DOI QR코드

DOI QR Code

The Modulatory Effect of Sodium Propionate Treatment in the Expression of Inflammatory Cytokines and Intracellular Growth of Brucella abortus 544 in Raw 264.7 Cells

  • Heejin Kim (Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University) ;
  • Tran Xuan Ngoc Huy (Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University) ;
  • Trang Thi Nguyen (Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University) ;
  • Alisha Wehdnesday Bernardo Reyes (Department of Veterinary Paraclinical Sciences, College of Veterinary Medicine, University of the Philippines Los Banos) ;
  • WonGi Min (Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University) ;
  • Hu Jang Lee (Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University) ;
  • Jin Hur (College of Veterinary Medicine, Chonbuk National University) ;
  • Suk Kim (Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University)
  • Received : 2023.03.28
  • Accepted : 2023.05.12
  • Published : 2023.08.28

Abstract

In this study, we investigated the effects of sodium propionate (SP) treatment on intracellular mechanism of murine macrophages and its contribution to host immunity during Brucella abortus 544 infection. The intracellular growth assay revealed that SP inhibited Brucella replication inside the macrophages. To determine intracellular signaling involved during SP treatment after Brucella infection, we analyzed the change of five different cytokines production relevant to SP such as TNF-α, IL-10, IFN-γ, IL-1β, and IL-6, and the results indicated that the boost with IL-10 was apparent throughout the culture period for 48 h as well as IL-1β which was apparent at 24 h post-infection and IFN-γ which was apparent at 24 h and 48 h in comparison to SP untreated groups. On the other way, SP-treated cells displayed suppressed production of TNF-α and IL-6 at all time points tested and 48 h post-infection, respectively. Furthermore, we conducted western blot to establish a cellular mechanism, and the result suggested that SP treatment attenuated p50 phosphorylation, part of the NF-κB pathway. These findings indicated that the inhibitory effect of SP against Brucella infection could be attributed through induction of cytokine production and interference on intracellular pathway, suggesting SP as a potential candidate for treating brucellosis.

Keywords

Acknowledgement

This work was supported by Korea Institute of Planning and Evaluation for Technology in Food, Agriculture and Forestry (IPET) through Animal Disease Management Technology Development Program, funded by Ministry of Agriculture, Food and Rural Affairs (122001021SB01061382116530000).

References

  1. Hop HT, Reyes AWB, Huy TXN, Arayan LT, Min WG, Lee HJ, et al. 2017. Activation of NF-κB-mediated TNF-induced antimicrobial immunity is required for the efficient Brucella abortus clearance in RAW 264.7 cells. Front. Cell. Infect. Microbiol. 7: 437.
  2. Pasquevich KA, Estein SM, Samartino CG, Zwerdling A, Coria LM, Barrionuevo P, et al. 2009. Immunization with recombinant Brucella species outer membrane protein Omp16 or Omp19 in adjuvant induces specific CD4+ and CD8+ T cells as well as systemic and oral protection against Brucella abortus infection. Infect. Immun. 77: 436-445. https://doi.org/10.1128/IAI.01151-08
  3. He Y. 2012. Analyses of Brucella pathogenesis, host immunity, and vaccine targets using systems biology and bioinformatics. Front. Cell. Infect. Microbiol. 2: 2.
  4. Xavier MN, Winter MG, Spees AM, Nguyen K, Atluri VL, Silva TMA, et al. 2013. CD4+ T cell-derived IL-10 promotes Brucella abortus persistence via modulation of macrophage function. PLoS Pathog. 9: e1003454.
  5. Hop HT, Arayan LT, Reyes AWB, Huy TXN, Min WG, Lee HJ, et al. 2018. Heat-stress-modulated induction of NF-κB leads to brucellacidal pro-inflammatory defense against Brucella abortus infection in murine macrophages and in a mouse model. BMC Microbiol. 18: 44.
  6. Gutierrez MG, Mishra BB, Jordao L, Elliott E, Anes E, Griffiths G. 2008. NF-κB activation controls phagolysosome fusion-mediated killing of mycobacteria by macrophages. J. Immunol. 181: 2651-2663. https://doi.org/10.4049/jimmunol.181.4.2651
  7. Cox MA, Jackson J, Stanton M, Rojas-Triana A, Bober L, Laverty M, et al. 2009. Short-chain fatty acids act as anti-inflammatory mediators by regulating prostaglandin E2 and cytokines. World J. Gastroenterol. 15: 5549-5557. https://doi.org/10.3748/wjg.15.5549
  8. Wang J, Wei Z, Zhang X, Wang Y, Yang Z, Fu Y. 2017. Propionate protects against lipopolysaccharide-induced mastitis in mice by restoring blood-milk barrier disruption and suppressing inflammatory response. Front. Immunol. 8: 1108.
  9. Stranahan LW, Khalaf OH, Garcia-Gonzalez DG, Arenas-Gamboa AM. 2019. Characterization of Brucella canis infection in mice. PLoS One 14: e0218809.
  10. Inan MS, Rasoulpour RJ, Yin L, Hubbard AK, Rosenberg DW, Giardina C. 2000. The luminal short-chain fatty acid butyrate modulates NF-κB activity in a human colonic epithelial cell line. Gastroenterology 118: 724-734. https://doi.org/10.1016/S0016-5085(00)70142-9
  11. Liu T, Li J, Liu Y, Xiao N, Suo H, Xie K, et al. 2012. Short-chain fatty acids suppress lipopolysaccharide-induced production of nitric oxide and proinflammatory cytokines through inhibition of NF-κB pathway in RAW264.7 cells. Inflammation 35: 1676-1684. https://doi.org/10.1007/s10753-012-9484-z
  12. Celli J. 2006. Surviving inside a macrophage: the many ways of Brucella. Res. Microbiol. 157: 93-98. https://doi.org/10.1016/j.resmic.2005.10.002
  13. Jeong S, Kim HY, Kim AR, Yun CH, Han SH. 2019. Propionate ameliorates Staphylococcus aureus skin infection by attenuating bacterial growth. Front. Microbiol. 10: 1363.
  14. Guinan J, Wang S, Hazbun TR, Yadav H, Thangamani S. 2019. Antibiotic-induced decreases in the levels of microbial-derived short-chain fatty acids correlate with increased gastrointestinal colonization of Candida albicans. Sci. Rep. 9: 8872.
  15. Zhang J, Zhang H, Liu M, Lan Y, Sun H, Mai K, et al. 2020. Short-chain fatty acids promote intracellular bactericidal activity in head kidney macrophages from Turbot (Scophthalmus maximus L.) via hypoxia inducible factor-1α. Front. Immunol. 11: 615536.
  16. Vinolo MAR, Rodrigues HG, Hatanaka E, Sato FT, Sampaio SC, Curi R. 2011. Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils. J. Nutr. Biochem. 22: 849-855. https://doi.org/10.1016/j.jnutbio.2010.07.009
  17. Meijer K, De Vos P, Priebe MG. 2010. Butyrate and other short-chain fatty acids as modulators of immunity: what relevance for health? Curr. Opin. Clin. Nutr. Metab. Care 13: 715-721. https://doi.org/10.1097/MCO.0b013e32833eebe5
  18. Silva LG, Ferguson BS, Avila AS, Faciola AP. 2018. Sodium propionate and sodium butyrate effects on histone deacetylase (HDAC) activity, histone acetylation, and inflammatory gene expression in bovine mammary epithelial cells. J. Anim. Sci. 96: 5244-5252.
  19. Jiang X, Baldwin CL. 1993. Effects of cytokines on intracellular growth of Brucella abortus. Infect. Immun. 61: 124-134. https://doi.org/10.1128/iai.61.1.124-134.1993
  20. Van Deun K, Haesebrouck F, Van Immerseel F, Ducatelle R, Pasmans F. 2008. Short-chain fatty acids and L-lactate as feed additives to control Campylobacter jejuni infections in broilers. Avian Pathol. 37: 379-383. https://doi.org/10.1080/03079450802216603
  21. Filippone A, Lanza M, Campolo M, Casili G, Paterniti I, Cuzzocrea S, et al. 2020. The anti-inflammatory and antioxidant effects of sodium propionate. Int. J. Mol. Sci. 21: 3026.
  22. Luhrs H, Gerke T, Muller JG, Melcher R, Schauber J, Boxberger F, et al. 2002. Butyrate inhibits NF-κB activation in lamina propria macrophages of patients with ulcerative colitis. Scand. J. Gastroenterol. 37: 458-466. https://doi.org/10.1080/003655202317316105
  23. Singh N, Thangaraju M, Prasad PD, Martin PM, Lambert NA, Boettger T, et al. 2010. Blockade of dendritic cell development by bacterial fermentation products butyrate and propionate through a transporter (Slc5a8)-dependent inhibition of histone deacetylases. J. Biol. Chem. 285: 27601-27608. https://doi.org/10.1074/jbc.M110.102947