Virulent subspecies subverts macrophages during early stages of infection Free

Abstract

Virulent non-tuberculous Mycobacteria (NTMs) successfully reside and multiply within the phagosomes of phagocytic cells such as monocytes and macrophages. Macrophages play a very important role in the innate clearance of intracellular pathogens including NTMs. Attenuated subsp. 100 enters macrophages but is incapable of escaping these cells via canonical mycobacteria escape mechanisms. Alternatively, virulent subsp. 104 and subsp. are able to modify macrophages to suit their growth, survival and ultimately escape from macrophages, while non-virulent is readily killed by macrophages. In this study we focused on early infection of macrophages with NTMs to determine the phenotypic response of macrophages, M1 or M2 differentiation, and phosphorylation alterations that can affect cellular response to invading bacteria. Our findings indicate that infection of the macrophage with MAH 100 and favours the development of M1 macrophage, a pro-inflammatory phenotype associated with the killing of intracellular pathogens, while infection of the macrophage with MAH 104 and favoured the development of M2 macrophage, an anti-inflammatory phenotype associated with the healing process. Interference with the host post-translational mechanisms, such as protein phosphorylation, is a key strategy used by many intracellular bacterial pathogens to modulate macrophage phenotype and subvert macrophage function. By comparing protein phosphorylation patterns of infected macrophages, we observed that uptake of both MAH 100 and resulted in MARCKS-related protein phosphorylation, which has been associated with macrophage activation. In contrast, in macrophages infected with MAH 104 and , methionine adenosyltransferase IIβ, an enzyme that catalyses the biosynthesis of S-adenosylmethionine, a methyl donor for DNA methylation. Inhibition of DNA methylation with 5-aza-2 deoxycytidine, significantly impaired the survival of MAH 104 in macrophages. Our findings suggest that the virulent MAH 104 and enhance its survival in the macrophage possibly through interference with the epigenome responses.

Funding
This study was supported by the:
  • Microbiology Foundation of San Francisco (Award MA101)
    • Principle Award Recipient: LuizE. Bermudez
Loading

Article metrics loading...

/content/journal/micro/10.1099/mic.0.001133
2022-02-08
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/micro/168/2/mic001133.html?itemId=/content/journal/micro/10.1099/mic.0.001133&mimeType=html&fmt=ahah

References

  1. Zheng C, Fanta CH. Non-tuberculous mycobacterial pulmonary infection in the immunocompetent host. QJM 2013; 106:307–315 [View Article] [PubMed]
    [Google Scholar]
  2. Johnson MM, Odell JA. Nontuberculous mycobacterial pulmonary infections. J Thorac Dis 2014; 6:210–220 [View Article] [PubMed]
    [Google Scholar]
  3. Piersimoni C, Scarparo C. Extrapulmonary infections associated with nontuberculous mycobacteria in immunocompetent persons. Emerg Infect Dis 2009; 15:1351–1358 [View Article] [PubMed]
    [Google Scholar]
  4. Vande Weygaerde Y, Cardinaels N, Bomans P, Chin T, Boelens J et al. Clinical relevance of pulmonary non-tuberculous mycobacterial isolates in three reference centres in Belgium: a multicentre retrospective analysis. BMC Infect Dis 2019; 19:1061 [View Article] [PubMed]
    [Google Scholar]
  5. Lee M-R, Sheng W-H, Hung C-C, Yu C-J, Lee L-N et al. Mycobacterium abscessus Complex Infections in Humans. Emerg Infect Dis 2015; 21:1638–1646 [View Article] [PubMed]
    [Google Scholar]
  6. Saleeb P, Olivier KN. Pulmonary nontuberculous mycobacterial disease: new insights into risk factors for susceptibility, epidemiology, and approaches to management in immunocompetent and immunocompromised patients. Curr Infect Dis Rep 2010; 12:198–203 [View Article] [PubMed]
    [Google Scholar]
  7. Rocco JM, Irani VR. Mycobacterium avium and modulation of the host macrophage immune mechanisms. Int J Tuberc Lung Dis 2011; 15:447–452 [View Article] [PubMed]
    [Google Scholar]
  8. Bermudez LE, Goodman J, Petrofsky M. Role of complement receptors in uptake of Mycobacterium avium by macrophages in vivo: evidence from studies using CD18-deficient mice. Infect Immun 1999; 67:4912–4916 [View Article] [PubMed]
    [Google Scholar]
  9. Arsenault RJ, Maattanen P, Daigle J, Potter A, Griebel P et al. From mouth to macrophage: mechanisms of innate immune subversion by Mycobacterium avium subsp. paratuberculosis. Vet Res 2014; 45:54 [View Article] [PubMed]
    [Google Scholar]
  10. Danelishvili L, Wu M, Stang B, Harriff M, Cirillo SLG et al. Identification of Mycobacterium avium pathogenicity island important for macrophage and amoeba infection. Proc Natl Acad Sci U S A 2007; 104:11038–11043 [View Article] [PubMed]
    [Google Scholar]
  11. Jeffrey B, Rose SJ, Gilbert K, Lewis M, Bermudez LE. Comparative analysis of the genomes of clinical isolates of Mycobacterium avium subsp. hominissuis regarding virulence-related genes. J Med Microbiol 2017; 66:1063–1075 [View Article] [PubMed]
    [Google Scholar]
  12. Shin D-M, Yang C-S, Yuk J-M, Lee J-Y, Kim KH et al. Mycobacterium abscessus activates the macrophage innate immune response via a physical and functional interaction between TLR2 and dectin-1. Cell Microbiol 2008; 10:1608–1621 [View Article] [PubMed]
    [Google Scholar]
  13. Dubois V, Pawlik A, Bories A, Le Moigne V, Sismeiro O et al. Mycobacterium abscessus virulence traits unraveled by transcriptomic profiling in amoeba and macrophages. PLoS Pathog 2019; 15:e1008069 [View Article] [PubMed]
    [Google Scholar]
  14. Koul A, Herget T, Klebl B, Ullrich A et al. Interplay between mycobacteria and host signalling pathways. Nat Rev Microbiol 2004; 2:189–202 [View Article] [PubMed]
    [Google Scholar]
  15. Wagner D, Maser J, Lai B, Cai Z, Barry CE 3rd et al. Elemental analysis of Mycobacterium avium-, Mycobacterium tuberculosis-, and Mycobacterium smegmatis-containing phagosomes indicates pathogen-induced microenvironments within the host cell’s endosomal system. J Immunol 2005; 174:1491–1500 [View Article] [PubMed]
    [Google Scholar]
  16. Atri C, Guerfali FZ, Laouini D. Role of human macrophage polarization in inflammation during infectious diseases. Int J Mol Sci 2018; 19:E1801 [View Article] [PubMed]
    [Google Scholar]
  17. McClean CM, Tobin DM. Macrophage form, function, and phenotype in mycobacterial infection: lessons from tuberculosis and other diseases. Pathog Dis 2016; 74:ftw068 [View Article] [PubMed]
    [Google Scholar]
  18. Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili S-A et al. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol 2018; 233:6425–6440 [View Article] [PubMed]
    [Google Scholar]
  19. McWhorter FY, Davis CT, Liu WF. Physical and mechanical regulation of macrophage phenotype and function. Cell Mol Life Sci 2015; 72:1303–1316 [View Article] [PubMed]
    [Google Scholar]
  20. Thiriot JD, Martinez-Martinez YB, Endsley JJ, Torres AG et al. Hacking the host: exploitation of macrophage polarization by intracellular bacterial pathogens. Pathog Dis 2020; 78:ftaa009 [View Article] [PubMed]
    [Google Scholar]
  21. Refai A, Gritli S, Barbouche M-R, Essafi M et al. Mycobacterium tuberculosis virulent factor ESAT-6 drives macrophage differentiation toward the pro-inflammatory M1 phenotype and subsequently switches it to the anti-inflammatory M2 phenotype. Front Cell Infect Microbiol 2018; 8:327 [View Article] [PubMed]
    [Google Scholar]
  22. Stapels DAC, Hill PWS, Westermann AJ, Fisher RA, Thurston TL et al. Salmonella persisters undermine host immune defenses during antibiotic treatment. Science 2018; 362:1156–1160 [View Article] [PubMed]
    [Google Scholar]
  23. Panagi I, Jennings E, Zeng J, Günster RA, Stones CD et al. Salmonella Effector SteE Converts the Mammalian Serine/Threonine Kinase GSK3 into a Tyrosine Kinase to Direct Macrophage Polarization. Cell Host Microbe 2020; 27:41–53 [View Article] [PubMed]
    [Google Scholar]
  24. Laencina L, Dubois V, Le Moigne V, Viljoen A, Majlessi L et al. Identification of genes required for Mycobacterium abscessus growth in vivo with a prominent role of the ESX-4 locus. Proc Natl Acad Sci U S A 2018; 115:E1002–E1011 [View Article] [PubMed]
    [Google Scholar]
  25. Bhardwaj S, Srivastava N, Sudan R, Saha B et al. Leishmania interferes with host cell signaling to devise a survival strategy. J Biomed Biotechnol 2010; 2010:109189 [View Article] [PubMed]
    [Google Scholar]
  26. El Amri M, Fitzgerald U, Schlosser G. MARCKS and MARCKS-like proteins in development and regeneration. J Biomed Sci 2018; 25:43 [View Article] [PubMed]
    [Google Scholar]
  27. Lee SM, Suk K, Lee WH. Myristoylated alanine-rich C kinase substrate (MARCKS) regulates the expression of proinflammatory cytokines in macrophages through activation of p38/JNK MAPK and NF-κB. Cell Immunol 2015; 296:115–121 [View Article] [PubMed]
    [Google Scholar]
  28. Björkblom B, Padzik A, Mohammad H, Westerlund N, Komulainen E et al. c-Jun N-terminal kinase phosphorylation of MARCKSL1 determines actin stability and migration in neurons and in cancer cells. Mol Cell Biol 2012; 32:3513–3526 [View Article] [PubMed]
    [Google Scholar]
  29. Ramani K, Donoyan S, Tomasi ML, Park S et al. Role of methionine adenosyltransferase α2 and β phosphorylation and stabilization in human hepatic stellate cell trans-differentiation. J Cell Physiol 2015; 230:1075–1085 [View Article] [PubMed]
    [Google Scholar]
  30. Inderlied CB, Young LS, Yamada JK. Determination of in vitro susceptibility of Mycobacterium avium complex isolates to antimycobacterial agents by various methods. Antimicrob Agents Chemother 1987; 31:1697–1702 [View Article]
    [Google Scholar]
  31. Saunders BM, Dane A, Briscoe H, Britton WJ. Characterization of immune responses during infection with Mycobacterium avium strains 100, 101 and the recently sequenced 104. Immunol Cell Biol 2002; 80:544–549 [View Article] [PubMed]
    [Google Scholar]
  32. Torrelles JB, Ellis D, Osborne T, Hoefer A, Orme IM et al. Characterization of virulence, colony morphotype and the glycopeptidolipid of Mycobacterium avium strain 104. Tuberculosis (Edinb) 2002; 82:293–300 [View Article] [PubMed]
    [Google Scholar]
  33. Bertram MA, Inderlied CB, Yadegar S, Kolanoski P, Yamada JK et al. Confirmation of the beige mouse model for study of disseminated infection with Mycobacterium avium complex. J Infect Dis 1986; 154:194–195 [View Article] [PubMed]
    [Google Scholar]
  34. Blanchard JD, Elias V, Cipolla D, Gonda I, Bermudez LE. Effective treatment of Mycobacterium avium subsp. hominissuis and Mycobacterium abscessus species infections in macrophages, biofilm, and mice by using liposomal ciprofloxacin. Antimicrob Agents Chemother 2018; 62:e00440-18 [View Article] [PubMed]
    [Google Scholar]
  35. Kim B-R, Kim B-J, Kook Y-H, Kim B-J et al. Phagosome Escape of Rough Mycobacterium abscessus strains in murine macrophage via phagosomal rupture can lead to type I interferon production and their cell-to-cell spread. Front Immunol 2019; 10:125 [View Article] [PubMed]
    [Google Scholar]
  36. Tomioka H, Tatano Y, Maw WW, Sano C, Kanehiro Y et al. Characteristics of suppressor macrophages induced by mycobacterial and protozoal infections in relation to alternatively activated M2 macrophages. Clin Dev Immunol 2012; 2012:635451 [View Article] [PubMed]
    [Google Scholar]
  37. Xu W, Zhao X, Daha MR, van Kooten C et al. Reversible differentiation of pro- and anti-inflammatory macrophages. Mol Immunol 2013; 53:179–186 [View Article] [PubMed]
    [Google Scholar]
  38. Tarique AA, Logan J, Thomas E, Holt PG, Sly PD et al. Phenotypic, functional, and plasticity features of classical and alternatively activated human macrophages. Am J Respir Cell Mol Biol 2015; 53:676–688 [View Article] [PubMed]
    [Google Scholar]
  39. Alvarez GR, Zwilling BS, Lafuse WP. Mycobacterium avium inhibition of IFN-gamma signaling in mouse macrophages: Toll-like receptor 2 stimulation increases expression of dominant-negative STAT1 beta by mRNA stabilization. J Immunol 2003; 171:6766–6773 [View Article] [PubMed]
    [Google Scholar]
  40. Yadav M, Roach SK, Schorey JS. Increased mitogen-activated protein kinase activity and TNF-alpha production associated with Mycobacterium smegmatis- but not Mycobacterium avium-infected macrophages requires prolonged stimulation of the calmodulin/calmodulin kinase and cyclic AMP/protein kinase A pathways. J Immunol 2004; 172:5588–5597 [View Article] [PubMed]
    [Google Scholar]
  41. Roach SK, Schorey JS. Differential regulation of the mitogen-activated protein kinases by pathogenic and nonpathogenic mycobacteria. Infect Immun 2002; 70:3040–3052 [View Article] [PubMed]
    [Google Scholar]
  42. Schorey JS, Cooper AM. Macrophage signalling upon mycobacterial infection: the MAP kinases lead the way. Cell Microbiol 2003; 5:133–142 [View Article] [PubMed]
    [Google Scholar]
  43. Gutierrez MG, Mishra BB, Jordao L, Elliott E, Anes E et al. NF-kappa B activation controls phagolysosome fusion-mediated killing of mycobacteria by macrophages. J Immunol 2008; 181:2651–2663 [View Article] [PubMed]
    [Google Scholar]
  44. Ribet D, Cossart P. Post-translational modifications in host cells during bacterial infection. FEBS Lett 2010; 584:2748–2758 [View Article] [PubMed]
    [Google Scholar]
  45. Richter E, Mostertz J, Hochgräfe F. Proteomic discovery of host kinase signaling in bacterial infections. Proteomics Clin Appl 2016; 10:994–1010 [View Article] [PubMed]
    [Google Scholar]
  46. Karin M, Hunter T. Transcriptional control by protein phosphorylation: signal transmission from the cell surface to the nucleus. Curr Biol 1995; 5:747–757 [View Article] [PubMed]
    [Google Scholar]
  47. Scholz R, Imami K, Scott NE, Trimble WS, Foster LJ et al. Novel host proteins and signaling pathways in Enteropathogenic E. coli pathogenesis identified by global phosphoproteome analysis. Mol Cell Proteomics 2015; 14:1927–1945 [View Article] [PubMed]
    [Google Scholar]
  48. Nakayasu ES, Tempel R, Cambronne XA, Petyuk VA, Jones MB et al. Comparative phosphoproteomics reveals components of host cell invasion and post-transcriptional regulation during Francisella infection. Mol Cell Proteomics 2013; 12:3297–3309 [View Article] [PubMed]
    [Google Scholar]
  49. Pfalzer AC, Choi S-W, Tammen SA, Park LK, Bottiglieri T et al. S-adenosylmethionine mediates inhibition of inflammatory response and changes in DNA methylation in human macrophages. Physiol Genomics 2014; 46:617–623 [View Article] [PubMed]
    [Google Scholar]
  50. Fol M, Włodarczyk M, Druszczyńska M. Host epigenetics in intracellular pathogen infections. Int J Mol Sci 2020; 21:13 [View Article] [PubMed]
    [Google Scholar]
  51. Xu C, Fang L, Kong Y, Xiao C, Yang M et al. Knockdown of RMI1 impairs DNA repair under DNA replication stress. Biochem Biophys Res Commun 2017; 494:158–164 [View Article] [PubMed]
    [Google Scholar]
  52. Xu C, Wang Y, Wang L, Wang Q, Du L-Q et al. Accumulation and Phosphorylation of RecQ-Mediated Genome Instability Protein 1 (RMI1) at Serine 284 and Serine 292 during Mitosis. Int J Mol Sci 2015; 16:26395–26405 [View Article] [PubMed]
    [Google Scholar]
  53. Yang Y, Liu X, Yin W, Xie D, He W et al. 5-Aza-2′-deoxycytidine enhances the antimicrobial response of vitamin D receptor against Mycobacterium tuberculosis. RSC Adv 2016; 6:61740–61746 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/micro/10.1099/mic.0.001133
Loading
/content/journal/micro/10.1099/mic.0.001133
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF

Most cited Most Cited RSS feed