TLR2/4-mediated NF-κB pathway combined with the histone modification regulates β-defensins and interleukins expression by sodium phenyl butyrate in porcine intestinal epithelial cells

  • Xiujing Dou Northeast Agriculture University
  • Junlan Han
  • Qiuyuan Ma
  • Baojing Cheng
  • Anshan Shan
  • Nan Gao
  • Yu Yang
Keywords: Sodium phenylbutyrate; β-defensins; Inflammatory cytokines; Signaling pathway

Abstract

Background: Host defense peptides (HDPs) possess direct antibacterial, antineoplastic, and immunomodulatory abilities, playing a vital role in innate immunity. Dietary-regulated HDP holds immense potential as a novel pathway for preventing infection.

Objective: In this study, we examined the regulation mechanism of HDPs (pEP2C, pBD-1, and pBD-3) and cytokines (IL-8 and IL-18) expression by sodium phenylbutyrate (PBA).

Design: The effects of PBA on HDP induction and the mechanism involved were studied in porcine intestinal epithelial cell lines (IPEC J2).

Results: In this study, the results showed that HDPs (pEP2C, pBD-1, and pBD-3) and cytokines (IL-8 and IL- 18) expression was increased significantly upon stimulation with PBA in IPEC J2 cells. Furthermore, toll-like receptor 2 (TLR2) and TLR4 were required for the PBA-mediated upregulation of the HDPs. This process occurred and further activated the NF-κB pathway via the phosphorylation of p65 and an IκB α synthesis delay. Meanwhile, histone deacetylase (HDAC) inhibition and an increased phosphorylation of histone H3 on serine S10 also occurred in PBA-induced HDP expression independently with TLR2 and TLR4. Furthermore, p38-MAPK suppressed PBA-induced pEP2C, pBD-1 pBD-3, IL-8, and IL-18 expression, but ERK1/2 failed to abolish the regulation of pBD-3, IL-8, and IL-18. Moreover, epidermal growth factor receptor (EGFR) is involved in PBA-mediated HDP regulation.

Conclusions: We concluded that PBA induced HDP and cytokine increases but did not cause an excessive pro-inflammatory response, which proceeded through the TLR2 and TLR4-NF-κB pathway and histone modification in IPEC J2 cells.

Downloads

Download data is not yet available.

References


  1. O’Neill J. The review on antimicrobial resistance: tracking drug resistant infections globally. Wellcome Trust and the Department of Health of UK Government 2016.

  2. Hancock REW, Haney EF, Gill EE. The immunology of host defence peptides: beyond antimicrobial activity. Nat Rev Immunol 2016; 16: 321.

  3. Zhang LJ, Gallo RL. Antimicrobial peptides. Curr Biol Cb 2016; 26: R14.

  4. Sang Y, Blecha F. Porcine host defense peptides: expanding repertoire and functions. Dev Comp Immunol 2009; 33: 334–43.

  5. Choi MK, Le MT, Nguyen DT, Choi H, Kim W, Kim JH, et al. Genome-level identification, gene expression, and comparative analysis of porcine ß-defensin genes. Bmc Genet 2012; 13: 98.

  6. Zeng X, Sunkara LT, Jiang W, Bible M, Carter S, Ma X, et al. Induction of porcine host defense peptide gene expression by short-chain fatty acids and their analogs. Plos One 2013; 8: e72922.

  7. Lyu W, Curtis AR, Sunkara LT, Zhang G. Transcriptional regulation of antimicrobial host defense peptides. Curr Protein Pept Sci 2015; 16: 672–9.

  8. Xiong H, Guo B, Gan Z, Song D, Lu Z, Yi H, et al. Butyrate upregulates endogenous host defense peptides to enhance disease resistance in piglets via histone deacetylase inhibition. Sci Rep 2016; 6: 27070.

  9. Fischer N, Sechet E, Friedman R, Aurélien Amiot, Sobhani I, Nigro G, et al. Histone deacetylase inhibition enhances antimicrobial peptide but not inflammatory cytokine expression upon bacterial challenge. P Natl Acad Sci USA 2016; 113: 201605997.

  10. Coussens AK, Wilkinson RJ, Martineau AR. Phenylbutyrate is bacteriostatic against mycobacterium tuberculosis and regulates the macrophage response to infection, synergistically with 25-Hydroxy-Vitamin D3. Plos Pathogens 2015; 11: e1005007.

  11. Merzvinskyte R, Treigyte G, Savickiene J, Magnusson KE, Navakauskiene R. Effects of histone deacetylase inhibitors, sodium phenyl butyrate and vitamin B3, in combination with retinoic acid on granulocytic differntiation of human promyelocytic leukemia HL-60 cells. Ann Ny Acad Sci 2006; 1091: 356–67.

  12. Kulkarni NN, Yi Z, Huehnken C, Agerberth B, Gudmundsson GH. Phenylbutyrate induces cathelicidin expression via the vitamin D receptor: Linkage to inflammatory and growth factor cytokines pathways. Mol Immunol 2014; 63: 530–39.

  13. Kulkarni NN, Yi Z, Huehnken C, Agerberth B, Gudmundsson GH. Phenylbutyrate induces antimicrobial peptide expression. Antimicrob Agents Chemother 2009; 53: 5127.

  14. Mariani V, Palermo S, Fiorentini S, Lanubile A, Giuffra E. Gene expression study of two widely used pig intestinal epithelial cell lines: IPEC-J2 and IPI-2I. Vet Immunol Immunopathol 2009; 131: 278–84.

  15. Cao L, Ge X, Gao Y, Ren Y, Ren X, Li G. Porcine epidemic diarrhea virus infection induces NF-κB activation through the TLR2, TLR3 and TLR9 pathways in porcine intestinal epithelial cells. J Gen Virol 2015; 96: 1757.

  16. Dou X, Han J, Song W, Dong N, Xu X, Zhang W, et al. Sodium butyrate improves porcine host defense peptide expression and relieves the inflammatory response upon toll-like receptor 2 activation and histone deacetylase inhibition in porcine kidney cells. Oncotarget 2017; 8: 26532.

  17. Fukata M, Vamadevan AS, Abreu MT. Toll-like receptors (TLRs) and Nod-like receptors (NLRs) in inflammatory disorders. Semin Immunol 2009; 21: 242–53.

  18. Kamdar K, Nguyen V, Depaolo RW. Toll-like receptor signaling and regulation of intestinal immunity. Virulence 2013; 4: 207–12.

  19. Quivy V, Van LC. Regulation at multiple levels of NF-kappaB-mediated transactivation by protein acetylation. Biochem Pharmacol 2004; 68: 1221.

  20. Johnston A, Gudjonsson JE, Aphale A, Guzman AM, Stoll SW, Elder JT. EGFR and IL-1 signaling synergistically promote keratinocyte antimicrobial defenses in a differentiation-dependent manner. J Invest Dermatol 2011; 131: 329–37

  21. Robinson K, Deng Z, Hou Y, Zhang G. Regulation of the intestinal barrier function by host defense peptides. Front Vet Sci 2015; 2: 57.

  22. Peterson LW, Artis D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat Rev Immunol 2014; 14: 141.

  23. Alva-Murillo N, Téllez-Pérez AD, Sagrero-Cisneros E, López-Meza JE, Ochoa-Zarzosa A. Expression of antimicrobial peptides by bovine endothelial cells. Cell Immunol 2012; 280: 108–12.

  24. Kumar A, Zhang J, Yu FX. Toll-like receptor 2-mediated expression of β-defensin-2 in human corneal epithelial cells. Microb Infect 2006; 8: 380–9.

  25. Hertz CJ, Wu Q, Porter EM, Zhang YJ, Weismüller KH, Godowski PJ, et al. Activation of Toll-like receptor 2 on human tracheobronchial epithelial cells induces the antimicrobial peptide human beta defensin-2. J Immunol 2003; 171: 6820–6.

  26. Stadnyk AW. Intestinal epithelial cells as a source of inflammatory cytokines and chemokines. J Canadien de Gastroenterologie 2002; 16: 241–6.

  27. Montreekachon P, Nongparn S, Sastraruji T, Khongkhunthian S, Chruewkamlow N, Kasinrerk W, et al. Favorable interleukin-8 induction in human gingival epithelial cells by the antimicrobial peptide LL-37. Asian Pacific Journal of Allergy & Immunology 2014; 32: 251–60.

  28. François Niyonsaba, Ushio H, Nagaoka I, Okumura K, Ogawa H. The human beta-defensins (-1, -2, -3, -4) and cathelicidin LL-37 induce IL-18 secretion through p38 and ERK MAPK activation in primary human keratinocytes. J Immunol 2005; 175: 1776.

  29. Luo MC, Zhou SY, Feng DY, Xiao J, Li WY, Xu CD, et al. Runt-related transcription factor 1 (RUNX1) binds to p50 in macrophages and enhances TLR4-triggered inflammation and septic shock. J Biol Chem 2016; 291: 22011.

  30. Horion J, Gloire G, El Mjiyad N, Quivy V, Vermeulen L, Vanden Berghe W, et al. Histone deacetylase inhibitor trichostatin A sustains sodium pervanadate-induced NF-kappaB activation by delaying ikappaBalpha mRNA resynthesis: comparison with tumor necrosis factor alpha. J Biol Chem 2007; 282: 15383–93.

  31. Furumai R, Ito A, Ogawa K, Maeda S, Saito A, Nishino N, et al. Histone deacetylase inhibitors block nuclear factor-κB-dependent transcription by interfering with RNA polymerase II recruitment. Cancer Science 2011; 102: 1081.

  32. Wollebo HS, Bellizzi A, Cossari DH, Safak M, Khalili K, White MK. Epigenetic regulation of polyomavirus JC involves acetylation of specific lysine residues in NF-κB p65. J Neurovirol 2015; 21: 679–87.

  33. Leus NGJ, Zwinderman MRH, Dekker FJ. Histone deacetylase 3 (HDAC 3) as emerging drug target in NF-κB-mediated inflammation. Curr Opin Chem Biol 2016; 33: 160–8.

  34. Saccani S, Pantano S, Natoli G. p38-dependent marking of inflammatory genes for increased NF-|[kappa]|B recruitment. Nat Immunol 2002; 3: 69.

  35. Clayton AL, Mahadevan LC. MAP kinase-mediated phosphoacetylation of histone H3 and inducible gene regulation. Febs Lett 2003; 546: 51–8.

Published
2018-12-06
How to Cite
Dou X., Han J., Ma Q., Cheng B., Shan A., Gao N., & Yang Y. (2018). TLR2/4-mediated NF-κB pathway combined with the histone modification regulates β-defensins and interleukins expression by sodium phenyl butyrate in porcine intestinal epithelial cells. Food & Nutrition Research, 62. https://doi.org/10.29219/fnr.v62.1493
Section
Original Articles