the mRNA and protein levels, resulting in an enhanced susceptibility of cells in both lines to NK lysis. It also led to an elevated expression of heat shock protein 70 (HSP70) and transcription factor Sp1, and increased the binding of transcription factors Sp1 and heat shock transcription factor 1 (HSF1) to the MICA/B promoter, resulting in increased expression of MICA and MICB. siRNA targeting Sp1 significantly attenuate the enhancement of MICA expression by sodium butyrate. These results suggest that sodium butyrate and other HDAC inhibitors may have therapeutic potential by enhancing the immune response to cancer.
Literature
1.
2.
go back to reference Smyth MJ, Hayakawa Y, Takeda K, Yagita H (2002) New aspects of natural-killer-cell surveillance and therapy of cancer. Nat Rev Cancer 2:850–861PubMedCrossRef Smyth MJ, Hayakawa Y, Takeda K, Yagita H (2002) New aspects of natural-killer-cell surveillance and therapy of cancer. Nat Rev Cancer 2:850–861PubMedCrossRef
3.
4.
go back to reference Watzl C (2003) The NKG2D receptor and its ligands-recognition beyond the “missing self”? Microbes Infect 5:31–37PubMedCrossRef Watzl C (2003) The NKG2D receptor and its ligands-recognition beyond the “missing self”? Microbes Infect 5:31–37PubMedCrossRef
5.
go back to reference Cerwenka A, Lanier LL (2003) NKG2D ligands: unconventional MHC class I-like molecules exploited by viruses and cancer. Tissue Antigen 61:335–343CrossRef Cerwenka A, Lanier LL (2003) NKG2D ligands: unconventional MHC class I-like molecules exploited by viruses and cancer. Tissue Antigen 61:335–343CrossRef
6.
go back to reference López-Larrea C, Suárez-Alvarez B, López-Soto A, López-Vázquez A, Gonzalez S (2008) The NKG2D receptor: sensing stressed cells. Trends Mol Med 14:179–189PubMedCrossRef López-Larrea C, Suárez-Alvarez B, López-Soto A, López-Vázquez A, Gonzalez S (2008) The NKG2D receptor: sensing stressed cells. Trends Mol Med 14:179–189PubMedCrossRef
7.
go back to reference Gasser S, Orsulic S, Brown EJ, Raulet DH (2005) The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 436:1186–1190PubMedCrossRef Gasser S, Orsulic S, Brown EJ, Raulet DH (2005) The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 436:1186–1190PubMedCrossRef
8.
go back to reference Gasser S, Rault DH (2006) The DNA damage response arouses the immune system. Cancer Res 66:3959–3962PubMedCrossRef Gasser S, Rault DH (2006) The DNA damage response arouses the immune system. Cancer Res 66:3959–3962PubMedCrossRef
9.
go back to reference Yamamoto K, Fujiyama Y, Andoh A, Bamba T, Okabe H (2001) Oxidative stress increases MICA and MICB gene expression in the human colon carcinoma cell line (CaCo-2). Biochim Biophys Acta 1526:10–12PubMed Yamamoto K, Fujiyama Y, Andoh A, Bamba T, Okabe H (2001) Oxidative stress increases MICA and MICB gene expression in the human colon carcinoma cell line (CaCo-2). Biochim Biophys Acta 1526:10–12PubMed
11.
go back to reference Armeanu S, Bitzer M, Lauer UM, Venturelli S, Pathil A, Krusch M, Kaiser S, Jobst J, Smirnow I, Wagner A, Steinle A, Salih HR (2005) Natural killer cell-mediated lysis of hepatoma cells via specific induction of NKG2D ligands by the histone deacetylase inhibitor sodium valproate. Cancer Res 65:6321–6329PubMedCrossRef Armeanu S, Bitzer M, Lauer UM, Venturelli S, Pathil A, Krusch M, Kaiser S, Jobst J, Smirnow I, Wagner A, Steinle A, Salih HR (2005) Natural killer cell-mediated lysis of hepatoma cells via specific induction of NKG2D ligands by the histone deacetylase inhibitor sodium valproate. Cancer Res 65:6321–6329PubMedCrossRef
12.
go back to reference Kato N, Tanaka J, Sugita J, Toubai T, Miura Y, Ibata M, Syono Y, Ota S, Kondo T, Asaka M, Imamura M (2007) Regulation of the expression of MHC class I-related chain A, B (MICA, MICB) via chromatin remodeling and its impact on the susceptibility of leukemic cells to the cytotoxicity of NKG2D-expressing cells. Leukemia 21:2103–2108PubMedCrossRef Kato N, Tanaka J, Sugita J, Toubai T, Miura Y, Ibata M, Syono Y, Ota S, Kondo T, Asaka M, Imamura M (2007) Regulation of the expression of MHC class I-related chain A, B (MICA, MICB) via chromatin remodeling and its impact on the susceptibility of leukemic cells to the cytotoxicity of NKG2D-expressing cells. Leukemia 21:2103–2108PubMedCrossRef
13.
go back to reference Muhlethaler-Mottet A, Meier R, Flahaut M, Bourloud KB, Nardou K, Joseph JM, Gross N (2008) Complex molecular mechanisms cooperate to mediate histone deacetylase inhibitors anti-tumour activity in neuroblastoma cells. Mol Cancer 7:55PubMedCrossRef Muhlethaler-Mottet A, Meier R, Flahaut M, Bourloud KB, Nardou K, Joseph JM, Gross N (2008) Complex molecular mechanisms cooperate to mediate histone deacetylase inhibitors anti-tumour activity in neuroblastoma cells. Mol Cancer 7:55PubMedCrossRef
14.
go back to reference Zhang C, Niu J, Zhang J, Wang Y, Zhou Z, Zhang J, Tian Z (2008) Opposing effect of IFNα and IFNγ on expression of MHC class I chain-related A in tumors. Cancer Sci 99:1279–1286PubMedCrossRef Zhang C, Niu J, Zhang J, Wang Y, Zhou Z, Zhang J, Tian Z (2008) Opposing effect of IFNα and IFNγ on expression of MHC class I chain-related A in tumors. Cancer Sci 99:1279–1286PubMedCrossRef
15.
go back to reference Cartharius K, Frech K, Grote K, Klocke B, Haltmeier M, Klingenhoff A, Frisch M, Bayerlein M, Werner T (2005) MatInspector and beyond: promoter analysis based on transcription factor binding sites. Bioinformatics 21:2933–2942PubMedCrossRef Cartharius K, Frech K, Grote K, Klocke B, Haltmeier M, Klingenhoff A, Frisch M, Bayerlein M, Werner T (2005) MatInspector and beyond: promoter analysis based on transcription factor binding sites. Bioinformatics 21:2933–2942PubMedCrossRef
16.
go back to reference Nausch N, Florin L, Hartenstein B, Angel P, Schorpp-Kistner M, Cerwenka A (2006) Cutting edge: the AP-1 subunit JunB determines NK cell-mediated target cell killing by regulation of the NKG2D-ligand RAE-1epsilon. J Immunol 176:7–11PubMed Nausch N, Florin L, Hartenstein B, Angel P, Schorpp-Kistner M, Cerwenka A (2006) Cutting edge: the AP-1 subunit JunB determines NK cell-mediated target cell killing by regulation of the NKG2D-ligand RAE-1epsilon. J Immunol 176:7–11PubMed
17.
go back to reference Rodríguez-Rodero S, González S, Rodrigo L, Fernández-Morera JL, Martínez-Borra J, López-Vázquez A, López-Larrea C (2007) Transcriptional regulation of MICA and MICB: a novel polymorphism in MICB promoter alters transcriptional regulation by Sp1. Eur J Immunol 37:1938–1953PubMedCrossRef Rodríguez-Rodero S, González S, Rodrigo L, Fernández-Morera JL, Martínez-Borra J, López-Vázquez A, López-Larrea C (2007) Transcriptional regulation of MICA and MICB: a novel polymorphism in MICB promoter alters transcriptional regulation by Sp1. Eur J Immunol 37:1938–1953PubMedCrossRef
18.
go back to reference Venkataraman GM, Suciu D, Groh V, Boss JM, Spies T (2007) Promoter region architecture and transcriptional regulation of the genes for the MHC class I-related Chain A and B ligands of NKG2D. J Immunol 178:961–969PubMed Venkataraman GM, Suciu D, Groh V, Boss JM, Spies T (2007) Promoter region architecture and transcriptional regulation of the genes for the MHC class I-related Chain A and B ligands of NKG2D. J Immunol 178:961–969PubMed
20.
go back to reference Tomasi TB, Magner WJ, Khan AN (2006) Epigenetic regulation of immune escape genes in cancer. Cancer Immunol Immunother 55:1159–1184PubMedCrossRef Tomasi TB, Magner WJ, Khan AN (2006) Epigenetic regulation of immune escape genes in cancer. Cancer Immunol Immunother 55:1159–1184PubMedCrossRef
21.
go back to reference Cress WD, Seto E (2000) Histone deacetylases, transcriptional control, and cancer. J Cell Physiol 184:1–16PubMedCrossRef Cress WD, Seto E (2000) Histone deacetylases, transcriptional control, and cancer. J Cell Physiol 184:1–16PubMedCrossRef
22.
go back to reference Pan LN, Lu J, Huang B (2007) HDAC inhibitors: a potential new category of anti-tumor agents. Cell Mol Immunol 4:337–343PubMed Pan LN, Lu J, Huang B (2007) HDAC inhibitors: a potential new category of anti-tumor agents. Cell Mol Immunol 4:337–343PubMed
23.
go back to reference Xu J, Zhou JY, Wei WZ, Philipsen S, Wu GS (2008) Sp1-mediated TRAIL induction in chemosensitization. Cancer Res 68:6718–6726PubMedCrossRef Xu J, Zhou JY, Wei WZ, Philipsen S, Wu GS (2008) Sp1-mediated TRAIL induction in chemosensitization. Cancer Res 68:6718–6726PubMedCrossRef
24.
go back to reference Chinnaiyan P, Cerna D, Burgan WE, Beam K, Williams ES, Camphausen K, Tofilon PJ (2008) Postradiation sensitization of the histone deacetylase inhibitor valproic acid. Clin Cancer Res 14:5410–5415PubMedCrossRef Chinnaiyan P, Cerna D, Burgan WE, Beam K, Williams ES, Camphausen K, Tofilon PJ (2008) Postradiation sensitization of the histone deacetylase inhibitor valproic acid. Clin Cancer Res 14:5410–5415PubMedCrossRef
25.
go back to reference Ryu H, Lee J, Olofsson BA, Mwidau A, Dedeoglu A, Escudero M, Flemington E, Azizkhan-Clifford J, Ferrante RJ, Ratan RR (2003) Histone deacetylase inhibitors prevent oxidative neuronal death independent of expanded polyglutamine repeats via an Sp1-dependent pathway. Proc Natl Acad Sci USA 100:4281–4286PubMedCrossRef Ryu H, Lee J, Olofsson BA, Mwidau A, Dedeoglu A, Escudero M, Flemington E, Azizkhan-Clifford J, Ferrante RJ, Ratan RR (2003) Histone deacetylase inhibitors prevent oxidative neuronal death independent of expanded polyglutamine repeats via an Sp1-dependent pathway. Proc Natl Acad Sci USA 100:4281–4286PubMedCrossRef
26.
go back to reference Kim YK, Lee EK, Kang JK, Kim JA, You JS, Park JH, Seo DW, Hwang JW, Kim SN, Lee HY, Lee HW, Han JW (2006) Activation of NF-kappaB by HDAC inhibitor apicidin through Sp1-dependent de novo protein synthesis: its implication for resistance to apoptosis. Cell Death Differ 13:2033–2041PubMedCrossRef Kim YK, Lee EK, Kang JK, Kim JA, You JS, Park JH, Seo DW, Hwang JW, Kim SN, Lee HY, Lee HW, Han JW (2006) Activation of NF-kappaB by HDAC inhibitor apicidin through Sp1-dependent de novo protein synthesis: its implication for resistance to apoptosis. Cell Death Differ 13:2033–2041PubMedCrossRef
27.
go back to reference Andresen L, Jensen H, Pedersen MT, Hansen KA, Skov S (2007) Molecular regulation of MHC class I chain-related protein A expression after HDAC-inhibitor treatment of Jurkat T cells. J Immunol 179:8235–8242PubMed Andresen L, Jensen H, Pedersen MT, Hansen KA, Skov S (2007) Molecular regulation of MHC class I chain-related protein A expression after HDAC-inhibitor treatment of Jurkat T cells. J Immunol 179:8235–8242PubMed
28.
go back to reference Seo HR, Chung DY, Lee YJ, Lee DH, Kim JI, Bae S, Chung HY, Lee SJ, Jeoung D, Lee YS (2006) Heat shock protein 25 or inducible heat shock protein 70 activates heat shock factor 1: dephosphorylation on serine 307 through inhibition of ERK1/2 phosphorylation. J Biol Chem 281:17220–17227PubMedCrossRef Seo HR, Chung DY, Lee YJ, Lee DH, Kim JI, Bae S, Chung HY, Lee SJ, Jeoung D, Lee YS (2006) Heat shock protein 25 or inducible heat shock protein 70 activates heat shock factor 1: dephosphorylation on serine 307 through inhibition of ERK1/2 phosphorylation. J Biol Chem 281:17220–17227PubMedCrossRef
29.
go back to reference Kim YH, Park JW, Lee JY, Kwon TK (2004) Sodium butyrate sensitizes TRAIL-mediated apoptosis by induction of transcription from the DR5 gene promoter through Sp1 sites in colon cancer cells. Carcinogenesis 25:1813–1820PubMedCrossRef Kim YH, Park JW, Lee JY, Kwon TK (2004) Sodium butyrate sensitizes TRAIL-mediated apoptosis by induction of transcription from the DR5 gene promoter through Sp1 sites in colon cancer cells. Carcinogenesis 25:1813–1820PubMedCrossRef
30.
go back to reference Pajak B, Orzechowski A (2007) Sodium butyrate-dependent sensitization of human colon adenocarcinoma COLO 205 cells to TNF-alpha-induced apoptosis. J Physiol Pharmacol 58(Suppl 3):163–176PubMed Pajak B, Orzechowski A (2007) Sodium butyrate-dependent sensitization of human colon adenocarcinoma COLO 205 cells to TNF-alpha-induced apoptosis. J Physiol Pharmacol 58(Suppl 3):163–176PubMed
31.
go back to reference Kramer OH, Gottlicher M, Heinzel T (2001) Histone deacetylase as a therapeutic agent. Trends Endocrinol Metab 12:294–300PubMedCrossRef Kramer OH, Gottlicher M, Heinzel T (2001) Histone deacetylase as a therapeutic agent. Trends Endocrinol Metab 12:294–300PubMedCrossRef
32.
go back to reference McIntyne A, Gibson PR, Young GP (1993) Butyrate production from dietary fibers and protection against large bowel cancer in a rat model. Gut 34:386–391CrossRef McIntyne A, Gibson PR, Young GP (1993) Butyrate production from dietary fibers and protection against large bowel cancer in a rat model. Gut 34:386–391CrossRef
33.
go back to reference Glaser KB (2007) HDAC inhibitors: Clinical update and mechanism-based potential. Biochem Pharmacol 74:659–671PubMedCrossRef Glaser KB (2007) HDAC inhibitors: Clinical update and mechanism-based potential. Biochem Pharmacol 74:659–671PubMedCrossRef
34.
go back to reference Groh V, Wu J, Yee C, Spies T (2002) Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 419:734–738PubMedCrossRef Groh V, Wu J, Yee C, Spies T (2002) Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 419:734–738PubMedCrossRef
35.
go back to reference Doubrovina ES, Doubrovin MM, Vider E, Sisson RB, O’Reilly RJ, Dupont B, Vyas YM (2003) Evasion from NK cell immunity by MHC class I chain-related molecules expressing colon adenocarcinoma. J Immunol 171:6891–6899PubMed Doubrovina ES, Doubrovin MM, Vider E, Sisson RB, O’Reilly RJ, Dupont B, Vyas YM (2003) Evasion from NK cell immunity by MHC class I chain-related molecules expressing colon adenocarcinoma. J Immunol 171:6891–6899PubMed
36.
go back to reference Oppenheim DE, Roberts SJ, Clarke SL, Filler R, Lewis JM, Tigelaar RE, Girardi M, Hayday AC (2005) Sustained localized expression of ligand for the activating NKG2D receptor impairs natural cytotoxicity in vivo and reduces tumor immunosurveillance. Nat Immunol 6:928–937PubMedCrossRef Oppenheim DE, Roberts SJ, Clarke SL, Filler R, Lewis JM, Tigelaar RE, Girardi M, Hayday AC (2005) Sustained localized expression of ligand for the activating NKG2D receptor impairs natural cytotoxicity in vivo and reduces tumor immunosurveillance. Nat Immunol 6:928–937PubMedCrossRef
37.
go back to reference Roberts AI, Lee L, Schwarz E, Groh V, Spies T, Ebert EC (2001) Jabri B (2001) NKG2D receptors induced by IL–15 costimulate CD28-negative effector CTL in the tissue microenvironment. J Immunol 167:5527–5530PubMed Roberts AI, Lee L, Schwarz E, Groh V, Spies T, Ebert EC (2001) Jabri B (2001) NKG2D receptors induced by IL–15 costimulate CD28-negative effector CTL in the tissue microenvironment. J Immunol 167:5527–5530PubMed
38.
go back to reference Zhang C, Zhang J, Niu J, Zhang J, Tian Z (2008) Interleukin-15 improves cytotoxicity of natural killer cells via up-regulating NKG2D and cytotoxic effector molecule expression as well as STAT1 and ERK1/2 phosphorylation. Cytokine 42:128–136PubMedCrossRef Zhang C, Zhang J, Niu J, Zhang J, Tian Z (2008) Interleukin-15 improves cytotoxicity of natural killer cells via up-regulating NKG2D and cytotoxic effector molecule expression as well as STAT1 and ERK1/2 phosphorylation. Cytokine 42:128–136PubMedCrossRef
39.
go back to reference Zhang C, Zhang J, Niu J, Zhou Z, Zhang J, Tian Z (2008) Interleukin-12 improves cytotoxicity of natural killer cells via upregulated expression of NKG2D. Hum Immunol 69:490–500PubMedCrossRef Zhang C, Zhang J, Niu J, Zhou Z, Zhang J, Tian Z (2008) Interleukin-12 improves cytotoxicity of natural killer cells via upregulated expression of NKG2D. Hum Immunol 69:490–500PubMedCrossRef
40.
go back to reference Zhang C, Zhang J, Sun R, Feng J, Wei H, Tian Z (2005) Opposing effect of IFNγ and IFNα on expression of NKG2 receptor family: negative regulation of IFNγ on NK cells. Int Immunopharmacol 5:1057–1067PubMedCrossRef Zhang C, Zhang J, Sun R, Feng J, Wei H, Tian Z (2005) Opposing effect of IFNγ and IFNα on expression of NKG2 receptor family: negative regulation of IFNγ on NK cells. Int Immunopharmacol 5:1057–1067PubMedCrossRef
41.
go back to reference Wu JD, Higgins LM, Steinle A, Cosman D, Haugk K, Plymate SR (2004) Prevalent expression of the immunostimulatory MHC class I chain-related molecule is counteracted by shedding in prostate cancer. J Clin Invest 114:560–568PubMed Wu JD, Higgins LM, Steinle A, Cosman D, Haugk K, Plymate SR (2004) Prevalent expression of the immunostimulatory MHC class I chain-related molecule is counteracted by shedding in prostate cancer. J Clin Invest 114:560–568PubMed
Metadata
Title
Sodium butyrate upregulates expression of NKG2D ligand MICA/B in HeLa and HepG2 cell lines and increases their susceptibility to NK lysis
Authors
Cai Zhang
Yiping Wang
Zhixia Zhou
Jian Zhang
Zhigang Tian
Publication date
01-08-2009
Publisher
Springer-Verlag
Published in
Cancer Immunology, Immunotherapy / Issue 8/2009
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-008-0645-8

Other articles of this Issue 8/2009

Cancer Immunology, Immunotherapy 8/2009 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