1887

Abstract

The DosR/DosS two-component regulatory system of regulates the expression of numerous genes under stress conditions and is important for the long-term survival of in the host. The rv2626c gene of is one of the most strongly induced transcripts of the dormancy regulon. This study focused on the immunological effects and possible function of Rv2626c in maintaining mycobacterial survival under various stress conditions.

We heterologously expressed the Rv2626c protein in by constructing a recombinant strain Ms_rv2626c. The viability of Ms_rv2626c was evaluated both and . Different stress conditions, including acidified sodium nitrite, malachite green, low pH, SDS and lysozyme, were used to evaluate the effect of Rv2626c on bacterial resistance. An assay using a macrophage infection model was utilized to investigate the potential effect of Rv2626c to alter the immune response of host cell and its associated pathways. The effect of Rv2626c on cell necrosis was also explored.

The expression of Rv2626c-enhanced survival under hypoxia and nitric oxide stress , and this enhancement was maintained within macrophages and in mouse tissues. In addition, macrophages infected with expressing Rv2626c showed significantly higher interleukin-1β (IL-1β), IL-6, tumour necrosis factor-α (TNF-α) and inducible nitric oxide synthase (iNOS) expression, as well as a higher level of cell necrosis, compared with the control.

protein Rv2626c plays a significant role in stimulating macrophages to provoke a pro-inflammatory response and in mycobacterial survival during infection.

Loading

Article metrics loading...

/content/journal/jmm/10.1099/jmm.0.000511
2017-07-01
2024-04-25
Loading full text...

Full text loading...

/deliver/fulltext/jmm/66/7/1033.html?itemId=/content/journal/jmm/10.1099/jmm.0.000511&mimeType=html&fmt=ahah

References

  1. Raviglione M, Sulis G. Tuberculosis 2015: burden, challenges and strategy for control and elimination. Infect Dis Rep 2016; 8:6570 [View Article][PubMed]
    [Google Scholar]
  2. Bretl DJ, He H, Demetriadou C, White MJ, Penoske RM et al. MprA and DosR coregulate a Mycobacterium tuberculosis virulence operon encoding Rv1813c and Rv1812c. Infect Immun 2012; 80:3018–3033 [View Article][PubMed]
    [Google Scholar]
  3. He H, Bretl DJ, Penoske RM, Anderson DM, Zahrt TC. Components of the Rv0081-Rv0088 locus, which encodes a predicted formate hydrogenlyase complex, are coregulated by Rv0081, MprA, and DosR in Mycobacterium tuberculosis. J Bacteriol 2011; 193:5105–5118 [View Article][PubMed]
    [Google Scholar]
  4. Chauhan R, Ravi J, Datta P, Chen T, Schnappinger D et al. Reconstruction and topological characterization of the sigma factor regulatory network of Mycobacterium tuberculosis. Nat Commun 2016; 7:11062 [View Article][PubMed]
    [Google Scholar]
  5. He H, Hovey R, Kane J, Singh V, Zahrt TC. MprAB is a stress-responsive two-component system that directly regulates expression of sigma factors SigB and SigE in Mycobacterium tuberculosis. J Bacteriol 2006; 188:2134–2143 [View Article][PubMed]
    [Google Scholar]
  6. Bhattacharya M, Biswas A, das AK. Interaction analysis of TcrX/Y two component system from Mycobacterium tuberculosis. Biochimie 2010; 92:263–272 [View Article][PubMed]
    [Google Scholar]
  7. Sherman DR, Voskuil M, Schnappinger D, Liao R, Harrell MI et al. Regulation of the Mycobacterium tuberculosis hypoxic response gene encoding α-crystallin. Proc Natl Acad Sci USA 2001; 98:7534–7539 [View Article][PubMed]
    [Google Scholar]
  8. Honaker RW, Dhiman RK, Narayanasamy P, Crick DC, Voskuil MI. DosS responds to a reduced electron transport system to induce the Mycobacterium tuberculosis DosR regulon. J Bacteriol 2010; 192:6447–6455 [View Article][PubMed]
    [Google Scholar]
  9. Fallow A, Domenech P, Reed MB. Strains of the East Asian (W/Beijing) lineage of Mycobacterium tuberculosis are DosS/DosT-DosR two-component regulatory system natural mutants. J Bacteriol 2010; 192:2228–2238 [View Article][PubMed]
    [Google Scholar]
  10. Bashir N, Kounsar F, Mukhopadhyay S, Hasnain SE. Mycobacterium tuberculosis conserved hypothetical protein rRv2626c modulates macrophage effector functions. Immunology 2010; 130:34–45 [View Article][PubMed]
    [Google Scholar]
  11. Shi L, Jung YJ, Tyagi S, Gennaro ML, North RJ. Expression of Th1-mediated immunity in mouse lungs induces a Mycobacterium tuberculosis transcription pattern characteristic of nonreplicating persistence. Proc Natl Acad Sci USA 2003; 100:241–246 [View Article][PubMed]
    [Google Scholar]
  12. Danelishvili L, Everman J, Bermudez LE. Mycobacterium tuberculosis PPE68 and Rv2626c genes contribute to the host cell necrosis and bacterial escape from macrophages. Virulence 2016; 7:23–32 [View Article][PubMed]
    [Google Scholar]
  13. Leistikow RL, Morton RA, Bartek IL, Frimpong I, Wagner K et al. The Mycobacterium tuberculosis DosR regulon assists in metabolic homeostasis and enables rapid recovery from nonrespiring dormancy. J Bacteriol 2010; 192:1662–1670 [View Article][PubMed]
    [Google Scholar]
  14. Gengenbacher M, Rao SP, Pethe K, Dick T. Nutrient-starved, non-replicating Mycobacterium tuberculosis requires respiration, ATP synthase and isocitrate lyase for maintenance of ATP homeostasis and viability. Microbiology 2010; 156:81–87 [View Article][PubMed]
    [Google Scholar]
  15. Boon C, Dick T. How Mycobacterium tuberculosis goes to sleep: the dormancy survival regulator DosR a decade later. Future Microbiol 2012; 7:513–518 [View Article][PubMed]
    [Google Scholar]
  16. Boon C, Li R, Qi R, Dick T. Proteins of Mycobacterium bovis BCG induced in the Wayne dormancy model. J Bacteriol 2001; 183:2672–2676 [View Article][PubMed]
    [Google Scholar]
  17. Proudfoot M, Sanders SA, Singer A, Zhang R, Brown G et al. Biochemical and structural characterization of a novel family of cystathionine beta-synthase domain proteins fused to a Zn ribbon-like domain. J Mol Biol 2008; 375:301–315 [View Article][PubMed]
    [Google Scholar]
  18. Janosík M, Kery V, Gaustadnes M, Maclean KN, Kraus JP. Regulation of human cystathionine beta-synthase by S-adenosyl-L-methionine: evidence for two catalytically active conformations involving an autoinhibitory domain in the C-terminal region. Biochemistry 2001; 40:10625–10633 [View Article][PubMed]
    [Google Scholar]
  19. Zhu ZY, Liu Y, Wang HB, Xiao JZ, Qiu YF et al. Expression and diagnostic value of proteins in Mycobacterium tuberculosis. Genet Mol Res 2014; 13:7780–7790 [View Article][PubMed]
    [Google Scholar]
  20. Dheenadhayalan V, Delogu G, Brennan MJ. Expression of the PE_PGRS 33 protein in Mycobacterium smegmatis triggers necrosis in macrophages and enhanced mycobacterial survival. Microbes Infect 2006; 8:262–272 [View Article][PubMed]
    [Google Scholar]
  21. Singh P, Rao RN, Reddy JR, Prasad RB, Kotturu SK et al. PE11, a PE/PPE family protein of Mycobacterium tuberculosis is involved in cell wall remodeling and virulence. Sci Rep 2016; 6:21624 [View Article][PubMed]
    [Google Scholar]
  22. Li W, Liu M, Xie J. Rv3369 Induces cytokine interleukin-1β production and enhances Mycobacterium smegmatis intracellular survival. J Interferon Cytokine Res 2016; 36:140–147 [View Article][PubMed]
    [Google Scholar]
  23. Pelicic V, Reyrat JM, Gicquel B. Generation of unmarked directed mutations in mycobacteria, using sucrose counter-selectable suicide vectors. Mol Microbiol 1996; 20:919–925 [View Article][PubMed]
    [Google Scholar]
  24. Jin Y, Xin Y, Zhang W, Ma Y. Mycobacterium tuberculosis Rv1302 and Mycobacterium smegmatis MSMEG_4947 have WecA function and MSMEG_4947 is required for the growth of M. smegmatis. FEMS Microbiol Lett 2010; 310:54–61 [View Article][PubMed]
    [Google Scholar]
  25. Daim S, Kawamura I, Tsuchiya K, Hara H, Kurenuma T et al. Expression of the Mycobacterium tuberculosis PPE37 protein in Mycobacterium smegmatis induces low tumour necrosis factor alpha and interleukin 6 production in murine macrophages. J Med Microbiol 2011; 60:582–591 [View Article][PubMed]
    [Google Scholar]
  26. Awasthy D, Bharath S, Subbulakshmi V, Sharma U. Alanine racemase mutants of Mycobacterium tuberculosis require D-alanine for growth and are defective for survival in macrophages and mice. Microbiology 2012; 158:319–327 [View Article][PubMed]
    [Google Scholar]
  27. Hu X, Li X, Huang L, Chan J, Chen Y et al. Quantitative proteomics reveals novel insights into isoniazid susceptibility in mycobacteria mediated by a universal stress protein. J Proteome Res 2015; 14:1445–1454 [View Article][PubMed]
    [Google Scholar]
  28. Wayne LG, Hayes LG. An in vitro model for sequential study of shiftdown of Mycobacterium tuberculosis through two stages of nonreplicating persistence. Infect Immun 1996; 64:2062–2069[PubMed]
    [Google Scholar]
  29. Voskuil MI, Schnappinger D, Visconti KC, Harrell MI, Dolganov GM et al. Inhibition of respiration by nitric oxide induces a Mycobacterium tuberculosis dormancy program. J Exp Med 2003; 198:705–713 [View Article][PubMed]
    [Google Scholar]
  30. Hernandez-Abanto SM, Woolwine SC, Jain SK, Bishai WR. Tetracycline-inducible gene expression in mycobacteria within an animal host using modified Streptomyces tcp830 regulatory elements. Arch Microbiol 2006; 186:459–464 [View Article][PubMed]
    [Google Scholar]
  31. Ayala JC, Pimienta E, Rodríguez C, Anné J, Vallín C et al. Use of Strep-tag II for rapid detection and purification of Mycobacterium tuberculosis recombinant antigens secreted by Streptomyces lividans. J Microbiol Methods 2013; 94:192–198 [View Article][PubMed]
    [Google Scholar]
  32. Cole ST, Brosch R, Parkhill J, Garnier T, Churcher C et al. Deciphering the biology of Mycobacterium tuberculosis from the complete genome sequence. Nature 1998; 393:537–544 [View Article][PubMed]
    [Google Scholar]
  33. Hedstrom L. IMP dehydrogenase: structure, mechanism, and inhibition. Chem Rev 2009; 109:2903–2928 [View Article][PubMed]
    [Google Scholar]
  34. Li W, Zhao Q, Deng W, Chen T, Liu M et al. Mycobacterium tuberculosis Rv3402c enhances mycobacterial survival within macrophages and modulates the host pro-inflammatory cytokines production via NF-kappa B/ERK/p38 signaling. PLoS One 2014; 9:e94418 [View Article][PubMed]
    [Google Scholar]
  35. Singh SK, Kumari R, Singh DK, Tiwari S, Singh PK et al. Putative roles of a proline-glutamic acid-rich protein (PE3) in intracellular survival and as a candidate for subunit vaccine against Mycobacterium tuberculosis. Med Microbiol Immunol 2013; 202:365–377 [View Article][PubMed]
    [Google Scholar]
  36. van der Woude AD, Stoop EJ, Stiess M, Wang S, Ummels R et al. Analysis of SecA2-dependent substrates in Mycobacterium marinum identifies protein kinase G (PknG) as a virulence effector. Cell Microbiol 2014; 16:280–295 [View Article][PubMed]
    [Google Scholar]
  37. Garg R, Tripathi D, Kant S, Chandra H, Bhatnagar R et al. The conserved hypothetical protein Rv0574c is required for cell wall integrity, stress tolerance, and virulence of Mycobacterium tuberculosis. Infect Immun 2015; 83:120–129 [View Article][PubMed]
    [Google Scholar]
  38. Rao SP, Alonso S, Rand L, Dick T, Pethe K. The protonmotive force is required for maintaining ATP homeostasis and viability of hypoxic, nonreplicating Mycobacterium tuberculosis. Proc Natl Acad Sci USA 2008; 105:11945–11950 [View Article][PubMed]
    [Google Scholar]
  39. Roupie V, Romano M, Zhang L, Korf H, Lin MY et al. Immunogenicity of eight dormancy regulon-encoded proteins of Mycobacterium tuberculosis in DNA-vaccinated and tuberculosis-infected mice. Infect Immun 2007; 75:941–949 [View Article][PubMed]
    [Google Scholar]
  40. Peña D, Rovetta AI, Hernández Del Pino RE, Amiano NO, Pasquinelli V et al. A Mycobacterium tuberculosis dormancy antigen differentiates latently infected bacillus calmette-guérin-vaccinated individuals. EBioMedicine 2015; 2:884–890 [View Article][PubMed]
    [Google Scholar]
  41. Ni LJ, Wang NN, Zhang LG, Guo YZ, Shi WZ. Evaluation of the effects of active fractions of chinese medicine formulas on IL-1β, IL-6, and TNF-α release from ANA-1 murine macrophages. J Ethnopharmacol 2016; 179:420–431 [View Article][PubMed]
    [Google Scholar]
  42. Luo H, Zeng J, Huang Q, Liu M, Abdalla AE et al. Mycobacterium tuberculosis Rv1265 promotes mycobacterial intracellular survival and alters cytokine profile of the infected macrophage. J Biomol Struct Dyn 2016; 34:585–599 [View Article][PubMed]
    [Google Scholar]
  43. Algood HM, Lin PL, Flynn JL. Tumor necrosis factor and chemokine interactions in the formation and maintenance of granulomas in tuberculosis. Clin Infect Dis 2005; 41:S189–S193 [View Article][PubMed]
    [Google Scholar]
  44. Yihao D, Hongyun H, Maodan T. Latency-associated protein Rv2660c of Mycobacterium tuberculosis augments expression of proinflammatory cytokines in human macrophages by interacting with TLR2. Infect Dis 2015; 47:168–177 [View Article][PubMed]
    [Google Scholar]
  45. Sledziewska J, Rowińska-Zakrzewska E. [The role of gamma interferon in host immune response to tuberculosis]. Pneumonol Alergol Pol 2000; 68:388–393[PubMed]
    [Google Scholar]
  46. Jayaraman P, Sada-Ovalle I, Nishimura T, Anderson AC, Kuchroo VK et al. IL-1β promotes antimicrobial immunity in macrophages by regulating TNFR signaling and caspase-3 activation. J Immunol 2013; 190:4196–4204 [View Article][PubMed]
    [Google Scholar]
  47. Darwin KH, Ehrt S, Gutierrez-Ramos JC, Weich N, Nathan CF. The proteasome of Mycobacterium tuberculosis is required for resistance to nitric oxide. Science 2003; 302:1963–1966 [View Article][PubMed]
    [Google Scholar]
  48. Flesch IE, Hess JH, Oswald IP, Kaufmann SH. Growth inhibition of Mycobacterium bovis by IFN-γ stimulated macrophages: regulation by endogenous tumor necrosis factor-α and by IL-10. Int Immunol 1994; 6:693–700 [View Article][PubMed]
    [Google Scholar]
  49. Schulze-Osthoff K, Ferrari D, Los M, Wesselborg S, Peter ME. Apoptosis signaling by death receptors. Eur J Biochem 1998; 254:439–459 [View Article][PubMed]
    [Google Scholar]
  50. Martinez AN, Mehra S, Kaushal D. Role of interleukin 6 in innate immunity to Mycobacterium tuberculosis infection. J Infect Dis 2013; 207:1253–1261 [View Article][PubMed]
    [Google Scholar]
  51. Engele M, Stössel E, Castiglione K, Schwerdtner N, Wagner M et al. Induction of TNF in human alveolar macrophages as a potential evasion mechanism of virulent Mycobacterium tuberculosis. J Immunol 2002; 168:1328–1337 [View Article][PubMed]
    [Google Scholar]
  52. Denis M, Gregg EO. Recombinant interleukin-6 increases the intracellular and extracellular growth of Mycobacterium avium. Can J Microbiol 1991; 37:479–483 [View Article][PubMed]
    [Google Scholar]
  53. Galagan JE, Minch K, Peterson M, Lyubetskaya A, Azizi E et al. The Mycobacterium tuberculosis regulatory network and hypoxia. Nature 2013; 499:178–183 [View Article][PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jmm/10.1099/jmm.0.000511
Loading
/content/journal/jmm/10.1099/jmm.0.000511
Loading

Data & Media loading...

Supplements

Supplementary File 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error