Добавил:
kiopkiopkiop18@yandex.ru Вовсе не секретарь, но почту проверяю Опубликованный материал нарушает ваши авторские права? Сообщите нам.
Вуз: Предмет: Файл:

5 курс / Пульмонология и фтизиатрия / Clinical_Tuberculosis_Friedman_Lloyd_N_,_Dedicoat

.pdf
Скачиваний:
1
Добавлен:
24.03.2024
Размер:
25.27 Mб
Скачать

70  Pathogenesis of Tuberculosis

\130.\ Rook GAW. Immunity and hypersensitivity. Practitioner. 1983;227:iv.

\131.\ Orme IM. Induction of nonspecific acquired resistance and delayed type hypersensitivity, but not specific acquired resistance, in mice inoculated with killed mycobacterial vaccines. Infect Immun. 1988;56:3310.

\132.\ Cooper AM, Callahan JE, Keen M, Belisle JT, and Orme IM. Expression of memory immunity in the lung following reexposure to Mycobacterium tuberculosis. Tuberc Lung Dis. 1997;78:67.

\133.\ Orme IM, Miller ES, Roberts AD, Furney SK, Griffin JP, Dobos EM, Chi D, Rivoire B, and Brennan PJ. T-lymphocytes mediating protection and cellular cytolysis during the course of Mycobacterium tuberculosis. J Immunol. 1992;148:189.

\134.\ Orme IM. Characteristics and specificity of acquired immunologic memory to MTB infection. J Immunol. 1988;140:3589.

\135.\ Muller I, Cobbold S, Waldmann H, and Kaufmann SME. Impaired resistance to MTB after selective in vivo depletion of L3T4+ and Lyt -2+ T-cells. Infect Immun. 1987;55:2037.

\136.\ Fiorentino DF, Bond MW, and Mosmann TR. Two types of mouse T helper cell IV. TH-2 clones secrete a factor that inhibits cytokine production by TH-1 clones. J Exp Med. 1989;170:2081.

\137.\ Cooper MA, Dalton DK, Stewart TA, Griffin JP, Russell DG, and Orme IM. Disseminated tuberculosis in interferon-γ gene-disrupted mice. J Exp Med. 1993;178:2243.

\138.\ Flynn JL, Chan J, Triebold KJ, Dalton DK, Stewart TA, and Bloom BR. An essential role for interferon-γ in resistance to

Mycobacterium tuberculosis. J Exp Med. 1993;178:2249. \139.\ Flynn JL, Goldstein MM, Triebold KJ, Koller B, and Bloom

BR. Major histocompatibility complex class I-restricted T cells are required for resistance to Mycobacterium tuberculosis infection. Proc Natl Acad Sci USA. 1992;89:12013.

\140.\ Cooper AM, D’Souza C, Frank AA, and Orme IM. The course of Mycobacterium tuberculosis infection in the lungs of mice lacking expression of either perforinor granzymemediated cytolytic mechanisms. Infect Immun. 1997;65:1317.

\141.\ Kaech SM, and Wherry EJ. Heterogeneity and cell-fate decisions in effector and memory CD8+ T cell differentiation during viral infection. Immunity 2007;27:39.

\142.\ Takemoto N, Intlekofer AM, Northrup JT, Wherry EJ, and Reiner SL. Cutting edge: IL-12 inversely regulates T-bet and eomesodermin expression during pathogen-induced CD8+ T cell differentiation. J Immunol. 2006;177:755.

\143.\ Sallusto F, Geginat J, and Lanzavecchia A. Central memory and effector memory T cell subsets: Function, generation, and maintenance. Annu Rev Immunol. 2004;22:745.

\144.\ Sallusto F, Lenig D, Forster R, Lipp M, and Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions Nature. 1999;401:708–12.

145.FritschRD,ShenX,SimsGP,HathcockKS,HodesRJ,andLipsky PE. Stepwise differentiation of CD4 memory T cells defined by expression of CCR7 and CD27. J Immunol. 2005;175:6489.

\146.\ Gattinoni L et al. A human memory T cell subset with stem cell-like properties. Nat Med. 2011 Sep 18;17(10):1290–7. doi: 10.1038/nm.2446.

\147.\ Boom WH, Wallis RS, and Chervenak KA. Human MTBreactive CD4+ T-cell clones: Heterogeneity in antigen recognition, cytokine production, and cytotoxicity for mononuclear phagocytes. Infect Immun. 1991;59:2737.

\148.\ Vilaplana C, Ruiz-Manzano J, Gil O, Cuchillo F, Montané E, Singh M, Spallek R, Ausina V, and Cardona PJ. The tuberculin skin test increases the responses measured by T cell interferon-gamma release assays. Scand J Immunol. 2008;67:610–7.

\ 149.\ Tan JS, Canaday DH, Boom WH, Balaji KN, Schwander SK, and Rich EA. Human alveolar T lymphocyte responses to

Mycobacterium tuberculosis antigens. J Immunol. 1997;159:290. \150.\ Berger HW and Mejia E. Tuberculous pleurisy. Chest.

1973;63:88.

\151.\ Fujiwara H, Okuda Y, Fukukawa T, and Tsuyuguchi I. In vitro tuberculin reactivity of lymphocytes from patients with tuberculous pleurisy, Infect Immun. 1982;35:402.

\152.\ Barnes PF, Mistry SD, Cooper CL, Pirmez C, Rea TH,

and Modlin

RL. Compartmentalization of

a CD4+

T-lymphocyte

subpopulation in tuberculous

pleuritis.

J Immunol. 1989;142:1114.

 

\153.\ Fujiwara H and Tsuyuguchi I. Frequency of tuberculinreactive T-lymphocytes in pleural fluid and blood from patients with tuberculous pleurisy. Chest 1984;89:530.

\154.\ Ellner JJ. Pleural fluid and peripheral blood lymphocyte function in tuberculosis, Ann Int Med. 1978;89:932.

\155.\ Barnes PF, Fong SJ, Brennan PJ, Twomey PE, Mazumder A, and Modlin RL. Local production of tumor necrosis factor and interferon-g in tuberculous pleuritis. J Immunol. 1990;145:149.

\156.\ Wallis RS, Alde SL, Havlir DV, Amir-Tahmasseb H, Daniel TM, and Ellner JJ. Identification of antigens of

Mycobacterium tuberculosis using human monoclonal antibodies. J Clin Invest. 1989;84:214.

\157.\ Nemes E et al. C-040-404 Study Team. Prevention of M. tuberculosis infection with H4:IC31 vaccine or BCG revaccination. N Engl J Med. 2018;379:138.

\158.\ Van Der Meeren O et al. Phase 2b controlled trial of M72/ AS01E vaccine to prevent tuberculosis. N Engl J Med. 2018.

\159.\ Anderseen P. Host responses and antigens involved in protective immunity to Mycobacterium tuberculosis. Scand J Immunol. 1997;45:115.

\160.\ Ottenhoff THM, Kale B, van Embden JDA, Thole JER, and Kiessling R. The recombinant 65 kD heat shock protein of Mycobacterium bovis Bacillus Calmette-Guérin/MTB is a target molecule for CD4+ cytotoxic T-lymphocytes that lyse human monocytes. J Exp Med. 1988;168:1947.

\161.\ Munk ME, Schoel B, and Kaufmann SHE. T cell responses of normal individuals towards recombinant protein antigens of MTB. Eur J Immunol. 1988;18:1835.

\162.\ Havlir DV, Wallis RS, Boom WH, Daniel TM, Chervenak K, and Ellner JJ. Human immune response to MTB antigens. Infect Immun. 1991;59:665.

\163.\ Schoel B, Gulle M, and Kaufmann SHE. Heterogeneity of the repertoire of T cells of tuberculosis patients and healthy contacts to MTB antigens separated by high resolution techniques. Infect Immun. 1992;60:1717.

Книга в списке рекомендаций к покупке и прочтению сайта https://meduniver.com/

References  71

164.Young DB, Kaufmann SHE, Hermans PWM, and Thole JER. Mycobacterial proteins, a compilation. Mol Microbiol. 1992;6:133.

165.Wiker HG, Sletten K, Nagai S, and Harboe M. Evidence for three separate genes encoding the proteins of the mycobacterial antigen 85 complex. Infect Immun. 1990;58:272.

166.Rambukkhan A, Das PK, Chand A, Baas JG, Grothuis DG, and Kold AHJ. Subcellular distribution of monoclonal anti- body-defined epitopes on immunuo dominant 33-kilodal- ton proteins of MTB: Identification and localization of 29/33 kilodalton doublet proteins in mycobacterial cell walls.

Scand J Immunol. 1991;33:763.

167.Abou-Zeid C, Ratliff TL, Wiker HG, Harboe M, Bennedsen J, and Rook GAW. Characterization of fibronectin-binding antigens released by MTB and M. bovis BCG. Infect Immun. 1988;56:3046.

168.Abou-Zeid C, Smith I, Grange JM, Ratliff TL, Steele J, and Rook GAW. The secreted antigens of MTB and their relationship to those recognized by the available antibodies. J Gen Microbiol. 1988;134:531.

169.Wiker HG, Harboe M, and Lea TE. Purification and characterization of two protein antigens from the heterogeneous BCG 85 complex in M. bovis BCG. Int Arch Allerg Appl Immunol. 1986;81:298.

170.Salata RA, Sanson AJ, Malhotra IJ, Wiker HG, Harboe HG, Phillips NB, and Daniel TM. Purification and characterization of the 30,000 dalton native antigen of Mycobacterium tuberculosis and characterization of six monoclonal antibodies reactive with a major epitope of this antigen. J Lab Clin Med. 1991;118:589.

171.Matsuo K, Yamaguchi R, Yamakazi A, Tasaka H, and Yamada T. Cloning and expression of the M. bovis BCG gene for extracellular alpha antigen. J Bacteriol. 1988;160:3847.

172.Pal PG and Horowitz MA. Immunization with extracellular proteins of M. tuberculosis induces cell-mediated immune responses with substantial protective immunity in a guinea pig model of pulmonary tuberculosis. Infect Immun. 1992;60:4782.

173.Huygen K, van Vooren JP, Turneer M, Bosmans R, Dierckx P, and De Bruyn J. Specific lymphoproliferation, gamma interferon production, and serum immunoglobulin G directed against a purified 32 kDA mycobacterial protein antigen (P32) in patients with active tuberculosis. Scand J Immunol. 1988;27:187.

174.Abou-Zeid C, Ratliff TL, Wiker HG, Harboe M, Bennedsen J, and Rook GAW. Characterization of fibronectin-binding antigens released by MTB and M. bovis BCG. Infect Immun. 1988;56:3046.

175.Yuan Y, Lee RE, Besra GS, Belisle JT, and Barry CE, III. Identification of a gene involved in the biosynthesis of cyclopropanated mycolic acids in Mycobacterium tuberculosis.

Proc Natl Acad Sci USA. 1995;92:6630.

176.Wallis RS, Fujiwara H, and Ellner JJ. Direct stimulation of monocyte release of interleukin-1 by mycobacterial protein antigens. J Immunol. 1986;36:193.

177.Toossi Z, Lapurga JP, Ondash R, Sedor JR, and Ellner JJ. Expression of functional interleukin-2 receptors by

peripheral blood monocytes from patients with active pulmonary tuberculosis. J Clin Invest. 1990;85:1777.

178.Toossi Z, Young TG, Averill LE, Hamilton BD, Shiratsuchi H, and Ellner JJ. Induction of transforming growth factor-β (TGF-β) by purified protein derivative (PPD) of

Mycobacterium tuberculosis. Infect Immun. 1995;63:224.

179.Harth G, Clemens DL, and Horwitz MA. Glutamine synthetase of Mycobacterium tuberculosis: Extracellular release and characterization of its enzymatic activity. Proc Natl Acad Sci USA. 1994;91:9342.

180.Wallis RS, Raranjape R, and Phillips M. Identification of 2-D gel electrophoresis of a 58 kD TNF-α-reducing protein of MTB. J Immun. 1993;61:627.

181.Toossi Z, Gogate P, Shiratsuchi H, Young T, and Ellner JJ. Enhanced production of TGF-β by blood monocytes from patients with active tuberculosis and presence of TGF-β in tuberculosis granulomatous lung lesions. J Immunol. 1995;154:465.

182.Rook GAW. Importance of recent advances in our understanding of antimicrobial cell-mediated immunity to the International Union for the Prevention of Tuberculosis. Bull Int Union Tuberc. 1983;58:60.

183.Zugmaier G et al. Transforming growth factor β1 induces cachexia and systemic fibrosis without an antitumor effect in nude mice. Cancer Res. 1991;51:3590.

184.Broekelmann TJ, Limper AH, Colby TV, and McDonald JA. Transforming growth factor β is present at sites of extracellular matrix gene expression in human pulmonary fibrosis.

Proc Natl Acad Sci USA. 1991;88:6642.

185.Border WA and Noble NA. Transforming growth factor β in tissue fibrosis. N Engl J Med. 1994;331:1286.

186.Denis M. Tumor necrosis factor and granulocyte macro- phage-colony stimulating factor stimulates human macrophages to restrict growth of virulent Mycobacterium avium and to kill avirulent M. avium. Killing effector mechanism depends on the generation of reactive nitrogen intermediates. J Leuk Biol. 1991;49:380.

187.Bermudez LE M. and Young LS. Tumor necrosis factor alone or in combination with IL-2 but not IFN-gamma, is associated with macrophage killing of Mycobacterium avium complex. J Immunol. 1988;140:3006.

188.Rook GAW, Steele J, Fraber L, Barker S, Karmali R, O’Riordan J, and Standford J. Vitamin D3, gamma interferon and control of proliferation of Mycobacterium tuberculosis by human monocytes. Immunology 1986;56:159.

189.Toossi Z, Hirsch CS, Hamilton BD, Kunuth CK, Friedlander MA, and Rich EA. Decreased production of transforming growth factor β1 (TGF-β1) in human alveolar macrophages. J Immunol. 1996;156:3461.

190.Kindler V, Sappino AP, Grau GE, Piquet PI, and Vassali P. The reducing role of tumor necrosis factor in the development of bactericidal granulomas during BCG infection. Cell 1989;56:731.

191.Flesch I and Kaufmann SH E. Mycobacterial growth inhibition by interferon-gamma activated bone marrow macrophages and differential susceptibility among strains of MTB. J Immunol. 1987;138:4408.

72  Pathogenesis of Tuberculosis

\192.\ Flesch IE and Kaufmann SH E. Mechanisms involved in mycobacterial growth inhibition by gamma interferon-acti- vated bone marrow macrophages: Role of reactive nitrogen intermediates. Infect Immun. 1991;59:3213.

\193.\ Douvas GS, Looker DL, Vatter AE, and Crowle AJ. Gamma interferon activates human macrophages to become tumoricidal and leishmanicidal but enhances replication of macrophage associated mycobacteria. Infect Immun. 1985;50:1.

\194.\ Rook GAW, Steele J, Fraher L, Barker S, Karmali R, O’Riordan J, and Stanfor J. Vitamin D3, gamma interferon, and control of proliferation of Mycobacterium tuberculosis by human monocytes. Immunology 1986;57:159.

\195.\ Hirsch CS, Yoneda T, Ellner JJ, Averill LE, and Toossi Z. Enhancement of intracellulargrowth of M. tuberculosis in human monocytes by transforming growth factor beta. J Infect Dis. 1994;170:1229.

\196.\ Bermudez LE. Production of transforming growth factor-β by Mycobacterium avium-infected human macrophages is associated with unresponsiveness to IFN-γ. J Immunol. 1993;150:1838.

\197.\ Hirsch CS, Jerrold JE, Blinkhorn R, and Toossi Z. In vitro restoration of T-cell responses in tuberculosis and augmentation of monocyte effector function against Mycobacterium tuberculosis by natural inhibitors of transforming growth factor-β. Proc. Natl. Acad. Sci. USA 1997;94:3926.

\198.\ Ding A, Nathan C, and Srimal S. Macrophage deactivating factor and TGF-β inhibit of macrophage nitrogen oxide synthesis by IFN-γ. J Immunol. 1990;145:940.

\199.\ Toossi Z, Mincek M, Seeholtzer E, Fulton SA, Hamilton BD, and Hirsch CS. Modulation of IL-12 by transforming growth factor-β (TGF-β) in Mycobacterium tuberculosis- infected mononuclear phagocytes and in patients with active tuberculosis. J Clin Lab Immunol. 1997;49:59.

\200.\ O’Garra A, Redford PS, McNab FW, Bloom CI, Wilkinson RJ, and Berry MPR. The immune response in tuberculosis. Annu Rev Immunol. 2013;31(1):475–527

\201.\ Mayer-Barber KD et al. Host-directed therapy of tuberculosis based on interleukin-1 and type I interferon crosstalk. Nature 03 July 2014;511:99–103, doi:10.1038/nature13489

\202.\ Boom WH, Wallis RS, and Chervenak KA. Human MTBreactive CD4+ T-cell clones: Heterogeneity in antigen recognition, cytokine production, and cytotoxicity for mononuclear phagocytes. Infect Immun. 1991;59:2737.

\203.\ Mustafa AS and Godal T. BCG-induced CD4+ cytotoxic T cells from BCG vaccinated healthy subjects: Relation between cytotoxicity and suppression in vitro. Clin Exp Immunol. 1987;69:255.

\204.\ Orme I, Andersen P, and Boom WH. T-cell responses to

Mycobacterium tuberculosis. J Infect Dis. 1993;167:1481. \205.\ Chan J, Mehta S, Bharrhan S, Chen Y, Achkar JM,

Casadevall A, and Flynn J. The role of B cells and humoral immunity in Mycobacterium tuberculosis infection. Semin Immunol. 2014;26(6):588–600. http://doi.org/10.1016/j. smim.2014.10.005

\206.\ Glatman-Freedman A. The role of antibody-mediated immunity in defense against Mycobacterium tuberculosis:

Advances toward a novel vaccine strategy. Tuberculosis (Edinburgh) 2006;86:191–7. [PubMed: 16584923]

\207.\ Achkar JM, Chan J, and Casadevall A. Role of B cells and antibodies in acquired immunity against Mycobacterium tuberculosis. Cold Spring Harb Perspect Med. 2015;5(3):a018432. http://doi.org/10.1101/cshperspect.a018432

\208.\ Maglione PJ, and Chan J. How B cells shape the immune response against Mycobacterium tuberculosis. Eur J Immunol. 2009;39:676–86.

\209.\ Chan J, Mehta S, Bharrhan S, Chen Y, Achkar JM, Casadevall A, and Flynn J. The role of B cells and humoral immunity in Mycobacterium tuberculosis infection. Semin Immunol. 2014;26:588.

\210.\ Li H, Wang XX, Wang B, Fu L, Liu G, Lu Y, Cao M, Huang H, and Javid B. Latently and uninfected healthcare workers exposed to TB make protective antibodies against

Mycobacterium tuberculosis. Proc Natl Acad Sci U S A. 2017;114:5023.

\211.\ Lu LL et al. A functional role for antibodies in tuberculosis. Cell 2016;167:433.

\212.\ Zimmermann N et al. Human isotype-dependent inhibitory antibody responses against Mycobacterium tuberculosis. EMBO Mol Med. 2016;8:1325.

\213.\ Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, Umesaki Y, Littman DR, Benoist C, and Mathis D. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 2010;32:817.

\214.\ Trompette A, Gollwitzer ES, Pattaroni C, Lopez-Mejia IC, Riva E, Pernot J, Ubags N, Fajas L, Nicod LP, and Marsland BJ, Dietary fiber confers protection against flu by shaping Ly6cpatrolling monocyte hematopoiesis and CD8+ T cell metabolism. J Immunity. 2018;4:992.

\215.\ Negatu DA, Liu JJJ, Zimmerman M, Kaya F, Dartois V, Aldrich CC, Gengenbacher M, and Dick T. Whole-cell screen of fragment library identifies gut microbiota metabolite indole propionic acid as antitubercular. Antimicrob Agents Chemother. 2018;62:31.

\216.\ Segal LN et al. Anaerobic bacterial fermentation products increase tuberculosis risk in antiretroviral-drug-treated HIV patients. Cell Host Microbe. 2017;21:530.

\217.\ Daniel TM, Oxtoby MJ, Pinto E, and Moreno E. The immune spectrum in patients with pulmonary tuberculosis. Am Rev Respir Dis. 1981;123:556.

\218.\ Nash DR and Douglass JE. Anergy in pulmonary tuberculosis: Comparison between positive and negative reactors and an evaluation of 5TU and 250 TU skin test doses, Chest, 1980;77:32.

\219.\ Rooney JJ, Crocco JA, Kramer S, and Lyons HA. Further observations on tuberculin reactions in tuberculosis. Am J Med. 1976;60:517.

\ 220.\ Sousa AO, Salem JI, Lee FK, Vercosa MC, Cruad P, Bloom BR, Lagrange PH, and David HL. An epidemic of tuberculosis with a high rate of tuberculin anergy among a population previously unexposed to tuberculosis, the Yanomami Indians of the Brazilian Amazon. Proc Natl Acad Sci USA. 1997;94:13227.

\221.\ Ellner JJ. Suppressor adherent cells in human tuberculosis. J Immunol. 1978;121:2573.

Книга в списке рекомендаций к покупке и прочтению сайта https://meduniver.com/

References  73

222.Kleinhenz ME and Ellner JJ. Antigen responsiveness during tuberculosis: Regulatory interaction of T-cell subpopulations and adherent cells. J Lab Clin Med. 1987;110:31.

223.Kleinhenz ME and Ellner JJ. Immunoregulatory adherent cells in human tuberculosis: Radiation sensitive antigenspecific suppression by monocytes. J Infect Dis. 1985;152:171.

224.Toossi Z, Edmonds KL, Tomford WJ, and Ellner JJ. Suppression of PPD-induced interleukin2 production by interaction of Leu-22 (CD16) lymphocytes and adherent mononuclear cells in tuberculosis. J Infect Dis. 1989;159:352.

225.Toossi Z, Kleinhenz ME, and Ellner JJ. Defective interleu- kin-2 production and responsiveness in human pulmonary tuberculosis. J Exp Med. 1986;163:1162.

226.Josefowicz SZ, Lu LF, and Rudensky AY. Regulatory T cells: Mechanisms of differentiation and function. Annu Rev Immunol. 2012;30:531–64. https://doi.org/10.1146/annurev. immunol.25.022106.141623

227.Ribeiro-RodriguesR,ResendeCoT,RojasR,ToossiZ,DietzeR, Boom WH, Maciel E, and Hirsch CS. A role for CD4+CD25+ T cells in regulation of the immune response during human tuberculosis. Clin Exp Immunol. 2006 Apr;144(1):25–34.

228.Vanham G et al. Generalized immune activation in pulmonary tuberculosis: Co-activation with HIV infection. Clin Exp Immunol. 1996;103:30.

229.Fujiwara H and Tsuyuguchi I. Frequency of tuberculinreactive T-lymphocytes in pleural fluid and blood from patients with tuberculous pleuritis. Chest 1984;89:530.

230.Carlucci S, Beschin A, Tuosto L, Ameglio F, Gandolfo G, Cocito C, Fiorucci F, Saltini C, and Piccolella E. Mycobacterial antigen complex A60-specific T-cell repertoire during the course of pulmonary tuberculosis, Infect Immun. 1993;61:439.

231.Hirsch CS, Hussain R, Toossi Z, Dawood G, Shahid F, and Ellner JJ. Cross modulation by transforming growth factor β in human tuberculosis: Suppression of antigen-driven blastogenesis and interferon γ production. Proc Natl Acad Sci (USA). 1995;93:3193.

232.Rakotosamimanana N et al. Variation in gamma interferon responses to different infecting strains of Mycobacterium tuberculosis in acid-fast bacillus smear-positive patients and household contacts in Antananarivo, Madagascar. Clin Vaccine Immunol. 2010;17:1094–103.

233.Lee J-S et al. Profiles of IFN-γ and its regulatory cytokines (IL-12, IL-18 and IL-10) in peripheral blood mononuclear cells from patients with multidrug-resistant tuberculosis. Clin Exp Immunol. 2002;128(3):516–24.

234.Adekambi T, Ibegbu CC, Cagle S, Kalokhe, AS, Wang YF, Hu Y, Day CL, Ray SM, and Rengarajan J. Biomarkers on patient T cells diagnose active tuberculosis and monitor treatment response. J Clin Invest. 2015;125:1827.

235.Adekambi T, Ibegbu CC, Cagle S, Ray SM, and Rengarajan J. High Frequencies of caspase-3 expressing Mycobacterium tuberculosis-specific cD4+ T cells are associated with active tuberculosis. Front Immunol. 2018;25:1481.

236.Andrade-Arzabe R, Machado IV, Fernandez B, Blanca I, Ramirez R, and Bianco NE. Cellular immunity in current active pulmonary tuberculosis. Am Rev Respir Dis. 1991;143:496.

237.Shiratsuchi H, Okuda Y, and Tsuyuguchi I. Recombinant human interleukin-2 reverses in vitro deficient cell-medi- ated immune responses to tuberculin purified protein derivative by lymphocytes of tuberculous patients. Infect Immun. 1987;55:2126.

238.Huygen K, van Vooren JP, Turneer M, Bosmans R, Dierckx P, and De Bruyn J. Specific lymphoproliferation, gamma interferon production, and serum immunoglobulin G directed against a purified 32 kDA mycobacterial protein antigen (P32) in patients with active tuberculosis. Scand J Immunol. 1988;27:187.

239. Vilcek J, Klion A, Henriksen-DeStefano D, Zemtsov A, Davidson DM, Davidson M, and Friedman-Kien A. Defective gamma-interferon production in peripheral blood leukocytes of patients with acute tuberculosis. J Clin Immunol. 1986;6:146.

240.Gazzinelli RT, Hieny S, Wynn TA, Wolf S, and Sher A. Interleukin 12 is required for the T-lymphocyte-independent induction of interferon γ by an intracellular parasite and induces resistance in T-cell-deficient hosts. Proc Natl Acad Sci. 1993;90:6115.

241.Sieling PA, Abrams JS, Yamamura M, Salgame P, Bloom BR, Rea TH, and Modlin RL. Immunosuppressive roles for IL-10 and IL-4 in human infection. J Immunol. 1993;150:5501.

242.Street NE and Mossman TR. Functional diversity of T lymphocytes due to secretion of different cytokine patterns, FASB J. 1991;5:171.

243.Bogdan C, Vodovotz Y, and Nathan C. Macrophage deactivation by interleukin 10. J Exp Med. 1991;174:1549.

244.Lehn MW, Weisner WY, Engelhorn S, Gillis S, and Remold

HG. IL-4 inhibits H2O2 production and antileishmanial capacity of human cultured monocytes mediated by IFN gamma. J Immunol. 1989;143:3020.

245.Sucrel HM, Tory-Blomberg M, Paulie S, Anderson G, Moreno C, Pasvol G, and Ivanyi J. TH-1/TH-2 profiles in tuberculosis, based on the proliferation and cytokine response of blood lymphocytes to mycobacterial antigens. Immunology 1994;81:171.

246.Hirsch CS, Toossi Z, Hussain R, and Ellner JJ. Suppression of T-cell responses by TGF-β in tuberculosis. J Invest Med. 1995;43:365A.

247.Schmitt E, Meuret G, and Stix L. Monocyte recruitment in tuberculosis and sarcoidosis. Brit J Hematol. 1977;35:11.

248.Toossi Z, Hamilton BD, Phillips MH, Averill LE, Ellner JJ, and Salvekar A. Regulation of nuclear factor-kB and its inhibitor IkB-a/ MAD-3 in monocytes by Mycobacterium tuberculosis and during human tuberculosis. J Immunol. 1997;159:4109.

249.Ahmed A, Adiga V, Nayak S, Uday Kumar JAJ, Dhar C, Sahoo PN, Sundararaj BN, Souza GD, and Vyakarnam A. Circulating HLA-DR+CD4+ effector memory T cells resistant to CCR5 and PD-L1 mediated suppression compromise regulatory T cell function in tuberculosis. PLoS Pathog. 2018;14(9):e1007289. https://doi.org/10.1371/journal.ppat.1007289

250.Lenzini L, Heather CJ, Rottoli L, and Rottoli P. Studies on bronchoalveolar cells in humans. I. Preliminary morphological studies in various respiratory diseases. Respiration 1978;36:145.

74  Pathogenesis of Tuberculosis

\251.\ Venet A, Niaudet P, Bach JF, and Even P. Study of alveolar lymphocytes obtained by bronchoalveolar lavage. Ann Anesthesiol Fr. 1980;6:634.

\252.\ Sharma SK, Pande JN, and Verma K. Bronchoalveolar lavage (BAL) in miliary tuberculosis. Tubercle 1988;69:175.

\253.\ Dhank R, De A, Ganguly NK, Gupta N, Jaswal S, Malik SK, and Kohli KK. Factors influencing the cellular response in bronchoalveolar lavage and peripheral blood of patients with pulmonary tuberculosis. Tubercle 1988;69:161.

\254.\ Baughman RP, Dohn MN, Loudon RG, and Trame PT. Bronchoscopy with bronchoalveolar lavage in tuberculosis and fungal infections. Chest 1991;99:92.

\255.\ Ozaki T, Nakahira S, Tani K, Ogushi F, Yasuoka S, and Ogura T. Differential cell analysis in bronchoalveolar lavage fluid from pulmonary lesions of patients with tuberculosis. Chest 1992;102:54.

\256.\ Schwander SK, Sada E, Torres M, Escobedo D, Sierra JG, Alt S, and Rich EA. T lymphocytic and immature macrophage alveolitis in active pulmonary tuberculosis. J Infect Dis. 1996;176:1267.

\257.\ Schwander SK, Torres M, Sada E, Carranza C, Ramos E, Tary-Lehmann M, Wallis RS, Sierra J, and Rich EA. Enhanced responses to Mycobacterium tuberculosis antigens by human alveolar lymphocytes during active pulmonary tuberculosis. J Infect Dis. 1998;178:1434.

\258.\ Taha RA, Kotsimbos TC, Song, Y-L., Menzies D, and Hamid Q. IFN-γ and IL-12 are increased in active compared with inactive tuberculosis. Am J Respir Crit Care Med. 1997;155:1135.

\259.\ Berry MPR et al. An interferon-inducible, neutrophildriven, blood transcriptional signature in tuberculosis. Nature. 2010;466:973.

\260.\ Cliff JM, Kaufmann SHE, McShane H, and van Helden P. The human immune response to tuberculosis and its treatment; a view from the blood. Immunol Rev. 2015;264:88.

\261.\ Moreira-Teixeira T, Mayer-Barber K, Sher A, and O’Garra A. Type 1 interferons in tuberculosis: Foe and occasionally friend. J Exp Med. 2018.

\262.\ Reider HL, Cauthen GM, Bloch AB, Cole CH, Holtzman D, Snider DE, Bigler WJ, and Witte JJ. Tuberculosis and AIDS: Florida. Arch Intern Med. 1989;149:1268.

\263.\ Daley CL, Small GF, Schecter GK, Schoolnik GK, McAdam RA, Jacobs WR, and Hopewell PC. An outbreak of tuberculosis with accelerated progression among persons infected with HIV. N Engl J Med. 1992;326:2131.

\264.\ Lucas S and Nelson AM. Pathogenesis of tuberculosis in human immunodeficiency virus-infected people. In: Bloom B. (ed.). Tuberculosis Pathogenesis, Protection, and Control. ASM Press: Materials Park, OH, 1994.

\265.\ Ellner JJ. [Editorial] Tuberculosis in the time of AIDS: The facts and the message. Chest 1990;98:1051.

\ 266.\ OkweraA,ErikiPP,GuayLA,BallP,andDanielTM.Tuberculin reactions in HIV-seropositive and HIV-seronegative healthy women in Uganda. MMWR. 1990;39:638.

\267.\ Johnson JL et al. Impact of human immunodeficiency virus type 1 infection on the initial bacteriologic and radiographic manifestations of pulmonary tuberculosis

in Uganda (Makerere University-Case Western Reserve Research Collaboration). Int J Tuberc Lung Dis. 1998;2:397.

\268.\ Wallis RS, Vjecha M, Amir-Tahmasseb M, Okwera A, Byekwaso F, Nyole J, Kabengera J, Mugerwa RD, and Ellner JJ. Influence of tuberculosis on HIV: Enhanced cytokine expression and elevated B2 microglobulin in HIV-1 associated tuberculosis. J Infect Dis. 1992;167:43.

\269.\ Kaforou M et al. Detection of tuberculosis in HIV-infected and -uninfected African adults using whole blood RNA expression signatures: A case-control study. PLoS Med. 2013;10(10):e1001538.

\270.\ Dawany N, Showe LC, Kossenkov AV, Chang C, Ive P, Conradie F, Stevens W, Sanne I, Azzoni L, and Montaner LJ. Identification of a 251 gene expression signature that can accurately detect M. tuberculosis in patients with and without HIV co-infection. PLOS ONE 2014;9(2):e89925.

\271.\ Maertzdorf J, McEwen G, Weiner J 3rd, Tian S, Lader E, Schriek U, Mayanja-Kizza H, Ota M, Kenneth J, and Kaufmann SH. Concise gene signature for point- of-care classification of tuberculosis. EMBO Mol Med. 2016;8(2):86–95.

\272.\ Laux da Costa L, Delcroix M, Dalla Costa ER, Prestes IV, Milano M, Francis SS, Unis G, Silva DR, Riley LW, and Rossetti ML. A real-time PCR signature to discriminate between tuberculosis and other pulmonary diseases.

Tuberculosis (Edinburgh) 2015;95(4):421–5.

\273.\ Sweeney TE, Braviak L, Tato CM, and Khatri P. Genomewide expression for diagnosis of pulmonary tuberculosis: A multicohort analysis. Lancet Respir Med. 2016;4(3):213–24.

\274.\ Sutherland JS et al. Differential gene expression of activating Fcgamma receptor classifies active tuberculosis regardless of human immunodeficiency virus status or ethnicity. Clin Microbiol Infect. 2014;20(4):O230–8.

\275.\ Roe JK et al. Blood transcriptomic diagnosis of pulmonary and extrapulmonary tuberculosis. JCI Insight 2016;1(16):e87238.

\276.\ Verma S et al. Tuberculosis in advanced HIV infection is associated with increased expression of IFNγ and its downstream targets. BMC Infect Dis. 2018;18(1):220. Published 2018 May 15. doi:10.1186/s12879-018-3127-4

\277.\ Lai RP, Nakiwala JK, Meintjes G, and Wilkinson RJ. The immunopathogenesis of the HIV tuberculosis immune reconstitution inflammatory syndrome. Eur J Immunol 2013;43:1995–2002.

\278.\ Meintjes G et al. Type 1 helper T cells and FoxP3-positive T cells in HIV–tuberculosis-associated immune reconstitution inflammatory syndrome. Am J Respir Crit Care Med. 2008;178(10):1083–9.

\279.\ Conradie F et al. Natural killer cell activation distinguishes Mycobacterium tuberculosis-mediated immune reconstitution syndrome from chronic HIV and HIV/MTB coinfection. J Acquired Immune Defic Syndr. 2011;58(3):309–18.

\280.\ Jinmin Ma J et al. Zinc finger and interferon-stimulated genes play a vital role in TB-IRIS following HAART in AIDS. Per Med. 2018;15(4):251–69.

\281.\ Tran HTT et al. Geert for the TB-IRIS study group. Modulation of the complement system in monocytes

Книга в списке рекомендаций к покупке и прочтению сайта https://meduniver.com/

References  75

contributes to tuberculosis-associated immune reconstitution inflammatory syndrome. AIDS July 17th, 2013;​ 27(11):1725–34.

282.Tran HTT, Van der Bergh R, Vu TN, Laukens K, Worodria W, Loembe MM, Colebunders R, Kestens L, De Baetselier P, Raes G for the TB-IRIS Study Group. The role of monocytes in the development of tuberculosis-associated immune reconstitution inflammatory syndrome. Immunobiology January 2014;219(1):37–44.

283.Narendran G et al. Paradoxical tuberculosis immune reconstitution inflammatory syndrome (TB-IRIS) in HIV patients with culture confirmed pulmonary tuberculosis in India and the potential role of IL-6 in prediction. PLOS ONE 2013;8(5):e63541. https://doi.org/10.1371/journal. pone.0063541

284.Meintjes G et al. Randomized placebo-controlled trial of prednisone for paradoxical tuberculosis-associated immune reconstitution inflammatory syndrome. AIDS 2010;24(15):2381–90.

285.Meintjes G et al. PredART Trial Team. Prednisone for the

prevention of paradoxical tuberculosis-associated IRIS. N Engl J Med. 2018 Nov 15;379(20):1915–25. doi: 10.1056/ NEJMoa1800762.

286.Cohen OJ, Kinter A, and Fauci AS. Host factors in the pathogenesis of HIV disease, Immunol Rev. 1997;159:31.

287.Lawn SD, Butera ST, and Shinnick TM. Tuberculosis unleashed: The impact of human immunodeficiency virus infection on the host granulomatous response to

Mycobacterium tuberculosis. Microbes Infect 2002;4:635e46.

288.Clerici M, and Shearer GM. A TH-1/TH-2 switch is a critical step in the etiology of HIV infection. Immunol Today 1993;14:107.

289.Zhang M, Gong J, Iyer D, Jones BE, Modlin RL, and Barnes PF. T-cell cytokine responses in persons with tuberculosis and human immunodeficiency virus infection. J Clin Invest. 1994;94:2435.

290.Orenstein JM, Fox C, and Wahl MS. Macrophages as a source of HIV during opportunistic infections, Science 1997;276:1857.

291.Meltzer MS, Skillman DR, Gomatos PJ, Kalter DC, and Gendelman HE. Role of mononuclear phagocytes in the pathogenesis of human immunodeficiency virus infection.

Ann Rev Immunol. 1990;8:169.

292.Twigg HL, Lipscomb MF, Yoffe B, Barbaro DJ, and Weissler JC. Enhanced accessory cell function by alveolar macrophages from patients infected with the human immunodeficiency virus: Potential role for depletion of CD4+ cells in the lung. Am J Respir Cell Mol Biol. 1989;1:391.

293.Murray HW, Gellene RA, Libby DM, Roth E, Armmel CD, and Rubin BY. Activation of tissue macrophages from AIDS patients: in vitro responses of AIDS alveolar macrophages to lymphokines and interferon-γ. J Immunol. 1985;​ 135:2374.

294.Buhl R, Jaffe HA, Holroyd KJ, Borok Z, Roum JH, Mastrangeli A, Wells FB, Kirby M, Saltini C, and Crystal RG. Activation of alveolar macrophages in asymptomatic HIV-infected individuals. J Immunol. 1993;150:1019.

295.Twigg HL, Iwamoto GK, and Soliman DM. Role of cytokines in alveolar macrophage accessory cell function in HIV-infected individuals. J Immunol. 1992;149:1462.

296.Trentin L, Barbisa S, Zambello R, Agostini C, Caenazzo C, di Francesco C, Cipriani A, Francavalla E, and Semenzato

G.Spontaneous production of IL-6 by alveolar macrophages form human immunodeficiency virus type 1-infected patients. J Infect Dis. 1992;166:731.

297.Agostini C et al. Alveolar macrophages from patients with AIDS and AIDS-related complex constitutively synthesize and release tumor necrosis factor alpha. Am Rev Respir Dis. 1991;144:195.

298.Plata F, Autran B, Pedroza Martins L, Wain-Hobson S, Raphael M, Mayaud C, Denis M, Guillon JM, and Debre P. AIDS virus-specific cytotoxic T-lymphocytes in lung disorders. Nature (London) 1987;328:348.

299.De Cock KM, Soro B, Coulibaly IM, and Lucas SB. Tuberculosis and HIV infection in sub-Saharan Africa. JAMA 1992;268:1581.

300.Whalen C, Horsburgh CR, Hom D, Lahart C, Simberkoff M, and Ellner JJ. Accelerated course of human immunodeficiency virus infection after tuberculosis. Am J Respir Crit Care Med. 1995;151:129.

301.Braun MM, Nsanga B, and Ryder RW. A retrospective cohort study of the risks of tuberculosis among women of childbearing age with HIV infection in Zaire. Am Rev Respir Dis. 1991;143:501.

302.Vjecha M et al. Predictors of mortality and drug toxicity in HIV-infected patients from Uganda treated for pulmonary tuberculosis, 8th International Conference on AIDS/3rd STD World Congress, Amsterdam, Netherlands, July 1992.

303.Goletti D, Weissman D, Jackson RW, Graham NM, Vlahov D, Klein RS, Munsiff SS, Ortona L, Cauda R, and Fauci AS. Effect of Mycobacterium tuberculosis on HIV replication: Role of immune activation. J Immunol. 1996;157:1271.

304.Toossi Z. Virological and immunological impact of tuberculosis on human immunodeficiency virus type 1 disease.

JInfect Dis. 15 October 2003;188(8):1146–55, https://doi. org/10.1086/378676

305.Lederman MM, Georges DL, Kusner DJ, Mudido P, Giam, C-Z, and Toossi Z. Mycobacterium tuberculosis and its purified protein derivative activate expression of the human immunodeficiency virus. J Acquired Immune Defic Syndr. 1994;7:727.

306.Toossi Z, Nicolacakis K, Xia L, Ferrari NA, and Rich EA. Activation of latent HIV-1 by Mycobacterium tuberculosis and its purified protein derivative in alveolar macrophages from HIV infected individuals in vitro. J Acquired Immune Defic Syndr Hum Retrovirol. 1997;15:325.

307.Toossi Z, Sierra-Madero JG, Blinkhorn RA, Mettler MA, and Rich EA. Enhanced susceptibility of blood monocytes from patients with pulmonary tuberculosis to productive infection with human immunodeficiency virus-1 (HIV-1).

JExp Med. 1993;177:1511.

308.Garrait CJ, Esvant H, Henry I, Morinet P, Mayaud C, and Israel-Biet D. Tuberculosis generates a microenvironment enhancing the productive infection of local lymphocytes by HIV. J Immunol. 1997;159:2824.

76  Pathogenesis of Tuberculosis

\309.\ Olson A, Ragan EJ, Nakiyingi L, Lin N, Jacobson KR, Ellner JJ, Manabe YC, and Sagar M. Pulmonary tuberculosis is associated with persistent systemic inflammation and decreased HIV-1 reservoir markers in co-infected Ugandans. J Acquired Immune Defic Syndr. 2018 Jul 26. doi: 10.1097/QAI.0000000000001823. [Epub ahead of print]

\310.\ Murugesan VS, Bin N, Dodd CE, and Schesinger LS. Macrophage immunoregulatory pathways in tuberculosis. Semin Immunol. 2014;26:471.

\311.\ Rich EA, Torres M, Sada E, Finegan CK, Hamilton BD, and Toossi Z. Mycobacterium tuberculosis (MTB)-stimulated production of nitric oxide by human alveolar macrophages and relationship of nitric oxide production to growth inhibition of MTB. Tuberc Lung Dis 1997;78:247.

\312.\ Hussain R, Dawood G, Obaid M, Toossi Z, Wallis RS, Minai A, Dojki M, Sturm AW, and Ellner JJ. Depressed cellular and augmented humoral responses in patients with active tuberculosis from Pakistan. Clin Diag Lab Immunol. 1995;2:726.

Книга в списке рекомендаций к покупке и прочтению сайта https://meduniver.com/

PART III

TRANSMISSION

5

Using Genotyping and Molecular Surveillance to Investigate Tuberculosis Transmission

79

 

Sarah Talarico, Laura F. Anderson, and Benjamin J. Silk

 

6

Tuberculosis Transmission Control

97

 

Edward A. Nardell

 

Книга в списке рекомендаций к покупке и прочтению сайта https://meduniver.com/

5

Using Genotyping and Molecular Surveillance to Investigate Tuberculosis Transmission

SARAH TALARICO, LAURA F. ANDERSON, AND BENJAMIN J. SILK

Introduction

Genotyping methods

Recent transmission

Clinical and public health applications

References

INTRODUCTION

Tuberculosis (TB) control programs prevent transmission of Mycobacterium tuberculosis by identifying and treating active cases of TB disease and ensuring that contacts of infectious cases are also identified, screened, and treated for active disease or latent infection. Genotyping and molecular surveillance methods can identify clusters of cases that share genetically similar M. tuberculosis strains, which may be related by recent transmission. Public health tools to investigate TB transmission have advanced by combining technologies for DNA sequencing and phylogenetic analysis with clinical and epidemiological information. Outbreak investigations are a priority because they can establish epidemiological links among patients and direct prevention measures toward populations and settings with ongoing transmission. The chapter reviews the use of genotyping to investigate transmission as well as how genotyping methods have been applied to study transmission dynamics, lineage, recurrent TB disease, mixed infections, and laboratory contamination.

GENOTYPING METHODS

Genotyping methods for M. tuberculosis can be divided into two main categories: (1) conventional genotyping methods that examine variation affecting a small portion of the M. tuberculosis genome and (2) whole-genome sequencing (WGS), which examines most of the M. tuberculosis genome at the nucleotide level. The three most commonly used conventional genotyping methods are restriction fragment length polymorphism (RFLP), spacer oligonucleotide typing (spoligotyping), and mycobacterial interspersed repetitive units-variable number tandem repeats (MIRU-VNTR). These methods are all based on repetitive DNA sequences found either interspersed throughout the bacterial

79

79

82

86

90

genome (insertion sequences) or at specific loci. Changes in these repetitive DNA sequences serve as a proxy for genetic diversification that is occurring throughout the entire genome. However, as DNA sequencing technologies have advanced, WGS is replacing the use of conventional genotyping methods for public health purposes to provide direct analysis of phylogenetic relationships at the nucleotide level.

Conventional methods based on repetitive sequences

RFLP typing for M. tuberculosis is based on the 1.3 kilobase repetitive insertion sequence IS6110, which is unique to M. tuberculosis complex (MTBC).1 IS6110 varies in the number of copies from 0 to 25 per strain and also by insertion site in the genome.2 These variations produce highly diverse hybridization patterns that are used to discriminate between strains (Figure 5.1). In the 1990s, IS6110 was the gold standard for genotyping M. tuberculosis due to the high discriminatory power and the stability of the biomarker, which has a half-life of approximately 3.2 years (i.e., the time it takes for a hybridization pattern to change).3 Disadvantages of IS6110-RFLP are that the technique itself is time-consuming, technically demanding, lacks inter-laboratory reproducibility, and requires specialized software for the analysis of results, which are difficult to compare between different laboratories and over time. Furthermore, the discriminatory power is poor for strains without a copy of IS6110 and for strains with a low (<6) copy number, such as M. bovis.

Spoligotyping is based on the presence or absence of 43 unique DNA “spacer” sequences that are interspersed between 36 base pair sequences.4,5 The results consist of binary data and the number and position of absent spacer sequences can be compared between strains (Figure 5.2). Results are reproducible, easy to interpret,

79