Heterogeneity in tuberculosis pathology, microenvironments and therapeutic responses

Anne Lenaerts, Clifton E Barry 3rd, Véronique Dartois, Anne Lenaerts, Clifton E Barry 3rd, Véronique Dartois

Abstract

Tuberculosis (TB) lesions are extremely complex and dynamic. Here, we review the multiple types and fates of pulmonary lesions that form following infection by Mycobacterium tuberculosis and the impact of this spatial and temporal heterogeneity on the bacteria they harbor. The diverse immunopathology of granulomas and cavities generates a plethora of microenvironments to which M. tuberculosis bacilli must adapt. This in turn affects the replication, metabolism, and relative density of bacterial subpopulations, and consequently their respective susceptibility to chemotherapy. We outline recent developments that support a paradigm shift in our understanding of lesion progression. The simple model according to which lesions within a single individual react similarly to the systemic immune response no longer prevails. Host-pathogen interactions within lesions are a dynamic process, driven by subtle and local differences in signaling pathways, resulting in diverging trajectories of lesions within a single host. The spectrum of TB lesions is a continuum with a large overlap in the lesion types found in latently infected and active TB patients. We hope this overview will guide TB researchers in the design, choice of read-outs, and interpretation of future studies in the search for predictive biomarkers and novel therapies.

Keywords: animal models; biomarkers; immunopathology; lesion dynamics; tuberculosis.

© 2015 The Authors. Immunological Reviews Published by John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
(A). Early non-necrotizing granuloma; the lymphocyte rim (L) appears bluish owing to the prominent nuclei and small cytoplasm; the center of the granuloma contains a majority of epitheloid histiocytes (H) or macrophages, which are large cells with pale abundant cytoplasm. (B). Necrotizing granuloma: the central necrosis appears as pink amorphous material (N), surrounded by a layer of cellular infiltrates as well as a fibrotic rim (F) which clearly defines the granuloma from the surrounding lung alveoli. (C). Masson's trichrome stain of a necrotizing granuloma showing the collagenous areas in blue. The thick fibrotic layer (F) is composed of fibroblasts, collagen and new vessels, and scattered chronic inflammatory cells. Note the prominent vascularization of the fibrotic layer. (D). Calcified granuloma with sclerotic rim (S) associated with absence of mononuclear infiltrates and no visible infectious activity; Ca: area of calcification/mineralization. (E). Tuberculous pneumonia: confluent aggregates of epitheloid histiocytes mixed with Langhans’ and multinuclear giant cells (arrows). The more bluish areas are lymphoid aggregates. These are the same cells that make up a granuloma, but in the absence of a fibrotic wall they have spread to fill alveolar space. (F). Wall of cavitating tuberculous granuloma demonstrating a trilaminar structure with inner necrosis (N), histiocyte aggregates (H) and peripheral fibrosis (F). The fibrotic wall contains mononuclear infiltrates which indicates a still active and progressing tuberculous lesion. The more friable caseous material has been removed during tissue processing.
Figure 2
Figure 2
TB lesions evolve independently in humans. This patient had non-responsive extensively drug-resistant TB and was enrolled into the delayed arm of a randomized clinical trial of linezolid. The subject had failed to respond to any chemotherapy and was hospitalized for 2 months while awaiting study drug. Upon admission, his baseline (18F)-2-fluorodeoxyglucose positron emission tomography (FDG-PET) computed tomography (CT) scan showed extensive bilateral disease. A coronal section of this subjects’ scan is shown in (A), projected at a maximum SUV (Standardized Uptake Value, a method to standardize FDG uptake accounting for body weight and decay time) of 10. The same coronal slice at the same SUV is shown from a repeated scan after 2 months (immediately prior to starting linezolid therapy) in (B). Apical lesions in the left lung (right side of PET/CT image) are resolving while apical lesions in the right lung have progressed. (C, D) The same scan data are presented from the full 3D volume of the chest cavity. White represents air, either in the trachea and airways or inside of cavitary lesions. Dark gray represents areas of radiodensity between −100 and 200 Hounsfeld Units associated with TB lesions in both lungs. FDG uptake is represented by light yellow (SUV 2-3) darker yellow (SUV 3-5) and red (SUV >5). These are posterior views, reversed from above for clarity. Each lesion complex appears to progress or regress independently of the others. The complex in the left superior lobe (on the left in these drawings) appears to resolve while the lesions in the right upper and lower lobes appear to progress significantly. The lesion in the left inferior lobe appears relatively stable with little change.
Figure 3
Figure 3
Heterogeneity in lesion types observed in C3HeB/FeJ mice infected with M. tuberculosis Erdman. Type I lesions (A) in C3HeB/FeJ mice are defined as the caseous necrotic lesions composed of a neutrophil-dominated central core region that degenerates over time into an amorphous, acellular caseum surrounded by a band of intact neutrophils and a distinct rim of foamy macrophages at the peripheral margin. The foamy macrophages contain numerous intracellular bacilli, while large numbers of extracellular bacilli are dispersed within the acellular caseum. The core region is encapsulated by a collagen rim deposited by fibroblasts intermixed with epithelioid and activated macrophages, and few lymphocytes. Type II lesions (B) observed in C3HeB/FeJ mice closely resemble PMN alveolitis occasionally observed in human tuberculosis patients, as described by Canetti (2). These lesions are predominantly composed of necrotizing neutrophils, but lack the fibrotic encapsulation seen in the Type I lesions. Type II lesions present as fulminant granulocytic pneumonia, containing high bacterial numbers extracellular and intracellular within in neutrophils, and very few if any detectable lymphocytes, and negatively impact mouse survival. Type III lesions (C) in C3HeB/FeJ mice are similar to lesions observed in BALB/c mice following aerosol infection. These inflammatory lesions are composed predominantly of epithelioid and foamy macrophages with large numbers of lymphocytes present, only containing few bacteria which are primarily located within macrophages.

References

    1. Barnes DS. The Making of a Social Disease – Tuberculosis in Nineteenth-Century France. Berkeley, Los Angeles, London: University of California Press; 1995.
    1. Canetti G. The Tubercle Bacillus. New York: Springer Publishing COmpany; 1955.
    1. Dannenberg AM., Jr . Pathogenesis of Human Pulmonary Tuberculosis. Washington, DC: ASM Press; 2006.
    1. Via LE, et al. Infection dynamics and response to chemotherapy in a Rabbit model of tuberculosis using (18F)2-fluoro-deoxy-D-glucose positron emission tomography and computed tomography. Antimicrob Agents Chemother. 2012;56:4391–4402.
    1. Via LE, et al. Differential virulence and disease progression following Mycobacterium tuberculosis complex infection of the common marmoset (Callithrix jacchus) Infect Immun. 2013;81:2909–2919.
    1. Bagci U, et al. A computational pipeline for quantification of pulmonary infections in small animal models using serial PET-CT imaging. EJNMMI Res. 2013;3:55.
    1. Murawski AM, et al. Imaging the evolution of reactivation pulmonary tuberculosis in mice using 18F-FDG PET. J Nucl Med. 2014 doi: .
    1. Esmail H, et al. The ongoing challenge of latent tuberculosis. Philos Trans R Soc Lond B Biol Sci. 2014;369:20130437.
    1. Lawn SD, Wood R, Wilkinson RJ. Changing concepts of “latent tuberculosis infection” in patients living with HIV infection. Clin Dev Immunol. 2011;2011 doi: .
    1. Barry CER, et al. The spectrum of latent tuberculosis: rethinking the biology and intervention strategies. Nat Rev Microbiol. 2009;7:845–855.
    1. Young DB, Gideon HP, Wilkinson RJ. Eliminating latent tuberculosis. Trends Microbiol. 2009;17:183–188.
    1. Capuano SV, 3rd, 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. Lin PL, et al. Quantitative comparison of active and latent tuberculosis in the cynomolgus macaque model. Infect Immun. 2009;77:4631–4642.
    1. Berry MP, et al. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature. 2010;466:973–977.
    1. Bloom CI, et al. Transcriptional blood signatures distinguish pulmonary tuberculosis, pulmonary sarcoidosis, pneumonias and lung cancers. PLoS ONE. 2013;8:e70630.
    1. Lee M, et al. Linezolid for treatment of chronic extensively drug-resistant tuberculosis. N Engl J Med. 2012;367:1508–1518.
    1. Coleman MT, et al. PET/CT monitoring demonstrates a therapeutic response to oxazolidinones in Mycobacterium tuberculosis infected macaques and humans. Sci Transl Med. 2014;6:265ra167.
    1. Aber VR, Nunn AJ. Short term chemotherapy of tuberculosis. Factors affecting relapse following short term chemotherapy. Bull Int Union Tuberc. 1978;53:276–280.
    1. Chang KC, et al. A nested case-control study on treatment-related risk factors for early relapse of tuberculosis. Am J Respir Crit Care Med. 2004;170:1124–1130.
    1. Benator D, et al. Rifapentine and isoniazid once a week versus rifampicin and isoniazid twice a week for treatment of drug-susceptible pulmonary tuberculosis in HIV-negative patients: a randomised clinical trial. Lancet. 2002;360:528–534.
    1. Kramnik I, et al. Genetic control of resistance to experimental infection with virulent Mycobacterium tuberculosis. Proc Natl Acad Sci USA. 2000;97:8560–8565.
    1. Driver ER, et al. Evaluation of a mouse model of necrotic granuloma formation using C3HeB/FeJ mice for testing of drugs against Mycobacterium tuberculosis. Antimicrob Agents Chemother. 2012;56:3181–3195.
    1. Davis SL, et al. Noninvasive pulmonary (18F)-2-fluoro-deoxy-D-glucose positron emission tomography correlates with bactericidal activity of tuberculosis drug treatment. Antimicrob Agents Chemother. 2009;53:4879–4884.
    1. Irwin SM, et al. Limited activity of clofazimine as a single drug in a mouse model of tuberculosis exhibiting caseous necrotic granulomas. Antimicrob Agents Chemother. 2014;58:4026–4034.
    1. Skerry C, et al. Adjunctive TNF inhibition with standard treatment enhances bacterial clearance in a murine model of necrotic TB granulomas. PLoS ONE. 2012;7:e39680.
    1. Vilaplana C, et al. Ibuprofen therapy resulted in significantly decreased tissue bacillary loads and increased survival in a new murine experimental model of active tuberculosis. J Infect Dis. 2013;208:199–202.
    1. Marzo E, et al. Damaging role of neutrophilic infiltration in a mouse model of progressive tuberculosis. Tuberculosis. 2014;94:55–64.
    1. Obregon-Henao A, et al. Gr1(int)CD11b+ myeloid-derived suppressor cells in Mycobacterium tuberculosis infection. PLoS ONE. 2013;8:e80669.
    1. Lin PL, et al. Tumor necrosis factor neutralization results in disseminated disease in acute and latent Mycobacterium tuberculosis infection with normal granuloma structure in a cynomolgus macaque model. Arthritis Rheum. 2010;62:340–350.
    1. Lin PL, et al. Radiologic responses in cynomolgous macaques for assessing tuberculosis chemotherapy regimens. Antimicrob Agents Chemother. 2013;57:4237–4244.
    1. Sharpe SA, et al. Establishment of an aerosol challenge model of tuberculosis in rhesus macaques and an evaluation of endpoints for vaccine testing. Clin Vaccine Immunol. 2010;17:1170–1182.
    1. Darrah PA, et al. Aerosol vaccination with AERAS-402 elicits robust cellular immune responses in the lungs of rhesus macaques but fails to protect against high-dose Mycobacterium tuberculosis challenge. J Immunol. 2014;193:1799–1811.
    1. Manabe YC, et al. Different strains of Mycobacterium tuberculosis cause various spectrums of disease in the rabbit model of tuberculosis. Infect Immun. 2003;71:6004–6011.
    1. Manabe YC, et al. The aerosol rabbit model of TB latency, reactivation and immune reconstitution inflammatory syndrome. Tuberculosis. 2008;88:187–196.
    1. Subbian S, et al. Spontaneous latency in a rabbit model of pulmonary tuberculosis. Am J Pathol. 2012;181:1711–1724.
    1. Subbian S, et al. Chronic pulmonary cavitary tuberculosis in rabbits: a failed host immune response. Open Biol. 2011;1:110016. doi: .
    1. Kjellsson MC, et al. Pharmacokinetic evaluation of the penetration of antituberculosis agents in rabbit pulmonary lesions. Antimicrob Agents Chemother. 2012;56:446–457.
    1. Lin PL, et al. Sterilization of granulomas is common in active and latent tuberculosis despite within-host variability in bacterial killing. Nat Med. 2014;20:75–79.
    1. Tobin DM, Ramakrishnan L. TB: the Yin and Yang of lipid mediators. Curr Opin Pharmacol. 2013;13:641–645.
    1. Mayer-Barber KD, et al. Host-directed therapy of tuberculosis based on interleukin-1 and type I interferon crosstalk. Nature. 2014;511:99–103.
    1. Behar SM, Divangahi M, Remold HG. Evasion of innate immunity by Mycobacterium tuberculosis: is death an exit strategy? Nat Rev Microbiol. 2010;8:668–674.
    1. Tobin DM, et al. The lta4h locus modulates susceptibility to mycobacterial infection in zebrafish and humans. Cell. 2010;140:717–730.
    1. Tobin DM, et al. Host genotype-specific therapies can optimize the inflammatory response to mycobacterial infections. Cell. 2012;148:434–446.
    1. Lalvani A, Behr MA, Sridhar S. Innate immunity to TB: a druggable balancing act. Cell. 2012;148:389–391.
    1. Mattila JT, et al. Microenvironments in tuberculous granulomas are delineated by distinct populations of macrophage subsets and expression of nitric oxide synthase and arginase isoforms. J Immunol. 2013;191:773–784.
    1. Subbian S, et al. Early innate immunity determines outcome of Mycobacterium tuberculosis pulmonary infection in rabbits. Cell Commun Signal. 2013;11:60.
    1. Palanisamy GS, et al. Clinical strains of Mycobacterium tuberculosis display a wide range of virulence in guinea pigs. Tuberculosis. 2009;89:203–209.
    1. Reed MB, et al. A glycolipid of hypervirulent tuberculosis strains that inhibits the innate immune response. Nature. 2004;431:84–87.
    1. Martineau AR, et al. Neutrophil-mediated innate immune resistance to mycobacteria. J Clin Invest. 2007;117:1988–1994.
    1. Barnes PF, et al. Predictors of short-term prognosis in patients with pulmonary tuberculosis. J Infect Dis. 1988;158:366–371.
    1. Lowe DM, et al. Neutrophilia independently predicts death in tuberculosis. Eur Respir J. 2013;42:1752–1757.
    1. Condos R, et al. Local immune responses correlate with presentation and outcome in tuberculosis. Am J Respir Crit Care Med. 1998;157(3 Pt 1):729–735.
    1. Eum SY, et al. Neutrophils are the predominant infected phagocytic cells in the airways of patients with active pulmonary TB. Chest. 2010;137:122–128.
    1. Lowe DM, et al. Neutrophils in tuberculosis: friend or foe? Trends Immunol. 2012;33:14–25.
    1. Corleis B, et al. Escape of Mycobacterium tuberculosis from oxidative killing by neutrophils. Cell Microbiol. 2012;14:1109–1121.
    1. Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 2013;13:159–175.
    1. Tsuda Y, et al. Three different neutrophil subsets exhibited in mice with different susceptibilities to infection by methicillin-resistant Staphylococcus aureus. Immunity. 2004;21:215–226.
    1. Nandi B, Behar SM. Regulation of neutrophils by interferon-gamma limits lung inflammation during tuberculosis infection. J Exp Med. 2011;208:2251–2262.
    1. Ernst JD. The immunological life cycle of tuberculosis. Nat Rev Immunol. 2012;12:581–591.
    1. Orme IM. A new unifying theory of the pathogenesis of tuberculosis. Tuberculosis. 2014;94:8–14.
    1. Ulrichs T, et al. Modified immunohistological staining allows detection of Ziehl-Neelsen-negative Mycobacterium tuberculosis organisms and their precise localization in human tissue. J Pathol. 2005;205:633–640.
    1. Deb C, et al. A novel in vitro multiple-stress dormancy model for Mycobacterium tuberculosis generates a lipid-loaded, drug-tolerant, dormant pathogen. PLoS ONE. 2009;4:e6077.
    1. Bhatt A, et al. Deletion of kasB in Mycobacterium tuberculosis causes loss of acid-fastness and subclinical latent tuberculosis in immunocompetent mice. Proc Natl Acad Sci USA. 2007;104:5157–5162.
    1. Yuan Y, et al. The effect of oxygenated mycolic acid composition on cell wall function and macrophage growth in Mycobacterium tuberculosis. Mol Microbiol. 1998;29:1449–1458.
    1. Seiler P, et al. Cell-wall alterations as an attribute of Mycobacterium tuberculosis in latent infection. J Infect Dis. 2003;188:1326–1331.
    1. Ryan GJ, et al. Multiple M. tuberculosis phenotypes in mouse and guinea pig lung tissue revealed by a dual-staining approach. PLoS ONE. 2010;5:e11108.
    1. Steingart KR, et al. Fluorescence versus conventional sputum smear microscopy for tuberculosis: a systematic review. Lancet Infect Dis. 2006;6:570–581.
    1. Ryan GJ, Shapiro HM, Lenaerts AJ. Improving acid-fast fluorescent staining for the detection of mycobacteria using a new nucleic acid staining approach. Tuberculosis. 2014;94:511–518.
    1. Munoz-Elias EJ, et al. Replication dynamics of Mycobacterium tuberculosis in chronically infected mice. Infect Immun. 2005;73:546–551.
    1. Galagan JE, et al. The Mycobacterium tuberculosis regulatory network and hypoxia. Nature. 2013;499:178–183.
    1. Wayne LG, Sohaskey CD. Nonreplicating persistence of mycobacterium tuberculosis. Annu Rev Microbiol. 2001;55:139–163.
    1. Boon C, Dick T. How Mycobacterium tuberculosis goes to sleep: the dormancy survival regulator DosR a decade later. Future Microbiol. 2012;7:513–518.
    1. Tsai MC, et al. Characterization of the tuberculous granuloma in murine and human lungs: cellular composition and relative tissue oxygen tension. Cell Microbiol. 2006;8:218–232.
    1. Via LE, et al. Tuberculous granulomas are hypoxic in guinea pigs, rabbits, and non-human primates. Infect Immun. 2008;76:2333–2340.
    1. Aly S, et al. Oxygen status of lung granulomas in Mycobacterium tuberculosis-infected mice. J Pathol. 2006;210:298–305.
    1. Lenaerts AJ, et al. Location of persisting mycobacteria in a Guinea pig model of tuberculosis revealed by r207910. Antimicrob Agents Chemother. 2007;51:3338–3345.
    1. Harper J, et al. Mouse model of necrotic tuberculosis granulomas develops hypoxic lesions. J Infect Dis. 2012;205:595–602.
    1. Voskuil MI, et al. Inhibition of respiration by nitric oxide induces a Mycobacterium tuberculosis dormancy program. J Exp Med. 2003;198:705–713.
    1. 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.
    1. Raleigh JA, et al. Hypoxia and vascular endothelial growth factor expression in human squamous cell carcinomas using pimonidazole as a hypoxia marker. Cancer Res. 1998;58:3765–3768.
    1. Koller F, Leuthardt F. Nekrose und autolyse beitrag zur kenntnis der dystrophischen verkalkung. Klin Wochenschr. 1934;43:1527–1529.
    1. Watanabe S, et al. Fumarate reductase activity maintains an energized membrane in anaerobic Mycobacterium tuberculosis. PLoS Pathog. 2011;7:e1002287.
    1. Kim MJ, et al. Caseation of human tuberculosis granulomas correlates with elevated host lipid metabolism. EMBO Mol Med. 2010;2:258–274.
    1. Deb C, et al. A novel lipase belonging to the hormone-sensitive lipase family induced under starvation to utilize stored triacylglycerol in Mycobacterium tuberculosis. J Biol Chem. 2006;281:3866–3875.
    1. Raju B, et al. Gene expression profiles of bronchoalveolar cells in pulmonary TB. Tuberculosis. 2008;88:39–51.
    1. Pandey AK, Sassetti CM. Mycobacterial persistence requires the utilization of host cholesterol. Proc Natl Acad Sci USA. 2008;105:4376–4380.
    1. Dhouib R, et al. Watching intracellular lipolysis in mycobacteria using time lapse fluorescence microscopy. Biochim Biophys Acta. 2011;1811:234–241.
    1. Daniel J, et al. Mycobacterium tuberculosis uses host triacylglycerol to accumulate lipid droplets and acquires a dormancy-like phenotype in lipid-loaded macrophages. PLoS Pathog. 2011;7:e1002093.
    1. Garton NJ, et al. Cytological and transcript analyses reveal fat and lazy persister-like bacilli in tuberculous sputum. PLoS Med. 2008;5:e75.
    1. Caire-Brandli I, et al. Reversible lipid accumulation and associated division arrest of Mycobacterium avium in lipoprotein-induced foamy macrophages may resemble key events during latency and reactivation of tuberculosis. Infect Immun. 2014;82:476–490.
    1. Simeone R, et al. Phagosomal rupture by Mycobacterium tuberculosis results in toxicity and host cell death. PLoS Pathog. 2012;8:e1002507.
    1. Vergne I, et al. Mechanism of phagolysosome biogenesis block by viable Mycobacterium tuberculosis. Proc Natl Acad Sci USA. 2005;102:4033–4038.
    1. Cappelli G, et al. Profiling of Mycobacterium tuberculosis gene expression during human macrophage infection: upregulation of the alternative sigma factor G, a group of transcriptional regulators, and proteins with unknown function. Res Microbiol. 2006;157:445–455.
    1. Schnappinger D, et al. Transcriptional adaptation of Mycobacterium tuberculosis within macrophages: insights into the phagosomal environment. J Exp Med. 2003;198:693–704.
    1. Tailleux L, et al. Probing host pathogen cross-talk by transcriptional profiling of both Mycobacterium tuberculosis and infected human dendritic cells and macrophages. PLoS ONE. 2008;3:e1403.
    1. Talaat AM, et al. The temporal expression profile of Mycobacterium tuberculosis infection in mice. Proc Natl Acad Sci USA. 2004;101:4602–4607.
    1. Bishai W. Lipid lunch for persistent pathogen. Nature. 2000;406:683–685.
    1. McKinney JD, et al. Persistence of Mycobacterium tuberculosis in macrophages and mice requires the glyoxylate shunt enzyme isocitrate lyase. Nature. 2000;406:735–738.
    1. Munoz-Elias EJ, McKinney JD. Mycobacterium tuberculosis isocitrate lyases 1 and 2 are jointly required for in vivo growth and virulence. Nat Med. 2005;11:638–644.
    1. Marrero J, et al. Gluconeogenic carbon flow of tricarboxylic acid cycle intermediates is critical for Mycobacterium tuberculosis to establish and maintain infection. Proc Natl Acad Sci USA. 2010;107:9819–9824.
    1. Fritz C, et al. Dependence of Mycobacterium bovis BCG on anaerobic nitrate reductase for persistence is tissue specific. Infect Immun. 2002;70:286–291.
    1. Hutter B, Dick T. Molecular genetic characterisation of whiB3, a mycobacterial homologue of a Streptomyces sporulation factor. Res Microbiol. 1999;150:295–301.
    1. Weber I, et al. Anaerobic nitrate reductase (narGHJI) activity of Mycobacterium bovis BCG in vitro and its contribution to virulence in immunodeficient mice. Mol Microbiol. 2000;35:1017–1025.
    1. Kaplan G, et al. Mycobacterium tuberculosis growth at the cavity surface: a microenvironment with failed immunity. Infect Immun. 2003;71:7099–7108.
    1. Weiss C, Tabachnick J, Cohen HP. Mechanism of softening of tubercles. III. Hydrolysis of protein and nucleic acid during anaerobic autolysis of normal and tuberculous lung tissue in vitro. AMA Arch Pathol. 1954;57:179–193.
    1. Rojas-Espinosa O, et al. The role of cathepsin D in the pathogenesis of tuberculosis. A histochemical study employing unlabeled antibodies and the peroxidase-antiperoxidase complex. Am J Pathol. 1974;74:1–17.
    1. Yamamura Y, Ogawa Y, Maeda H. Prevention of tuberculous cavity formation by desensitization with tuberculin-active peptide. Am Rev Respir Dis. 1974;109:594–601.
    1. Luna B, et al. In vivo prediction of tuberculosis cavity formation in rabbits. J Infect Dis. 2014;211:481–485.
    1. Langermans JA, et al. Divergent effect of bacillus Calmette-Guerin (BCG) vaccination on Mycobacterium tuberculosis infection in highly related macaque species: implications for primate models in tuberculosis vaccine research. Proc Natl Acad Sci USA. 2001;98:11497–11502.
    1. Ulrichs T, Kaufmann SH. New insights into the function of granulomas in human tuberculosis. J Pathol. 2006;208:261–269.
    1. Leong FJ. Pathology of tuberculosis in the human lung. In: Leong FJ, Dartois V, Dick T, editors. A Color Atlas of Comparative Pathology of Pulmonary Tuberculosis. CRC Press; 2011. pp. 53–81.
    1. Aly S, et al. Interferon-gamma-dependent mechanisms of mycobacteria-induced pulmonary immunopathology: the role of angiostasis and CXCR3-targeted chemokines for granuloma necrosis. J Pathol. 2007;212:295–305.
    1. Van de Velde S, et al. Contrasting effects of human THP-1 cell differentiation on levofloxacin and moxifloxacin intracellular accumulation and activity against Staphylococcus aureus and Listeria monocytogenes. J Antimicrob Chemother. 2008;62:518–521.
    1. Rey-Jurado E, et al. Activity and interactions of levofloxacin, linezolid, ethambutol and amikacin in three-drug combinations against Mycobacterium tuberculosis isolates in a human macrophage model. Int J Antimicrob Agents. 2013;42:524–530.
    1. Baik J, et al. Multiscale distribution and bioaccumulation analysis of clofazimine reveals a massive immune system-mediated xenobiotic sequestration response. Antimicrob Agents Chemother. 2013;57:1218–1230.
    1. Prideaux B, et al. High-sensitivity MALDI-MRM-MS imaging of moxifloxacin distribution in tuberculosis-infected rabbit lungs and granulomatous lesions. Anal Chem. 2011;83:2112–2118.
    1. Prideaux B, Stoeckli M. Mass spectrometry imaging for drug distribution studies. J Prot. 2012;75:4999–5013.
    1. Skerry C, et al. Simvastatin increases the in vivo activity of the first-line tuberculosis regimen. J Antimicrob Chemother. 2014;69:2453–2457.
    1. Subbian S, et al. Phosphodiesterase-4 inhibition combined with isoniazid treatment of rabbits with pulmonary tuberculosis reduces macrophage activation and lung pathology. Am J Pathol. 2011;179:289–301.
    1. Salamon H, et al. Cutting edge: vitamin D regulates lipid metabolism in Mycobacterium tuberculosis infection. J Immunol. 2014;193:30–34.
    1. Corson MA. Darapladib: an emerging therapy for atherosclerosis. Ther Adv Cardiovasc Dis. 2010;4:241–248.
    1. Serruys PW, et al. Effects of the direct lipoprotein-associated phospholipase A(2) inhibitor darapladib on human coronary atherosclerotic plaque. Circulation. 2008;118:1172–1182.
    1. Ridker PM, Luscher TF. Anti-inflammatory therapies for cardiovascular disease. Eur Heart J. 2014;35:1782–1791.
    1. Fox W, Ellard GA, Mitchison DA. Studies on the treatment of tuberculosis undertaken by the British Medical Research Council tuberculosis units, 1946-1986, with relevant subsequent publications. Int J Tuberc Lung Dis. 1999;3(10 Suppl 2):S231–S279.
    1. Mitchison DA. The action of antituberculosis drugs in short-course chemotherapy. Tubercle. 1985;66:219–225.
    1. Yano T, et al. Reduction of clofazimine by mycobacterial type 2 NADH:quinone oxidoreductase: a pathway for the generation of bactericidal levels of reactive oxygen species. J Biol Chem. 2011;286:10276–10287.
    1. Wallis RS, et al. Whole blood bactericidal activity during treatment of pulmonary tuberculosis. J Infect Dis. 2003;187:270–278.
    1. Zhu T, et al. Population pharmacokinetic/pharmacodynamic analysis of the bactericidal activities of sutezolid (PNU-100480) and its major metabolite against intracellular mycobacterium tuberculosis in ex vivo whole-blood cultures of patients with pulmonary tuberculosis. Antimicrob Agents Chemother. 2014;58:3306–3311.
    1. Dhillon J, et al. Metronidazole has no antibacterial effect in Cornell model murine tuberculosis. Int J Tuberc Lung Dis. 1998;2:736–742.
    1. Lin PL, et al. Metronidazole prevents reactivation of latent Mycobacterium tuberculosis infection in macaques. Proc Natl Acad Sci USA. 2012;109:14188–14193.
    1. Hoff DR, et al. Metronidazole lacks antibacterial activity in guinea pigs infected with Mycobacterium tuberculosis. Antimicrob Agents Chemother. 2008;52:4137–4140.
    1. Carroll MW, et al. Efficacy and safety of metronidazole for pulmonary multidrug-resistant tuberculosis. Antimicrob Agents Chemother. 2013;57:3903–3909.
    1. Ramakrishnan L. Looking within the zebrafish to understand the tuberculous granuloma. Adv Exp Med Biol. 2013;783:251–266.
    1. Swaim LE, et al. Mycobacterium marinum infection of adult zebrafish causes caseating granulomatous tuberculosis and is moderated by adaptive immunity. Infect Immun. 2006;74:6108–6117.
    1. Basaraba RJ. Experimental tuberculosis: the role of comparative pathology in the discovery of improved tuberculosis treatment strategies. Tuberculosis. 2008;88(Suppl 1):S35–S47.
    1. Rosenthal IM, et al. Dose-ranging comparison of rifampin and rifapentine in two pathologically distinct murine models of tuberculosis. Antimicrob Agents Chemother. 2012;56:4331–4340.
    1. Singhal A, et al. BCG induces protection against Mycobacterium tuberculosis infection in the Wistar rat model. PLoS ONE. 2011;6:e28082.
    1. Palanisamy GS, et al. Disseminated disease severity as a measure of virulence of Mycobacterium tuberculosis in the guinea pig model. Tuberculosis. 2008;88:295–306.
    1. Gonzalez-Juarrero M, et al. Experimental aerosol Mycobacterium bovis model of infection in goats. Tuberculosis. 2013;93:558–564.
    1. Sanchez J, et al. Microscopical and immunological features of tuberculoid granulomata and cavitary pulmonary tuberculosis in naturally infected goats. J Comp Pathol. 2011;145:107–117.
    1. Gil O, et al. Granuloma encapsulation is a key factor for containing tuberculosis infection in minipigs. PLoS ONE. 2010;5:e10030.
    1. Kaushal D, et al. The non-human primate model of tuberculosis. J Med Primatol. 2012;41:191–201.
    1. Lin PL, et al. Early events in Mycobacterium tuberculosis infection in cynomolgus macaques. Infect Immun. 2006;74:3790–3803.

Source: PubMed

3
Se inscrever