Granuloma encapsulation is a key factor for containing tuberculosis infection in minipigs

Olga Gil, Ivan Díaz, Cristina Vilaplana, Gustavo Tapia, Jorge Díaz, María Fort, Neus Cáceres, Sergio Pinto, Joan Caylà, Leigh Corner, Mariano Domingo, Pere-Joan Cardona, Olga Gil, Ivan Díaz, Cristina Vilaplana, Gustavo Tapia, Jorge Díaz, María Fort, Neus Cáceres, Sergio Pinto, Joan Caylà, Leigh Corner, Mariano Domingo, Pere-Joan Cardona

Abstract

A transthoracic infection involving a low dose of Mycobacterium tuberculosis has been used to establish a new model of infection in minipigs. The 20-week monitoring period showed a marked Th1 response and poor humoral response for the whole infection. A detailed histopathological analysis was performed after slicing the formalin-fixed whole lungs of each animal. All lesions were recorded and classified according to their microscopic aspect, their relationship with the intralobular connective network and their degree of maturity in order to obtain a dissemination ratio (DR) between recent and old lesions. CFU counts and evolution of the DR with time showed that the proposed model correlated with a contained infection, decreasing from week 9 onwards. These findings suggest that the infection induces an initial Th1 response, which is followed by local fibrosis and encapsulation of the granulomas, thereby decreasing the onset of new lesions. Two therapeutic strategies were applied in order to understand how they could influence the model. Thus, chemotherapy with isoniazid alone helped to decrease the total number of lesions, despite the increase in DR after week 9, with similar kinetics to those of the control group, whereas addition of a therapeutic M. tuberculosis fragment-based vaccine after chemotherapy increased the Th1 and humoral responses, as well as the number of lesions, but decreased the DR. By providing a local pulmonary structure similar to that in humans, the mini-pig model highlights new aspects that could be key to a better understanding tuberculosis infection control in humans.

Conflict of interest statement

Competing Interests: PJC is co-inventor of the patent claiming RUTI® as a therapeutic vaccine. Regulatory approval and further clinical development is currently being undertaken by a spin-off, the biopharmaceutical company Archivel Farma S.L., in collaboration with the Institut Germans Trias i Pujol. PJC is the Scientific Director of this spin-off. The authors confirm that this does not alter their adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. CFU values in the lungs.
Figure 1. CFU values in the lungs.
Data (mean and standard deviation) are presented by comparing an inoculated lobe with others (pictures A and B), lung lymph nodes (C), and extrapulmonary samples (D). Every sample collected weighted 2 g. approximately. * signifies a statistically significant difference in the one-way ANOVA test (p<0.005). CT  =  control group; INH  =  chemotherapy with isoniazid; VAC  =  group treated with vaccine therapy.
Figure 2. Evaluation of the cellular immune…
Figure 2. Evaluation of the cellular immune response by ELISA assay.
ESAT-6 and PPD-specific IFN-γ, IL-12 and TNF-α production after stimulation of PBMCs, as determined by ELISA assay. Means and standard error of the means (SEM) are drawn in gray and connected by dotted lines. The black boxes show the period of INH treatment and the black arrows the time of therapeutic vaccination.
Figure 3. Evaluation of the cellular immune…
Figure 3. Evaluation of the cellular immune response by ELISPOT assay.
Evaluation of ESAT-6, Ag85B, 16 kDa and PPD-specific IFN-γ production after stimulation of PBMCs, as determined by ELISPOT assay. Means and SEM are drawn in black and connected by dotted lines. The black boxes show the period of INH treatment and the black arrows the time of therapeutic vaccination.
Figure 4. Evaluation of the humoral response.
Figure 4. Evaluation of the humoral response.
Humoral response against PPD is presented. The dotted line indicates the background threshold. Means and SEM are drawn in black and connected by dotted lines. The dark-grey boxes show the period of INH treatment and the black arrows the time of therapeutic vaccination.
Figure 5. Macroscopic pathology of the whole…
Figure 5. Macroscopic pathology of the whole lung showing the intralobular septa network.
Macroscopic pathology of the whole lung (A), sliced lobes (B) and the inoculation site (C). White arrows indicate normal septa and the increased thickness of those surrounding lesions (D).
Figure 6. Microscopic evolution of recent lesions,…
Figure 6. Microscopic evolution of recent lesions, showing the relationship between the granulomas and the intralobular septa.
A–D: Phase I lesions; E and F: Phase II lesions. Images A and B show the initial evolution phase where the granuloma touches an intralobular septa but there is still no fibroblast proliferation. This can be seen in images C and D, where the septa increase in thickness and start to surround the granuloma, as shown by the white dotted lines. Images E and F show how the granuloma is finally surrounded by a thick collagenic mantle. Pictures A, C and D were stained with haematoxylin and eosin (H&E), while B, D and F were stained with Masson's trichromic. The original magnification of the large images is ×40 whereas all insets, except F (x100), are magnified ×400. The green arrows show the septa and the dotted white lines the trajectory of the capsule.
Figure 7. Microscopic evolution of the old…
Figure 7. Microscopic evolution of the old lesions.
Once the granuloma is structured, the necrotic process starts and calcification appears. Images A and B (phase III) show a well-structured and encapsulated granuloma with necrotic calcification. Images C and D show lesions of an advanced evolution (phase IV), with granulomas containing a large amount of calcification and fibrosis. Samples A and C were stained with H&E, whereas samples B and D were half stained with Masson's trichromic and von Kossa stain, which shows calcification in black. Pictures E to G were stained with H&E, Masson's trichromic and von Kossa stain, respectively. Lesions 1 and 2 are phase II lesions and differ only in the initial mineralization seen in lesion 2. The other lesions are all phase III lesions that have progressed differently. Original magnification is ×10.
Figure 8. Evolution of the area of…
Figure 8. Evolution of the area of granulomas according to their evolutive phase.
Individual data are presented in relation to the evolutive state of the lesion (A) or its encapsulation status (B). Mean and quartiles are presented in each case. Inter-group differences were determined by Dunn's One Anova on ranks test and are marked with * if statistically significant (p<0.05).
Figure 9. Macroscopic evolution of the lesions.
Figure 9. Macroscopic evolution of the lesions.
Classification of the fixed pulmonary lesions as they appear under the stereoscopic microscope. Considering the histological evolution of the lesions, and taking into account the sequential appearance of encapsulation and calcification, we have divided the lesions into four phases. Phase I is characterized by the presence of cellular infiltration (A and B). The intragranulomatous necrosis, which is characterized by the presence of an opaque zone inside the granuloma (C and D), appears during Phase II and structuration of the lesion starts. Phase III (E and F) involves the onset of calcification, which gives a shiny aspect to the central opacity, which grows in size. These lesions are characterized by the cartilaginous texture of the lesion when touched with the forceps. Phase IV lesions (G and H) are characterized by predominance of the calcification and a thin surrounding infiltration. Original magnification is ×10. Scale bar: 1 mm.
Figure 10. Evolution of recent and old…
Figure 10. Evolution of recent and old lesions.
The results show the median of the values represented in Table 2. The gray bar shows the INH treatment period and vaccine inoculation is represented by the black arrows. CT  =  control group; INH  =  chemotherapy with isoniazid; VAC  =  group treated with vaccine therapy.
Figure 11. Evolution of the dissemination rate…
Figure 11. Evolution of the dissemination rate (DR).
Individual data points are represented by full or open circles in control (CT) and isoniazid-treated (INH) groups respectively, and are adjusted to an exponential regression. The continuous and dotted lines represents the adjustment for the CT (y = 5620.45−1.071·x + 0.5460) and INH (y = 295.62−0.4848·x +0.8793) groups. Both adjustments were statistically significant (p<0.0001)
Figure 12. Characterization of the dual fibrotic…
Figure 12. Characterization of the dual fibrotic responses in granuloma evolution.
Figures A, B and H show reticulin stain of initial Phase I (A) and Phase III granulomas (the peripheral capsule is marked with a green arrow in B and H). C and D present immunostaining with anti CD10 and show an increase in the center (C) or the periphery (D) of Phase I and III granulomas, respectively. E and F also show the differences between these evolutive phases with the same proliferation pattern stained with Ki 67. G shows recognition of the capsule by anti collagen type 1 antibodies, whereas I and J show the identification of myofibroblasts using anti smooth muscle and anti HHF35 antibodies respectively. Original magnification is ×200. Scale bar: 100 µm.

References

    1. WHO. Global tuberculosis control: epidemiology, strategy, financing: WHO report 2009. 2009. Geneva, Switzerland.
    1. Parrish N, Dick J, Bishai W. Mechanisms of latency in Mycobacterium tuberculosis. Trends Microbiol. 1998;6:107–112.
    1. Sterling TR, Bethel J, Goldberg S, Weinfurter P, Yun L, et al. The scope and impact of treatment of latent tuberculosis infection in the United States and Canada. Am J Respir Crit Care Med. 2006;173:927–931.
    1. Bucher HC, Griffith LE, Guyatt GH, Sudre P, Naef M, et al. Isoniazid prophylaxis for tuberculosis in HIV infection: a meta-analysis of randomized controlled trials. Aids. 1999;13:501–507.
    1. Targeted tuberculin testing and treatment of latent tuberculosis infection. This official statement of the American Thoracic Society was adopted by the ATS Board of Directors, July 1999. This is a Joint Statement of the American Thoracic Society (ATS) and the Centers for Disease Control and Prevention (CDC). This statement was endorsed by the Council of the Infectious Diseases Society of America. (IDSA), September 1999, and the sections of this statement. Am J Respir Crit Care Med. 2000;161:S221–247.
    1. Pai M, Riley LW, Colford JM., Jr Interferon-gamma assays in the immunodiagnosis of tuberculosis: a systematic review. Lancet Infect Dis. 2004;4:761–776.
    1. North RJ, Jung YJ. Immunity to tuberculosis. Annu Rev Immunol. 2004;22:599–623.
    1. Orme IM. Mouse and guinea pig models for testing new tuberculosis vaccines. Tuberculosis (Edinb) 2005;85:13–17.
    1. Basaraba RJ. Experimental tuberculosis: the role of comparative pathology in the discovery of improved tuberculosis treatment strategies. Tuberculosis (Edinb) 2008;88(Suppl 1):S35–47.
    1. Wallace JG. The heat resistance of tubercle bacilli in the lungs of infected mice. Am Rev Respir Dis. 1961;83:866–871.
    1. Munoz-Elias EJ, Timm J, Botha T, Chan WT, Gomez JE, et al. Replication dynamics of Mycobacterium tuberculosis in chronically infected mice. Infect Immun. 2005;73:546–551.
    1. Cardona PJ, Llatjos R, Gordillo S, Diaz J, Ojanguren I, et al. Evolution of granulomas in lungs of mice infected aerogenically with Mycobacterium tuberculosis. Scand J Immunol. 2000;52:156–163.
    1. Cardona PJ, Gordillo S, Diaz J, Tapia G, Amat I, et al. Widespread bronchogenic dissemination makes DBA/2 mice more susceptible than C57BL/6 mice to experimental aerosol infection with Mycobacterium tuberculosis. Infect Immun. 2003;71:5845–5854.
    1. Dheda K, Booth H, Huggett JF, Johnson MA, Zumla A, et al. Lung remodeling in pulmonary tuberculosis. J Infect Dis. 2005;192:1201–1209.
    1. Lenaerts AJ, Hoff D, Aly S, Ehlers S, Andries K, et al. Location of persisting mycobacteria in a Guinea pig model of tuberculosis revealed by r207910. Antimicrob Agents Chemother. 2007;51:3338–3345.
    1. Basaraba R, Smith E, Shanley C, Orme I. Pulmonary lymphatics are primary sites of Mycobacterium tuberculosis infection in guinea pigs infected by aerosol. Infect Immun. 2006;74:5397–5401.
    1. Bezuidenhout J, Schneider J. Pathology and pathogenesis of tuberculosis. In: Zumla SHa., editor. Tuberculosis A comprehensive clinical reference. Europe: Saunders Elsevier; 2009. pp. 117–128.
    1. Davis J, Ramakrishnan L. The role of the granuloma in expansion and dissemination of early tuberculous infection. Cell. 2009;136:37–49.
    1. Buddle BM, Skinner MA, Wedlock DN, de Lisle GW, Vordermeier HM, et al. Cattle as a model for development of vaccines against human tuberculosis. Tuberculosis (Edinb) 2005;85:19–24.
    1. Capuano SV, 3rd, Croix DA, Pawar S, Zinovik A, Myers A, et al. Experimental Mycobacterium tuberculosis infection of cynomolgus macaques closely resembles the various manifestations of human M. tuberculosis infection. Infect Immun. 2003;71:5831–5844.
    1. Canetti G. Histobacteriology and its bearing on the therapy of pulmonary tuberculosis. New York: Springer Publishing Company, Inc; 1955. The tubercle bacillus in the pulmonary lesion of man.
    1. Fraser RS. Histology and gross anatomy of the respiratory tract. In: Hamid Q, Shannon J, Martin J, editors. Physiologic basis of respiratory disease. Hamilton: B.C. Decker inc; 2005.
    1. Plopper CG, Harkema JR. The respiratory system and its use in research. The laboratory primate. 2005;30:503–526.
    1. Thoen CO. Tuberculosis in wild and domestic mammals. In: Bloom BR, editor. Tuberculosis: pathogenesis, protection, and control. Washington D.C.: American Society for Microbiology; 1994. pp. 157–162.
    1. Thoen CO, Steele JH. Ames: Iowa State University Press; 1995. Mycobacterium bovis infection in animals and humans.
    1. Bolin CA, Whipple DL, Khanna KV, Risdahl JM, Peterson PK, et al. Infection of swine with Mycobacterium bovis as a model of human tuberculosis. J Infect Dis. 1997;176:1559–1566.
    1. Lurie M, Abramson S, Heppleston A. On the response of genetically resistant and susceptible rabbits to the quantitative inhalation of human type tubercle bacilli and the nature of resistance to tuberculosis. J Exp Med. 1952;95:119–134.
    1. McMurray DN. Hematogenous reseeding of the lung in low-dose, aerosol-infected guinea pigs: unique features of the host-pathogen interface in secondary tubercles. Tuberculosis (Edinb) 2003;83:131–134.
    1. Ulrichs T, Kaufmann S. New insights into the function of granulomas in human tuberculosis. J Pathol. 2006;208:261–269.
    1. Opie EL, Aronson JD. Tubercle bacilli in latent tuberculous lesions and in lung tissue without tuberculous lesions. Archives of Pathology and Laboratory Medicine. 1927;4:1.
    1. Wright AE, Reid S. On the possibility of determining the presence or absence of tubercular infection by the examination of a patient's blood and tissue fluids. 1906. pp. 194–211. Proceedings of the Royal Society of London. Series B, Containing Papers of a Biological Character: Royal Society.
    1. Pottenger FM. St. Louis: The C.V. Mosby Company.; 1934. Tuberculosis in the child and the adult.611
    1. Cardona P. A dynamic reinfection hypothesis of latent tuberculosis infection. Infection. 2009;37:80–86.
    1. Cáceres N, Tapia G, Ojanguren I, Altare F, Gil O, et al. Evolution of foamy macrophages in the pulmonary granulomas of experimental tuberculosis models. Tuberculosis (Edinb) 2009;89:175–182.
    1. Peyron P, Vaubourgeix J, Poquet Y, Levillain F, Botanch C, et al. Foamy macrophages from tuberculous patients' granulomas constitute a nutrient-rich reservoir for M. tuberculosis persistence. PLoS Pathog. 2008;4:e1000204.
    1. Russell DG, Cardona PJ, Kim MJ, Allain S, Altare F. Foamy macrophages and the progression of the human tuberculosis granuloma. Nat Immunol. 2009;10:943–948.
    1. Hinz B, Phan SH, Thannickal VJ, Galli A, Bochaton-Piallat ML, et al. The myofibroblast: one function, multiple origins. Am J Pathol. 2007;170:1807–1816.
    1. Lenzi HL, Kimmel E, Schechtman H, Pelajo-Machado M, Vale BS, et al. Collagen arrangement in hepatic granuloma in mice infected with Schistosoma mansoni: dependence on fiber radiation centers. Braz J Med Biol Res. 1999;32:639–643.
    1. Mullarky IK, Szaba FM, Berggren KN, Kummer LW, Wilhelm LB, et al. Tumor necrosis factor alpha and gamma interferon, but not hemorrhage or pathogen burden, dictate levels of protective fibrin deposition during infection. Infect Immun. 2006;74:1181–1188.
    1. Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, et al. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest. 1998;101:890–898.
    1. Huynh ML, Fadok VA, Henson PM. Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF-beta1 secretion and the resolution of inflammation. J Clin Invest. 2002;109:41–50.
    1. D'Avila H, Roque NR, Cardoso RM, Castro-Faria-Neto HC, Melo RC, et al. Neutrophils recruited to the site of Mycobacterium bovis BCG infection undergo apoptosis and modulate lipid body biogenesis and prostaglandin E production by macrophages. Cell Microbiol. 2008;10:2589–2604.
    1. Wipff PJ, Rifkin DB, Meister JJ, Hinz B. Myofibroblast contraction activates latent TGF-beta1 from the extracellular matrix. J Cell Biol. 2007;179:1311–1323.
    1. Fujita J, Ohtsuki Y, Suemitsu I, Yamadori I, Shigeto E, et al. Immunohistochemical distribution of epithelioid cell, myofibroblast, and transforming growth factor-beta1 in the granuloma caused by Mycobacterium avium intracellulare complex pulmonary infection. Microbiol Immunol. 2002;46:67–74.
    1. Guirado E, Gil O, Cáceres N, Singh M, Vilaplana C, et al. Induction of a specific strong polyantigenic cellular immune response after short-term chemotherapy controls bacillary reactivation in murine and guinea pig experimental models of tuberculosis. Clin Vaccine Immunol. 2008;15:1229–1237.
    1. Torday JS, Rehan VK. The evolutionary continuum from lung development to homeostasis and repair. Am J Physiol Lung Cell Mol Physiol. 2007;292:L608–611.
    1. Nishikawa K, Arai H, Inoue K. Scavenger receptor-mediated uptake and metabolism of lipid vesicles containing acidic phospholipids by mouse peritoneal macrophages. J Biol Chem. 1990;265:5226–5231.
    1. Nishikawa K, Sato Y, Arai H, Inoue K. Mobilization of acyl chains from endogenous cellular phospholipids into cholesteryl esters during foam-cell formation in mouse peritoneal macrophages. Biochim Biophys Acta. 1993;1169:257–263.
    1. Huitema LF, Vaandrager AB. What triggers cell-mediated mineralization? Front Biosci. 2007;12:2631–2645.
    1. Hsu HH, Abbo BG. Role of bicarbonate/CO2 buffer in the initiation of vesicle-mediated calcification: mechanisms of aortic calcification related to atherosclerosis. Biochim Biophys Acta. 2004;1690:118–123.
    1. Cardona P, Julián E, Vallès X, Gordillo S, Muñoz M, et al. Production of antibodies against glycolipids from the Mycobacterium tuberculosis cell wall in aerosol murine models of tuberculosis. Scand J Immunol. 2002;55:639–645.
    1. Cardona P, Amat I, Gordillo S, Arcos V, Guirado E, et al. Immunotherapy with fragmented Mycobacterium tuberculosis cells increases the effectiveness of chemotherapy against a chronical infection in a murine model of tuberculosis. Vaccine. 2005;23:1393–1398.
    1. Guirado E, Amat I, Gil O, Díaz J, Arcos V, et al. Passive serum therapy with polyclonal antibodies against Mycobacterium tuberculosis protects against post-chemotherapy relapse of tuberculosis infection in SCID mice. Microbes Infect. 2006;8:1252–1259.
    1. Ferebee SH. Controlled chemoprophylaxis trials in tuberculosis. A general review. Bibl Tuberc. 1970;26:28–106.
    1. Andersen P. Host responses and antigens involved in protective immunity to Mycobacterium tuberculosis. Scand J Immunol. 1997;45:115–131.
    1. Grosset J. Mycobacterium tuberculosis in the extracellular compartment: an underestimated adversary. Antimicrob Agents Chemother. 2003;47:833–836.
    1. Jindani A, Doré C, Mitchison D. Bactericidal and sterilizing activities of antituberculosis drugs during the first 14 days. Am J Respir Crit Care Med. 2003;167:1348–1354.
    1. Guirado E, Gil O, Caceres N, Singh M, Vilaplana C, et al. Induction of a specific strong polyantigenic cellular immune response after short-term chemotherapy controls bacillary reactivation in murine and guinea pig experimental models of tuberculosis. Clin Vaccine Immunol. 2008;15:1229–1237.
    1. Domingo M, Gil O, Serrano E, Guirado E, Nofrarias M, et al. Effectiveness and safety of a treatment regimen based on isoniazid plus vaccination with Mycobacterium tuberculosis cells' fragments: field-study with naturally Mycobacterium caprae-infected goats. Scand J Immunol. 2009;69:500–507.
    1. Corner LA, Costello E, Lesellier S, O'Meara D, Sleeman DP, et al. Experimental tuberculosis in the European badger (Meles meles) after endobronchial inoculation of Mycobacterium bovis: I. Pathology and bacteriology. Res Vet Sci. 2007;83:53–62.
    1. Krasnow I, Wayne LG. Sputum digestion. I. The mortality rate of tubercle bacilli in various digestion systems. Tech Bull Regist Med Technol. 1966;36:34–37.
    1. Kiernan JA. Bloxham: Scion.; 2008. Histological and histochemical methods: theory and practice. p. xvi, 606.
    1. Diaz I, Mateu E. Use of ELISPOT and ELISA to evaluate IFN-gamma, IL-10 and IL-4 responses in conventional pigs. Vet Immunol Immunopathol. 2005;106:107–112.

Source: PubMed

3
Abonner