1887

Abstract

. Intraocular tuberculosis (IOTB) is a significant cause of visual morbidity in tuberculosis (TB)-endemic countries. Although () has been detected in both the retinal pigment epithelial (RPE) cells and in the intraocular fluid (IOF) in some cases, IOTB is paucibacillary in the vast majority of patients. As a result, pathogenesis in the ocular compartment is poorly defined.

The transcriptional profile of in the ocular compartment will differ from those of in environments that represent earlier stages of infection.

. Our aim is to shed light on the pathogenesis of in a clinically relevant but challenging environment to study.

Whole-genome microarray analysis was performed on grown in an IOF model (artificial IOF; AIOF) over 6 days against reference log phase bacteria grown in 7H9. Results were compared to published transcriptomes in other physiologically relevant environments, e.g. RPE cell line.

replicates slowly in AIOF. Genes involved in active replication and aerobic respiration as well as lipid metabolism were either downregulated or not differentially expressed. Yet, in AIOF downregulated genes of the DosR regulon, indicating the suppression of dormancy, similar to in RPE cells. This transcriptional profile is distinct from the active and virulent transcriptomes of in alveolar epithelial cells and blood.

likely acquires a non-invasive and quiescent phenotype, between active infection and dormancy, upon reaching an extrapulmonary niche, i.e. the ocular environment.

Funding
This study was supported by the:
  • Fogarty International Center (Award FIC/NIHD43TW009588)
    • Principle Award Recipient: SumanLaal
Loading

Article metrics loading...

/content/journal/jmm/10.1099/jmm.0.001649
2023-02-09
2024-05-21
Loading full text...

Full text loading...

References

  1. Shakarchi FI. Ocular tuberculosis: current perspectives. Clin Ophthalmol 2015; 9:2223–2227 [View Article]
    [Google Scholar]
  2. Gupta V, Gupta A, Rao NA. Intraocular tuberculosis--an update. Surv Ophthalmol 2007; 52:561–587 [View Article]
    [Google Scholar]
  3. Helm CJ, Holland GN. Ocular tuberculosis. Surv Ophthalmol 1993; 38:229–256 [View Article] [PubMed]
    [Google Scholar]
  4. Rao NA, Albini TA, Kumaradas M, Pinn ML, Fraig MM et al. Experimental ocular tuberculosis in guinea pigs. Arch Ophthalmol 2009; 127:1162–1166 [View Article]
    [Google Scholar]
  5. Abhishek S, Ryndak MB, Choudhary A, Sharma S, Gupta A et al. Transcriptional signatures of Mycobacterium tuberculosis in mouse model of intraocular tuberculosis. Pathog Dis 2019; 77:ftz045 [View Article]
    [Google Scholar]
  6. Takaki K, Ramakrishnan L, Basu S. A zebrafish model for ocular tuberculosis. PLoS One 2018; 13:e0194982 [View Article]
    [Google Scholar]
  7. Damera SK, Panigrahi RK, Mitra S, Basu S. Role of extracellular mycobacteria in blood-retinal barrier invasion in a Zebrafish model of ocular TB. Pathogens 2021; 10:333 [View Article]
    [Google Scholar]
  8. Rao NA, Saraswathy S, Smith RE. Tuberculous uveitis: distribution of Mycobacterium tuberculosis in the retinal pigment epithelium. Arch Ophthalmol 2006; 124:1777–1779 [View Article]
    [Google Scholar]
  9. Aggarwal K, Agarwal A, Sehgal S, Sharma S, Singh N et al. An unusual presentation of intraocular tuberculosis in a monocular patient: clinicopathological correlation. J Ophthalmic Inflamm Infect 2016; 6:46 [View Article] [PubMed]
    [Google Scholar]
  10. Bajgai P, Sharma SP, Singh R. Auramine-rhodamine stain-detection of Mycobacteria in ocular fluid. Ophthalmol Retina 2017; 1:513 [View Article]
    [Google Scholar]
  11. Macri A, Marini V, Sangalli G, Fucile C, Iester M et al. An artificial aqueous humor as a standard matrix to assess drug concentration in the anterior chamber by high performance liquid chromatography methods. Clin Lab 2015; 61:47–52 [View Article] [PubMed]
    [Google Scholar]
  12. Dubnau E, Fontán P, Manganelli R, Soares-Appel S, Smith I. Mycobacterium tuberculosis genes induced during infection of human macrophages. Infect Immun 2002; 70:2787–2795 [View Article] [PubMed]
    [Google Scholar]
  13. Ryndak MB, Singh KK, Peng Z, Zolla-Pazner S, Li H et al. Transcriptional profiling of Mycobacterium tuberculosis replicating ex vivo in blood from HIV- and HIV+ subjects. PLoS One 2014; 9:e94939 [View Article]
    [Google Scholar]
  14. Schlingemann J, Thuerigen O, Ittrich C, Toedt G, Kramer H et al. Effective transcriptome amplification for expression profiling on sense-oriented oligonucleotide microarrays. Nucleic Acids Res 2005; 33:e29 [View Article]
    [Google Scholar]
  15. Garton NJ, Waddell SJ, Sherratt AL, Lee S-M, Smith RJ et al. Cytological and transcript analyses reveal fat and lazy persister-like bacilli in tuberculous sputum. PLoS Med 2008; 5:e75 [View Article]
    [Google Scholar]
  16. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci 2001; 98:5116–5121 [View Article]
    [Google Scholar]
  17. Abhishek S, Saikia UN, Gupta A, Bansal R, Gupta V et al. Transcriptional profile of Mycobacterium tuberculosis in an in vitro model of intraocular tuberculosis. Front Cell Infect Microbiol 2018; 8:330 [View Article]
    [Google Scholar]
  18. Betts JC, Lukey PT, Robb LC, McAdam RA, Duncan K. Evaluation of a nutrient starvation model of Mycobacterium tuberculosis persistence by gene and protein expression profiling. Mol Microbiol 2002; 43:717–731 [View Article] [PubMed]
    [Google Scholar]
  19. Stewart GR, Wernisch L, Stabler R, Mangan JA, Hinds J et al. Dissection of the heat-shock response in Mycobacterium tuberculosis using mutants and microarraysaaA list of the 100 ORFs most highly induced by heat shock is provided as supplementary data with the online version of this paper ( http://mic.sgmjournals.org). Microbiology 2002; 148:3129–3138 [View Article]
    [Google Scholar]
  20. Dhandayuthapani S. Stress response of genes encoding putative stress signaling molecules of Mycobacterium tuberculosis. Front Biosci 2007; 12:4676–4681 [View Article] [PubMed]
    [Google Scholar]
  21. Klink M, Brzezinska M, Szulc I, Brzostek A, Kielbik M et al. Cholesterol oxidase is indispensable in the pathogenesis of Mycobacterium tuberculosis. PLoS One 2013; 8:e73333 [View Article]
    [Google Scholar]
  22. Cuthbertson L, Nodwell JR. The TetR family of regulators. Microbiol Mol Biol Rev 2013; 77:440–475 [View Article] [PubMed]
    [Google Scholar]
  23. Quigley J, Hughitt VK, Velikovsky CA, Mariuzza RA, El-Sayed NM et al. The cell wall lipid PDIM contributes to phagosomal escape and host cell exit of Mycobacterium tuberculosis. mBio 2017; 8:e00148-17 [View Article]
    [Google Scholar]
  24. Sharma S, Ryndak MB, Aggarwal AN, Yadav R, Sethi S et al. Transcriptome analysis of mycobacteria in sputum samples of pulmonary tuberculosis patients. PLoS One 2017; 12:e0173508 [View Article]
    [Google Scholar]
  25. Korch SB, Contreras H, Clark-Curtiss JE. Three Mycobacterium tuberculosis Rel toxin-antitoxin modules inhibit mycobacterial growth and are expressed in infected human macrophages. J Bacteriol 2009; 191:1618–1630 [View Article] [PubMed]
    [Google Scholar]
  26. Schnappinger D, Ehrt S, Voskuil MI, Liu Y, Mangan JA et al. Transcriptional adaptation of Mycobacterium tuberculosis within macrophages: insights into the phagosomal environment. J Exp Med 2003; 198:693–704 [View Article]
    [Google Scholar]
  27. Vilchèze C, Yan B, Casey R, Hingley-Wilson S, Ettwiller L et al. Commonalities of Mycobacterium tuberculosis transcriptomes in response to defined persisting macrophage stresses. Front Immunol 2022; 13:909904 [View Article]
    [Google Scholar]
  28. Karakousis PC, Yoshimatsu T, Lamichhane G, Woolwine SC, Nuermberger EL et al. Dormancy phenotype displayed by extracellular Mycobacterium tuberculosis within artificial granulomas in mice. J Exp Med 2004; 200:647–657 [View Article] [PubMed]
    [Google Scholar]
  29. Mehra S, Foreman TW, Didier PJ, Ahsan MH, Hudock TA et al. The DosR regulon modulates adaptive immunity and is essential for Mycobacterium tuberculosis persistence. Am J Respir Crit Care Med 2015; 191:1185–1196 [View Article]
    [Google Scholar]
  30. 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]
  31. Lin PL, Flynn JL. The end of the binary era: revisiting the spectrum of tuberculosis. J Immunol 2018; 201:2541–2548 [View Article]
    [Google Scholar]
  32. Ryndak MB, Singh KK, Peng Z, Laal S. Transcriptional profile of Mycobacterium tuberculosis replicating in type II alveolar epithelial cells. PLoS One 2015; 10:e0123745 [View Article]
    [Google Scholar]
  33. Ryndak MB, Laal S. Mycobacterium tuberculosis primary infection and dissemination: a critical role for alveolar epithelial cells. Front Cell Infect Microbiol 2019; 9:299 [View Article]
    [Google Scholar]
  34. Mehta PK, King CH, White EH, Murtagh JJ Jr, Quinn FD. Comparison of in vitro models for the study of Mycobacterium tuberculosis invasion and intracellular replication. Infect Immun 1996; 64:2673–2679 [View Article] [PubMed]
    [Google Scholar]
  35. Barrios-Payán J, Saqui-Salces M, Jeyanathan M, Alcántara-Vazquez A, Castañon-Arreola M et al. Extrapulmonary locations of Mycobacterium tuberculosis DNA during latent infection. J Infect Dis 2012; 206:1194–1205 [View Article]
    [Google Scholar]
  36. Naing C, Mak JW, Maung M, Wong SF, Kassim AIBM. Meta-analysis: the association between HIV infection and extrapulmonary tuberculosis. Lung 2013; 191:27–34 [View Article] [PubMed]
    [Google Scholar]
  37. Mehta S, Peters RP, Smit DP, Gupta V. Ocular tuberculosis in HIV-infected individuals. Ocul Immunol Inflamm 2020; 28:1251–1258 [View Article]
    [Google Scholar]
  38. Garweg JG, Garweg S-DL, Flueckiger F, Jacquier P, Boehnke M. Aqueous humor and serum immunoblotting for immunoglobulin types G, A, M, and E in cases of human ocular toxoplasmosis. J Clin Microbiol 2004; 42:4593–4598 [View Article]
    [Google Scholar]
  39. Wang ZJ, Zhou M, Cao WJ, Ji J, Bi YW et al. Evaluation of the Goldmann-Witmer coefficient in the immunological diagnosis of ocular toxocariasis. Acta Trop 2016; 158:20–23 [View Article] [PubMed]
    [Google Scholar]
  40. Rolando I, Vilchez G, Olarte L, Lluncor M, Carrillo C et al. Brucellar uveitis: intraocular fluids and biopsy studies. Int J Infect Dis 2009; 13:e206–11 [View Article] [PubMed]
    [Google Scholar]
  41. Stiens J, Arnvig KB, Kendall SL, Nobeli I. Challenges in defining the functional, non-coding, expressed genome of members of the Mycobacterium tuberculosis complex. Mol Microbiol 2022; 117:20–31 [View Article] [PubMed]
    [Google Scholar]
  42. Hafneh NA. Investigation of Molecular Factors Involved in Mycobacterial Stress Responses and Non-Replicating Persistence University of Leicester; 2018
    [Google Scholar]
  43. Houghton J, Cortes T, Schubert O, Rose G, Rodgers A et al. A small RNA encoded in the Rv2660c locus of Mycobacterium tuberculosis is induced during starvation and infection. PLoS One 2013; 8:e80047 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jmm/10.1099/jmm.0.001649
Loading
/content/journal/jmm/10.1099/jmm.0.001649
Loading

Data & Media loading...

Supplements

Supplementary material 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