Main Article Content

Authors

Background:

The hallmark of tuberculosis is the granuloma, an organized cellular accumulation playing a key role in host defense against Mycobacterium tuberculosis. These structures sequester and contain mycobacterial cells preventing active disease, while long term maintenance of granulomas leads to latent disease. Clear understanding on mechanisms involved in granuloma formation and maintenance is lacking.

Objective:

to monitor granuloma formation and to determine gene expression profiles induced during the granulomatous response to M. tuberculosis (H37Ra).

Methods:

We used a previously characterized in vitro human model. Cellular aggregation was followed daily with microscopy and Wright staining for 5 days. Granulomas were collected at 24 h, RNA extracted and hybridized to Affymetrix human microarrays.

Results:

Daily microscopic examination revealed gradual formation of granulomas in response to mycobacterial infection. Granulomatous structures persisted for 96 h, and then began to disappear.

Conclusions:

Microarray analysis identified genes in the innate immune response and antigen presentation pathways activated during the in vitro granulomatous response to live mycobacterial cells, revealing very early changes in gene expression of the human granulomatous response.

 

Reyes, N., Bettin, A., Reyes, I., & Geliebter, J. (2015). Microarray analysis of the in vitro granulomatous response to Mycobacterium tuberculosis H37Ra. Colombia Medica, 46(1), 26–32. https://doi.org/10.25100/cm.v46i1.1570

WHO . Global Tuberculosis. Geneva, Switzerland: 2012.

Parrish NM, Dick JD, Bishai WR. Mechanisms of latency in Mycobacterium tuberculosis. Trends Microbiol. 1998 ;6(3): 107–12.

Glickman MS, Jacobs WR Jr. Microbial pathogenesis of Mycobacterium tuberculosis: dawn of a discipline. Cell. 2001; 104(4): 477–85.

Ramakrishnan L. Revisiting the role of the granuloma in tuberculosis. Nat Rev Immunol. 2012; 12(5): 352–66.

Zhu G, Xiao H, Mohan VP, Tsen F, Salgame P, and Chan J. Gene expression in the tuberculous granuloma: analysis by laser capture microdissection and real-time PCR. Cell Microbiol. 2003; 5(7): 445–53.

Davis JM, Ramakrishnan L. The role of the granuloma in expansion and dissemination of early tuberculous infection. Cell. 2009;136(1):37–49.

Guirado E, Schlesinger LS. Modeling the Mycobacterium tuberculosis granuloma - the critical battlefield in host immunity and disease. Front Immunol. 2013; 4: 98.

Kunkel SL, Lukacs NW, Strieter RM, Chensue SW. Animal models of granulomatous inflammation. Semin Respir Infect. 1998; 13(3): 221–8.

Davis JMC, H Lewis, J. L. Ghori, N. Herbomel, P. Ramakrishnan, L. Real-time visualization of Mycobacterium-macrophage interactions leading to initiation of granuloma formation in zebrafish embryos. Immunity. 2002; 17(6): 693–702.

Kapoor N, Pawar S, Sirakova TD, Deb C, Warren WL, Kolattukudy PE. Human granuloma in vitro model, for TB dormancy and resuscitation. PLoS One. 2013;8(1): e53657.

Puissegur MP, Botanch C, Duteyrat JL, Delsol G, Caratero C, Altare F. An in vitro dual model of mycobacterial granulomas to investigate the molecular interactions between mycobacteria and human host cells. Cell Microbiol. 2004; 6(5): 423–33.

Birkness KA, Guarner J, Sable SB, Tripp RA, Kellar KL, Bartlett J, et al. An in vitro model of the leukocyte interactions associated with granuloma formation in Mycobacterium tuberculosis infection. Immunol Cell Biol. 2007; 85(2): 160–8.

Rivero-Lezcano OM. In vitro infection of human cells with Mycobacterium tuberculosis. Tuberculosis (Edinb) 2012; 93(2): 123–9.

Li C, Hung Wong W. Model-based analysis of oligonucleotide arrays: model validation, design issues and standard error application. Genome Biol. 2001; 2(8): RESEARCH0032.

Dahlquist KD, Salomonis N, Vranizan K, Lawlor SC, Conklin BR. GenMAPP, a new tool for viewing and analyzing microarray data on biological pathways. Nat Genet. 2002; 31(1): 19–20.

Rajeevan MS, Ranamukhaarachchi DG, Vernon SD, Unger ER. Use of real-time quantitative PCR to validate the results of cDNA array and differential display PCR technologies. Methods. 2001; 25(4): 443–51.

Russell DG. Mycobacterium tuberculosis: here today, and here tomorrow. Nat Rev Mol Cell Biol. 2001; 2(8): 569–77.

Russell DG. Who puts the tubercle in tuberculosis. Nat Rev Microbiol. 2007; 5(1): 39–47.

Drage MG, Pecora ND, Hise AG, Febbraio M, Silverstein RL, Golenbock DT, et al. TLR2 and its co-receptors determine responses of macrophages and dendritic cells to lipoproteins of Mycobacterium tuberculosis. Cell Immunol. 2009; 258(1): 29–37.

Vignal C, Guerardel Y, Kremer L, Masson M, Legrand D, Mazurier J, et al. Lipomannans, but not lipoarabinomannans, purified from Mycobacterium chelonae and Mycobacterium kansasii induce TNF-alpha and IL-8 secretion by a CD14-toll-like receptor 2-dependent mechanism. J Immunol. 2003; 171(4): 2014–23.

Quesniaux VJ, Nicolle DM, Torres D, Kremer L, Guerardel Y, Nigou J, et al. Toll-like receptor 2 (TLR2)-dependent-positive and TLR2-independent-negative regulation of proinflammatory cytokines by mycobacterial lipomannans. J Immunol. 2004; 172(7): 4425–34.

Harding CV, Boom WH. Regulation of antigen presentation by Mycobacterium tuberculosis: a role for Toll-like receptors. Nat Rev Microbiol. 2010; 8(4): 296–307.

Bergeron A, Bonay M, Kambouchner M, Lecossier D, Riquet M, Soler P, et al. Cytokine patterns in tuberculous and sarcoid granulomas: correlations with histopathologic features of the granulomatous response. J Immunol. 1997; 159(6): 3034–43.

Law K, Weiden M, Harkin T, Tchou-Wong K, Chi C, Rom WN. Increased release of interleukin-1 beta, interleukin-6, and tumor necrosis factor-alpha by bronchoalveolar cells lavaged from involved sites in pulmonary tuberculosis. Am J Respir Crit Care Med. 1996; 153(2): 799–804.

Mehra S, Pahar B, Dutta NK, Conerly CN, Philippi-Falkenstein K, Alvarez X, et al. Transcriptional reprogramming in nonhuman primate (Rhesus macaque) tuberculosis granulomas. PLoS One. 2010; 5(8): e12266.

Jo EK, Park JK, Dockrell HM. Dynamics of cytokine generation in patients with active pulmonary tuberculosis. Curr Opin Infect Dis. 2003; 16(3): 205–10.

Sachse F, Ahlers F, Stoll W, Rudack C. Neutrophil chemokines in epithelial inflammatory processes of human tonsils. Clin Exp Immunol. 2005; 140(2): 293–300.

Donninger H, Glashoff R, Haitchi HM, Syce JA, Ghildyal R, van Rensburg E, et al. Rhinovirus induction of the CXC chemokine epithelial-neutrophil activating peptide-78 in bronchial epithelium. J Infect Dis. 2003; 187(11): 1809–17.

Jang S, Uzelac A, Salgame P. Distinct chemokine and cytokine gene expression pattern of murine dendritic cells and macrophages in response to Mycobacterium tuberculosis infection. J Leukoc Biol. 2008; 84(5): 1264–70.

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(3): S189–93.

Kaufmann SH. Protection against tuberculosis: cytokines, T cells, and macrophages. Ann Rheum Dis. 2002; 61(2): 54–8.

Lazarevic V, Yankura DJ, DiVito SJ, Flynn JL. Induction of Mycobacterium tuberculosis-specific primary and secondary T-cell responses in interleukin-15-deficient mice. Infect Immun. 2005; 73(5): 2910–22.

O'Kane CM, Boyle JJ, Horncastle DE, Elkington PT, Friedland JS. Monocyte-dependent fibroblast CXCL8 secretion occurs in tuberculosis and limits survival of mycobacteria within macrophages. J Immunol. 2007; 178(6): 3767–76.

Perskvist N, Long M, Stendahl O, Zheng L. Mycobacterium tuberculosis promotes apoptosis in human neutrophils by activating caspase-3 and altering expression of Bax/Bcl-xL via an oxygen-dependent pathway. J Immunol. 2002; 168(12): 6358–65.

Placido R, Mancino G, Amendola A, Mariani F, Vendetti S, Piacentini M, et al. Apoptosis of human monocytes/macrophages in Mycobacterium tuberculosis infection. J Pathol. 1997; 181(1): 31–8.

Behar SM, Martin CJ, Booty MG, Nishimura T, Zhao X, Gan HX, et al. Apoptosis is an innate defense function of macrophages against Mycobacterium tuberculosis. Mucosal Immunol. 2011; 4(3): 279–87.

Lieberman J. Granzyme A activates another way to die. Immunol Rev. 2010; 235(1): 93–104.

Getachew Y, Stout-Delgado H, Miller BC, Thiele DL. Granzyme C supports efficient CTL-mediated killing late in primary alloimmune responses. J Immunol. 2008; 181(11): 7810–7.

Stoeckle C, Gouttefangeas C, Hammer M, Weber E, Melms A, Tolosa E. Cathepsin W expressed exclusively in CD8+ T cells and NK cells, is secreted during target cell killing but is not essential for cytotoxicity in human CTLs. Exp Hematol. 2009; 37(2): 266–75.

Ruttkay-Nedecky B, Nejdl L, Gumulec J, Zitka O, Masarik M, Eckschlager T, et al. The role of metallothionein in oxidative stress. Int J Mol Sci. 2013; 14(3): 6044–66.

Aydemir TB, Blanchard RK, Cousins RJ. Zinc supplementation of young men alters metallothionein, zinc transporter, and cytokine gene expression in leukocyte populations. Proc Natl Acad Sci USA. 2006; 103(6): 1699–704.

Ochieng J, Chaudhuri G. Cystatin superfamily. J Health Care Poor Underserved. 2010; 21(Suppl 1): 51–70.

Volkman HE, Pozos TC, Zheng J, Davis JM, Rawls JF, Ramakrishnan L. Tuberculous granuloma induction via interaction of a bacterial secreted protein with host epithelium. Science. 2010; 327(5964): 466–9.

Elass E, Aubry L, Masson M, Denys A, Guérardel Y, Maes E, et al. Mycobacterial lipomannan induces matrix metalloproteinase-9 expression in human macrophagic cells through a Toll-like receptor 1 (TLR1)/TLR2- and CD14-dependent mechanism. Infect Immun. 2005; 73(10): 7064–8.

Kaufmann SH. The contribution of immunology to the rational design of novel antibacterial vaccines. Nat Rev Microbiol. 2007; 5(7): 491–504.

Martinez AN, Mehra S, Kaushal D. Role of interleukin 6 in innate immunity to Mycobacterium tuberculosis infection. J Infect Dis. 2013; 207(8): 1253–61.

Downloads

Download data is not yet available.
Received 2014-05-08
Accepted 2015-02-02
Published 2015-03-30