Skip to main content
Top
Published in: BMC Medicine 1/2016

Open Access 01-12-2016 | Opinion

‘”Why me, why now?” Using clinical immunology and epidemiology to explain who gets nontuberculous mycobacterial infection

Authors: M. Alexandra Lake, Lyn R. Ambrose, Marc C. I. Lipman, David M. Lowe

Published in: BMC Medicine | Issue 1/2016

Login to get access

Abstract

Background

The prevalence of nontuberculous mycobacterial (NTM) disease is rising. An understanding of known risk factors for disease sheds light on the immunological and physical barriers to infection, and how and why they may be overcome. This review focuses on human NTM infection, supported by experimental and in vitro data of relevance to the practising clinician who seeks to understand why their patient has NTM infection and how to further investigate.

Discussion

First, the underlying immune response to NTM disease is examined. Important insights regarding NTM disease susceptibility come from nature's own knockouts, the primary immune deficiency disorders. We summarise the current knowledge surrounding interferon-gamma (IFNγ)-interleukin-12 (IL-12) axis abnormalities, followed by a review of phagocytic defects, T cell lymphopenia and rarer genetic conditions known to predispose to NTM disease. We discuss how these define key immune pathways involved in the host response to NTM. Iatrogenic immunosuppression is also important, and we evaluate the impact of novel biological therapies, as well as bone marrow transplant and chemotherapy for solid organ malignancy, on the epidemiology and presentation of NTM disease, and discuss the host defence dynamics thus revealed. NTM infection and disease in the context of other chronic illnesses including HIV and malnutrition is reviewed. The role of physical barriers to infection is explored. We describe how their compromise through different mechanisms including cystic fibrosis, bronchiectasis and smoking-related lung disease can result in pulmonary NTM colonisation or infection. We also summarise further associations with host factors including body habitus and age.

Summary

We use the presented data to develop an over-arching model that describes human host defences against NTM infection, where they may fail, and how this framework can be applied to investigation in routine clinical practice.
Literature
1.
go back to reference Chetchotisakd P, Kiertiburanakul S, Mootsikapun P, Assanasen S, Chaiwarith R, Anunnatsiri S. Disseminated nontuberculous mycobacterial infection in patients who are not infected with HIV in Thailand. Clin Infect Dis. 2007;45:421–7.PubMedCrossRef Chetchotisakd P, Kiertiburanakul S, Mootsikapun P, Assanasen S, Chaiwarith R, Anunnatsiri S. Disseminated nontuberculous mycobacterial infection in patients who are not infected with HIV in Thailand. Clin Infect Dis. 2007;45:421–7.PubMedCrossRef
2.
go back to reference Prevots DR, Shaw PA, Strickland D, Jackson LA, Raebel MA, Blosky MA, et al. Nontuberculous mycobacterial lung disease prevalence at four integrated health care delivery systems. Am J Respir Crit Care Med. 2010;182:970–6.PubMedPubMedCentralCrossRef Prevots DR, Shaw PA, Strickland D, Jackson LA, Raebel MA, Blosky MA, et al. Nontuberculous mycobacterial lung disease prevalence at four integrated health care delivery systems. Am J Respir Crit Care Med. 2010;182:970–6.PubMedPubMedCentralCrossRef
3.
go back to reference Adjemian J, Olivier KN, Seitz AE, Holland SM, Prevots DR. Prevalence of nontuberculous mycobacterial lung disease in U.S. Medicare beneficiaries. Am J Respir Crit Care Med. 2012;185:881–6.PubMedPubMedCentralCrossRef Adjemian J, Olivier KN, Seitz AE, Holland SM, Prevots DR. Prevalence of nontuberculous mycobacterial lung disease in U.S. Medicare beneficiaries. Am J Respir Crit Care Med. 2012;185:881–6.PubMedPubMedCentralCrossRef
4.
go back to reference Shah N, Davidson J, Anderson L, Lalor M, Kim J, Thomas H, et al. BMC Infectious Diseases Pulmonary Mycobacterium avium-intracellulare is the main driver of the rise in non-tuberculous mycobacteria incidence in England, Wales and Northern Ireland, 2007–2012. BMC Infect Dis. 2016. In Press. Shah N, Davidson J, Anderson L, Lalor M, Kim J, Thomas H, et al. BMC Infectious Diseases Pulmonary Mycobacterium avium-intracellulare is the main driver of the rise in non-tuberculous mycobacteria incidence in England, Wales and Northern Ireland, 2007–2012. BMC Infect Dis. 2016. In Press.
5.
go back to reference Griffith DE, Aksamit T, Brown-Elliott BA, Catanzaro A, Daley C, Gordin F, et al. An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med. 2007;175:367–416.PubMedCrossRef Griffith DE, Aksamit T, Brown-Elliott BA, Catanzaro A, Daley C, Gordin F, et al. An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med. 2007;175:367–416.PubMedCrossRef
6.
go back to reference Satta G, McHugh TD, Mountford J, Abubakar I, Lipman M. Managing pulmonary nontuberculous mycobacterial infection: Time for a patient-centered approach. Ann Am Thorac Soc. 2014;11:117–21.PubMedPubMedCentralCrossRef Satta G, McHugh TD, Mountford J, Abubakar I, Lipman M. Managing pulmonary nontuberculous mycobacterial infection: Time for a patient-centered approach. Ann Am Thorac Soc. 2014;11:117–21.PubMedPubMedCentralCrossRef
7.
go back to reference Casanova JL, Blanche S, Emile JF, Jouanguy E, Lamhamedi S, Altare F, et al. Idiopathic disseminated bacillus Calmette-Guérin infection: a French national retrospective study. Pediatrics. 1996;98(4 Pt 1):774–8.PubMed Casanova JL, Blanche S, Emile JF, Jouanguy E, Lamhamedi S, Altare F, et al. Idiopathic disseminated bacillus Calmette-Guérin infection: a French national retrospective study. Pediatrics. 1996;98(4 Pt 1):774–8.PubMed
8.
go back to reference Prando C, Samarina A, Bustamante J, Boisson-Dupuis S, Cobat A, Picard C, et al. Inherited IL-12p40 deficiency: genetic, immunologic, and clinical features of 49 patients from 30 kindreds. Medicine (Baltimore). 2013;92:109–22.CrossRef Prando C, Samarina A, Bustamante J, Boisson-Dupuis S, Cobat A, Picard C, et al. Inherited IL-12p40 deficiency: genetic, immunologic, and clinical features of 49 patients from 30 kindreds. Medicine (Baltimore). 2013;92:109–22.CrossRef
9.
go back to reference Picard C, Fieschi C, Altare F, Al-Jumaah S, Al-Hajjar S, Feinberg J, et al. Inherited interleukin-12 deficiency: IL12B genotype and clinical phenotype of 13 patients from six kindreds. Am J Hum Genet. 2002;70:336–48.PubMedPubMedCentralCrossRef Picard C, Fieschi C, Altare F, Al-Jumaah S, Al-Hajjar S, Feinberg J, et al. Inherited interleukin-12 deficiency: IL12B genotype and clinical phenotype of 13 patients from six kindreds. Am J Hum Genet. 2002;70:336–48.PubMedPubMedCentralCrossRef
10.
go back to reference Fieschi C, Dupuis S, Catherinot E, Feinberg J, Bustamante J, Breiman A, et al. Low penetrance, broad resistance, and favorable outcome of interleukin 12 receptor beta1 deficiency: medical and immunological implications. J Exp Med. 2003;197:527–35.PubMedPubMedCentralCrossRef Fieschi C, Dupuis S, Catherinot E, Feinberg J, Bustamante J, Breiman A, et al. Low penetrance, broad resistance, and favorable outcome of interleukin 12 receptor beta1 deficiency: medical and immunological implications. J Exp Med. 2003;197:527–35.PubMedPubMedCentralCrossRef
11.
go back to reference Newport MJ, Huxley CM, Huston S, Hawrylowicz CM, Oostra BA, Williamson R, et al. A mutation in the interferon-gamma-receptor gene and susceptibility to mycobacterial infection. N Engl J Med. 1996;335:1941–9.PubMedCrossRef Newport MJ, Huxley CM, Huston S, Hawrylowicz CM, Oostra BA, Williamson R, et al. A mutation in the interferon-gamma-receptor gene and susceptibility to mycobacterial infection. N Engl J Med. 1996;335:1941–9.PubMedCrossRef
12.
go back to reference Jouanguy E, Dupuis S, Pallier A, Döffinger R, Fondanèche MC, Fieschi C, et al. In a novel form of IFN-gamma receptor 1 deficiency, cell surface receptors fail to bind IFN-gamma. J Clin Invest. 2000;105:1429–36.PubMedPubMedCentralCrossRef Jouanguy E, Dupuis S, Pallier A, Döffinger R, Fondanèche MC, Fieschi C, et al. In a novel form of IFN-gamma receptor 1 deficiency, cell surface receptors fail to bind IFN-gamma. J Clin Invest. 2000;105:1429–36.PubMedPubMedCentralCrossRef
13.
go back to reference Holland SM, Dorman SE, Kwon A, Pitha-Rowe IF, Frucht DM, Gerstberger SM, et al. Abnormal regulation of interferon-gamma, interleukin-12, and tumor necrosis factor-alpha in human interferon-gamma receptor 1 deficiency. J Infect Dis. 1998;178:1095–104.PubMedCrossRef Holland SM, Dorman SE, Kwon A, Pitha-Rowe IF, Frucht DM, Gerstberger SM, et al. Abnormal regulation of interferon-gamma, interleukin-12, and tumor necrosis factor-alpha in human interferon-gamma receptor 1 deficiency. J Infect Dis. 1998;178:1095–104.PubMedCrossRef
14.
go back to reference Dorman SE, Holland SM. Mutation in the signal-transducing chain of the interferon-gamma receptor and susceptibility to mycobacterial infection. J Clin Invest. 1998;101:2364–9.PubMedPubMedCentralCrossRef Dorman SE, Holland SM. Mutation in the signal-transducing chain of the interferon-gamma receptor and susceptibility to mycobacterial infection. J Clin Invest. 1998;101:2364–9.PubMedPubMedCentralCrossRef
15.
go back to reference Dupuis S, Dargemont C, Fieschi C, Thomassin N, Rosenzweig S, Harris J, et al. Impairment of mycobacterial but not viral immunity by a germline human STAT1 mutation. Science. 2001;293:300–3.PubMedCrossRef Dupuis S, Dargemont C, Fieschi C, Thomassin N, Rosenzweig S, Harris J, et al. Impairment of mycobacterial but not viral immunity by a germline human STAT1 mutation. Science. 2001;293:300–3.PubMedCrossRef
16.
go back to reference Chapgier A, Kong X-F, Boisson-Dupuis S, Jouanguy E, Averbuch D, Feinberg J, et al. A partial form of recessive STAT1 deficiency in humans. J Clin Invest. 2009;119:1502–14.PubMedPubMedCentralCrossRef Chapgier A, Kong X-F, Boisson-Dupuis S, Jouanguy E, Averbuch D, Feinberg J, et al. A partial form of recessive STAT1 deficiency in humans. J Clin Invest. 2009;119:1502–14.PubMedPubMedCentralCrossRef
17.
go back to reference Sampaio EP, Bax HI, Hsu AP, Kristosturyan E, Pechacek J, Chandrasekaran P, et al. A novel STAT1 mutation associated with disseminated mycobacterial disease. J Clin Immunol. 2012;32:681–9.PubMedPubMedCentralCrossRef Sampaio EP, Bax HI, Hsu AP, Kristosturyan E, Pechacek J, Chandrasekaran P, et al. A novel STAT1 mutation associated with disseminated mycobacterial disease. J Clin Immunol. 2012;32:681–9.PubMedPubMedCentralCrossRef
18.
go back to reference Averbuch D, Chapgier A, Boisson-Dupuis S, Casanova J-L, Engelhard D. The clinical spectrum of patients with deficiency of Signal Transducer and Activator of Transcription-1. Pediatr Infect Dis J. 2011;30:352–5.PubMedCrossRef Averbuch D, Chapgier A, Boisson-Dupuis S, Casanova J-L, Engelhard D. The clinical spectrum of patients with deficiency of Signal Transducer and Activator of Transcription-1. Pediatr Infect Dis J. 2011;30:352–5.PubMedCrossRef
19.
go back to reference Chapgier A, Boisson-Dupuis S, Jouanguy E, Vogt G, Feinberg J, Prochnicka-Chalufour A, et al. Novel STAT1 alleles in otherwise healthy patients with mycobacterial disease. PLoS Genet. 2006;2, e131.PubMedPubMedCentralCrossRef Chapgier A, Boisson-Dupuis S, Jouanguy E, Vogt G, Feinberg J, Prochnicka-Chalufour A, et al. Novel STAT1 alleles in otherwise healthy patients with mycobacterial disease. PLoS Genet. 2006;2, e131.PubMedPubMedCentralCrossRef
20.
go back to reference Kreins AY, Ciancanelli MJ, Okada S, Kong X-F, Ramírez-Alejo N, Kilic SS, et al. Human TYK2 deficiency: Mycobacterial and viral infections without hyper-IgE syndrome. J Exp Med. 2015;212:1641–62.PubMedPubMedCentralCrossRef Kreins AY, Ciancanelli MJ, Okada S, Kong X-F, Ramírez-Alejo N, Kilic SS, et al. Human TYK2 deficiency: Mycobacterial and viral infections without hyper-IgE syndrome. J Exp Med. 2015;212:1641–62.PubMedPubMedCentralCrossRef
21.
go back to reference Hambleton S, Salem S, Bustamante J, Bigley V, Boisson-Dupuis S, Azevedo J, et al. IRF8 mutations and human dendritic-cell immunodeficiency. N Engl J Med. 2011;365:127–38.PubMedPubMedCentralCrossRef Hambleton S, Salem S, Bustamante J, Bigley V, Boisson-Dupuis S, Azevedo J, et al. IRF8 mutations and human dendritic-cell immunodeficiency. N Engl J Med. 2011;365:127–38.PubMedPubMedCentralCrossRef
22.
go back to reference Bogunovic D, Byun M, Durfee LA, Abhyankar A, Sanal O, Mansouri D, et al. Mycobacterial disease and impaired IFN-γ immunity in humans with inherited ISG15 deficiency. Science. 2012;337:1684–8.PubMedPubMedCentralCrossRef Bogunovic D, Byun M, Durfee LA, Abhyankar A, Sanal O, Mansouri D, et al. Mycobacterial disease and impaired IFN-γ immunity in humans with inherited ISG15 deficiency. Science. 2012;337:1684–8.PubMedPubMedCentralCrossRef
23.
go back to reference Takeda K, Kawai T, Nakazawa Y, Komuro H, Shoji K, Morita K, et al. Augmentation of antitubercular therapy with IFNγ in a patient with dominant partial IFNγ receptor 1 deficiency. Clin Immunol. 2014;151:25–8.PubMedCrossRef Takeda K, Kawai T, Nakazawa Y, Komuro H, Shoji K, Morita K, et al. Augmentation of antitubercular therapy with IFNγ in a patient with dominant partial IFNγ receptor 1 deficiency. Clin Immunol. 2014;151:25–8.PubMedCrossRef
24.
go back to reference Hallstrand TS, Ochs HD, Zhu Q, Liles WC. Inhaled IFN-gamma for persistent nontuberculous mycobacterial pulmonary disease due to functional IFN-gamma deficiency. Eur Respir J. 2004;24:367–70.PubMedCrossRef Hallstrand TS, Ochs HD, Zhu Q, Liles WC. Inhaled IFN-gamma for persistent nontuberculous mycobacterial pulmonary disease due to functional IFN-gamma deficiency. Eur Respir J. 2004;24:367–70.PubMedCrossRef
25.
go back to reference Bax HI, Freeman AF, Ding L, Hsu AP, Marciano B, Kristosturyan E, et al. Interferon alpha treatment of patients with impaired interferon gamma signaling. J Clin Immunol. 2013;33:991–1001.PubMedPubMedCentralCrossRef Bax HI, Freeman AF, Ding L, Hsu AP, Marciano B, Kristosturyan E, et al. Interferon alpha treatment of patients with impaired interferon gamma signaling. J Clin Immunol. 2013;33:991–1001.PubMedPubMedCentralCrossRef
26.
go back to reference Ward CM, Jyonouchi H, Kotenko SV, Smirnov SV, Patel R, Aguila H, et al. Adjunctive treatment of disseminated Mycobacterium avium complex infection with interferon alpha-2b in a patient with complete interferon-gamma receptor R1 deficiency. Eur J Pediatr. 2007;166:981–5.PubMedCrossRef Ward CM, Jyonouchi H, Kotenko SV, Smirnov SV, Patel R, Aguila H, et al. Adjunctive treatment of disseminated Mycobacterium avium complex infection with interferon alpha-2b in a patient with complete interferon-gamma receptor R1 deficiency. Eur J Pediatr. 2007;166:981–5.PubMedCrossRef
27.
go back to reference Okada S, Markle JG, Deenick EK, Mele F, Averbuch D, Lagos M, et al. Impairment of immunity to Candida and Mycobacterium in humans with bi-allelic RORC mutations. Science (80-). 2015;349:606–13.CrossRef Okada S, Markle JG, Deenick EK, Mele F, Averbuch D, Lagos M, et al. Impairment of immunity to Candida and Mycobacterium in humans with bi-allelic RORC mutations. Science (80-). 2015;349:606–13.CrossRef
28.
go back to reference Nedorost ST, Elewski B, Tomford JW, Camisa C. Rosacea-like lesions due to familial Mycobacterium avium-intracellulare infection. Int J Dermatol. 1991;30:491–7.PubMedCrossRef Nedorost ST, Elewski B, Tomford JW, Camisa C. Rosacea-like lesions due to familial Mycobacterium avium-intracellulare infection. Int J Dermatol. 1991;30:491–7.PubMedCrossRef
29.
go back to reference Filipe-Santos O, Bustamante J, Haverkamp MH, Vinolo E, Ku C-L, Puel A, et al. X-linked susceptibility to mycobacteria is caused by mutations in NEMO impairing CD40-dependent IL-12 production. J Exp Med. 2006;203:1745–59.PubMedPubMedCentralCrossRef Filipe-Santos O, Bustamante J, Haverkamp MH, Vinolo E, Ku C-L, Puel A, et al. X-linked susceptibility to mycobacteria is caused by mutations in NEMO impairing CD40-dependent IL-12 production. J Exp Med. 2006;203:1745–59.PubMedPubMedCentralCrossRef
30.
go back to reference Deffert C, Cachat J, Krause K-H. Phagocyte NADPH oxidase, chronic granulomatous disease and mycobacterial infections. Cell Microbiol. 2014;16:1168–78.PubMedCrossRef Deffert C, Cachat J, Krause K-H. Phagocyte NADPH oxidase, chronic granulomatous disease and mycobacterial infections. Cell Microbiol. 2014;16:1168–78.PubMedCrossRef
31.
go back to reference Bustamante J, Arias AA, Vogt G, Picard C, Galicia LB, Prando C, et al. Germline CYBB mutations that selectively affect macrophages in kindreds with X-linked predisposition to tuberculous mycobacterial disease. Nat Immunol. 2011;12:213–21.PubMedPubMedCentralCrossRef Bustamante J, Arias AA, Vogt G, Picard C, Galicia LB, Prando C, et al. Germline CYBB mutations that selectively affect macrophages in kindreds with X-linked predisposition to tuberculous mycobacterial disease. Nat Immunol. 2011;12:213–21.PubMedPubMedCentralCrossRef
32.
go back to reference Nibbering PH, Pos O, Stevenhagen A, Van Furth R. Interleukin-8 enhances nonoxidative intracellular killing of Mycobacterium fortuitum by human granulocytes. Infect Immun. 1993;61:3111–6.PubMedPubMedCentral Nibbering PH, Pos O, Stevenhagen A, Van Furth R. Interleukin-8 enhances nonoxidative intracellular killing of Mycobacterium fortuitum by human granulocytes. Infect Immun. 1993;61:3111–6.PubMedPubMedCentral
33.
go back to reference Newman GW, Guarnaccia JR, Remold HG, Kazanjian PH. Cytokines enhance neutrophils from human immunodeficiency virus-negative donors and AIDS patients to inhibit the growth of Mycobacterium avium in vitro. J Infect Dis. 1997;175:891–900.PubMedCrossRef Newman GW, Guarnaccia JR, Remold HG, Kazanjian PH. Cytokines enhance neutrophils from human immunodeficiency virus-negative donors and AIDS patients to inhibit the growth of Mycobacterium avium in vitro. J Infect Dis. 1997;175:891–900.PubMedCrossRef
34.
go back to reference Hartmann P, Becker R, Franzen C, Schell-Frederick E, Römer J, Jacobs M, et al. Phagocytosis and killing of Mycobacterium avium complex by human neutrophils. J Leukoc Biol. 2001;69:397–404.PubMed Hartmann P, Becker R, Franzen C, Schell-Frederick E, Römer J, Jacobs M, et al. Phagocytosis and killing of Mycobacterium avium complex by human neutrophils. J Leukoc Biol. 2001;69:397–404.PubMed
35.
go back to reference Appelberg R, Castro AG, Gomes S, Pedrosa J, Silva MT. Susceptibility of beige mice to Mycobacterium avium: role of neutrophils. Infect Immun. 1995;63:3381–7.PubMedPubMedCentral Appelberg R, Castro AG, Gomes S, Pedrosa J, Silva MT. Susceptibility of beige mice to Mycobacterium avium: role of neutrophils. Infect Immun. 1995;63:3381–7.PubMedPubMedCentral
36.
go back to reference Bermudez LE, Petrofsky M, Stevens P. Treatment with recombinant granulocyte colony-stimulating factor (Filgrastin) stimulates neutrophils and tissue macrophages and induces an effective non-specific response against Mycobacterium avium in mice. Immunology. 1998;94:297–303.PubMedPubMedCentralCrossRef Bermudez LE, Petrofsky M, Stevens P. Treatment with recombinant granulocyte colony-stimulating factor (Filgrastin) stimulates neutrophils and tissue macrophages and induces an effective non-specific response against Mycobacterium avium in mice. Immunology. 1998;94:297–303.PubMedPubMedCentralCrossRef
37.
go back to reference Petrofsky M, Bermudez LE. Neutrophils from Mycobacterium avium-infected mice produce TNF-alpha, IL-12, and IL-1 beta and have a putative role in early host response. Clin Immunol. 1999;91:354–8.PubMedCrossRef Petrofsky M, Bermudez LE. Neutrophils from Mycobacterium avium-infected mice produce TNF-alpha, IL-12, and IL-1 beta and have a putative role in early host response. Clin Immunol. 1999;91:354–8.PubMedCrossRef
38.
go back to reference Hsu AP, Sampaio EP, Khan J, Calvo KR, Lemieux JE, Patel SY, et al. Mutations in GATA2 are associated with the autosomal dominant and sporadic monocytopenia and mycobacterial infection (MonoMAC) syndrome. Blood. 2011;118:2653–5.PubMedPubMedCentralCrossRef Hsu AP, Sampaio EP, Khan J, Calvo KR, Lemieux JE, Patel SY, et al. Mutations in GATA2 are associated with the autosomal dominant and sporadic monocytopenia and mycobacterial infection (MonoMAC) syndrome. Blood. 2011;118:2653–5.PubMedPubMedCentralCrossRef
39.
go back to reference Dickinson RE, Milne P, Jardine L, Zandi S, Swierczek SI, McGovern N, et al. The evolution of cellular deficiency in GATA2 mutation. Blood. 2014;123:863–74.PubMedPubMedCentralCrossRef Dickinson RE, Milne P, Jardine L, Zandi S, Swierczek SI, McGovern N, et al. The evolution of cellular deficiency in GATA2 mutation. Blood. 2014;123:863–74.PubMedPubMedCentralCrossRef
40.
go back to reference Spinner MA, Sanchez LA, Hsu AP, Shaw PA, Zerbe CS, Calvo KR, et al. GATA2 deficiency: a protean disorder of hematopoiesis, lymphatics, and immunity. Blood. 2014;123:809–21.PubMedPubMedCentralCrossRef Spinner MA, Sanchez LA, Hsu AP, Shaw PA, Zerbe CS, Calvo KR, et al. GATA2 deficiency: a protean disorder of hematopoiesis, lymphatics, and immunity. Blood. 2014;123:809–21.PubMedPubMedCentralCrossRef
42.
go back to reference Marciano BE, Huang C-Y, Joshi G, Rezaei N, Carvalho BC, Allwood Z, et al. BCG vaccination in patients with severe combined immunodeficiency: complications, risks, and vaccination policies. J Allergy Clin Immunol. 2014;133:1134–41.PubMedPubMedCentralCrossRef Marciano BE, Huang C-Y, Joshi G, Rezaei N, Carvalho BC, Allwood Z, et al. BCG vaccination in patients with severe combined immunodeficiency: complications, risks, and vaccination policies. J Allergy Clin Immunol. 2014;133:1134–41.PubMedPubMedCentralCrossRef
43.
go back to reference Ahmad DS, Esmadi M, Steinmann WC. Idiopathic CD4 Lymphocytopenia: Spectrum of opportunistic infections, malignancies, and autoimmune diseases. Avicenna J Med. 2013;3:37–47.PubMedPubMedCentralCrossRef Ahmad DS, Esmadi M, Steinmann WC. Idiopathic CD4 Lymphocytopenia: Spectrum of opportunistic infections, malignancies, and autoimmune diseases. Avicenna J Med. 2013;3:37–47.PubMedPubMedCentralCrossRef
44.
go back to reference Oksenhendler E, Gérard L, Fieschi C, Malphettes M, Mouillot G, Jaussaud R, et al. Infections in 252 patients with common variable immunodeficiency. Clin Infect Dis. 2008;46:1547–54.PubMedCrossRef Oksenhendler E, Gérard L, Fieschi C, Malphettes M, Mouillot G, Jaussaud R, et al. Infections in 252 patients with common variable immunodeficiency. Clin Infect Dis. 2008;46:1547–54.PubMedCrossRef
45.
go back to reference Winkelstein JA, Marino MC, Lederman HM, Jones SM, Sullivan K, Burks AW, et al. X-linked agammaglobulinemia: report on a United States registry of 201 patients. Medicine (Baltimore). 2006;85:193–202.CrossRef Winkelstein JA, Marino MC, Lederman HM, Jones SM, Sullivan K, Burks AW, et al. X-linked agammaglobulinemia: report on a United States registry of 201 patients. Medicine (Baltimore). 2006;85:193–202.CrossRef
46.
go back to reference Höflich C, Sabat R, Rosseau S, Temmesfeld B, Slevogt H, Döcke W-D, et al. Naturally occurring anti-IFN-gamma autoantibody and severe infections with Mycobacterium cheloneae and Burkholderia cocovenenans. Blood. 2004;103:673–5.PubMedCrossRef Höflich C, Sabat R, Rosseau S, Temmesfeld B, Slevogt H, Döcke W-D, et al. Naturally occurring anti-IFN-gamma autoantibody and severe infections with Mycobacterium cheloneae and Burkholderia cocovenenans. Blood. 2004;103:673–5.PubMedCrossRef
47.
go back to reference Döffinger R, Helbert MR, Barcenas-Morales G, Yang K, Dupuis S, Ceron-Gutierrez L, et al. Autoantibodies to interferon-gamma in a patient with selective susceptibility to mycobacterial infection and organ-specific autoimmunity. Clin Infect Dis. 2004;38:e10–4.PubMedCrossRef Döffinger R, Helbert MR, Barcenas-Morales G, Yang K, Dupuis S, Ceron-Gutierrez L, et al. Autoantibodies to interferon-gamma in a patient with selective susceptibility to mycobacterial infection and organ-specific autoimmunity. Clin Infect Dis. 2004;38:e10–4.PubMedCrossRef
48.
go back to reference Browne SK. Anticytokine autoantibody-associated immunodeficiency. Annu Rev Immunol. 2014;32:635–57.PubMedCrossRef Browne SK. Anticytokine autoantibody-associated immunodeficiency. Annu Rev Immunol. 2014;32:635–57.PubMedCrossRef
49.
go back to reference Otome O, O’Reilly M, Lim L. Disseminated Mycobacterium haemophilum skeletal disease in a patient with interferon-gamma deficiency. Intern Med J. 2015;45:1073–6.PubMedCrossRef Otome O, O’Reilly M, Lim L. Disseminated Mycobacterium haemophilum skeletal disease in a patient with interferon-gamma deficiency. Intern Med J. 2015;45:1073–6.PubMedCrossRef
50.
go back to reference Browne SK, Burbelo PD, Chetchotisakd P, Suputtamongkol Y, Kiertiburanakul S, Shaw PA, et al. Adult-onset immunodeficiency in Thailand and Taiwan. N Engl J Med. 2012;367:725–34.PubMedPubMedCentralCrossRef Browne SK, Burbelo PD, Chetchotisakd P, Suputtamongkol Y, Kiertiburanakul S, Shaw PA, et al. Adult-onset immunodeficiency in Thailand and Taiwan. N Engl J Med. 2012;367:725–34.PubMedPubMedCentralCrossRef
51.
go back to reference Czaja CA, Merkel PA, Chan ED, Lenz LL, Wolf ML, Alam R, et al. Rituximab as successful adjunct treatment in a patient with disseminated nontuberculous mycobacterial infection due to acquired anti-interferon-γ autoantibody. Clin Infect Dis. 2014;58:e115–8.PubMedPubMedCentralCrossRef Czaja CA, Merkel PA, Chan ED, Lenz LL, Wolf ML, Alam R, et al. Rituximab as successful adjunct treatment in a patient with disseminated nontuberculous mycobacterial infection due to acquired anti-interferon-γ autoantibody. Clin Infect Dis. 2014;58:e115–8.PubMedPubMedCentralCrossRef
52.
go back to reference Browne SK, Zaman R, Sampaio EP, Jutivorakool K, Rosen LB, Ding L, et al. Anti-CD20 (rituximab) therapy for anti-IFN-γ autoantibody-associated nontuberculous mycobacterial infection. Blood. 2012;119:3933–9.PubMedPubMedCentralCrossRef Browne SK, Zaman R, Sampaio EP, Jutivorakool K, Rosen LB, Ding L, et al. Anti-CD20 (rituximab) therapy for anti-IFN-γ autoantibody-associated nontuberculous mycobacterial infection. Blood. 2012;119:3933–9.PubMedPubMedCentralCrossRef
53.
go back to reference Meager A, Vincent A, Newsom-Davis J, Willcox N. Spontaneous neutralising antibodies to interferon--alpha and interleukin-12 in thymoma-associated autoimmune disease. Lancet (London, England). 1997;350:1596–7.CrossRef Meager A, Vincent A, Newsom-Davis J, Willcox N. Spontaneous neutralising antibodies to interferon--alpha and interleukin-12 in thymoma-associated autoimmune disease. Lancet (London, England). 1997;350:1596–7.CrossRef
54.
go back to reference Moulias R, Proust J, Wang A, Congy F, Marescot MR, Deville Chabrolle A, et al. Age-related increase in autoantibodies. Lancet (London, England). 1984;1:1128–9.CrossRef Moulias R, Proust J, Wang A, Congy F, Marescot MR, Deville Chabrolle A, et al. Age-related increase in autoantibodies. Lancet (London, England). 1984;1:1128–9.CrossRef
55.
go back to reference Kartalija M, Ovrutsky AR, Bryan CL, Pott GB, Fantuzzi G, Thomas J, et al. Patients with nontuberculous mycobacterial lung disease exhibit unique body and immune phenotypes. Am J Respir Crit Care Med. 2013;187:197–205.PubMedCrossRef Kartalija M, Ovrutsky AR, Bryan CL, Pott GB, Fantuzzi G, Thomas J, et al. Patients with nontuberculous mycobacterial lung disease exhibit unique body and immune phenotypes. Am J Respir Crit Care Med. 2013;187:197–205.PubMedCrossRef
56.
go back to reference Greinert U, Schlaak M, Rũsch-Gerdes S, Flad H-D, Ernst M. Low In Vitro Production of Interferon-γ and Tumor Necrosis Factor-α in HIV-Seronegative Patients with Pulmonary Disease Caused by Nontuberculous Mycobacteria. J Clin Immunol. 2000;20:445–52.PubMedCrossRef Greinert U, Schlaak M, Rũsch-Gerdes S, Flad H-D, Ernst M. Low In Vitro Production of Interferon-γ and Tumor Necrosis Factor-α in HIV-Seronegative Patients with Pulmonary Disease Caused by Nontuberculous Mycobacteria. J Clin Immunol. 2000;20:445–52.PubMedCrossRef
57.
go back to reference Kwon YS, Kim EJ, Lee S-H, Suh GY, Chung MP, Kim H, et al. Decreased cytokine production in patients with nontuberculous mycobacterial lung disease. Lung. 2007;185:337–41.PubMedCrossRef Kwon YS, Kim EJ, Lee S-H, Suh GY, Chung MP, Kim H, et al. Decreased cytokine production in patients with nontuberculous mycobacterial lung disease. Lung. 2007;185:337–41.PubMedCrossRef
58.
go back to reference Milanés-Virelles MT, García-García I, Santos-Herrera Y, Valdés-Quintana M, Valenzuela-Silva CM, Jiménez-Madrigal G, et al. Adjuvant interferon gamma in patients with pulmonary atypical Mycobacteriosis: a randomized, double-blind, placebo-controlled study. BMC Infect Dis. 2008;8:17.PubMedPubMedCentralCrossRef Milanés-Virelles MT, García-García I, Santos-Herrera Y, Valdés-Quintana M, Valenzuela-Silva CM, Jiménez-Madrigal G, et al. Adjuvant interferon gamma in patients with pulmonary atypical Mycobacteriosis: a randomized, double-blind, placebo-controlled study. BMC Infect Dis. 2008;8:17.PubMedPubMedCentralCrossRef
59.
go back to reference Kim RD, Greenberg DE, Ehrmantraut ME, Guide SV, Ding L, Shea Y, et al. Pulmonary nontuberculous mycobacterial disease: prospective study of a distinct preexisting syndrome. Am J Respir Crit Care Med. 2008;178:1066–74.PubMedPubMedCentralCrossRef Kim RD, Greenberg DE, Ehrmantraut ME, Guide SV, Ding L, Shea Y, et al. Pulmonary nontuberculous mycobacterial disease: prospective study of a distinct preexisting syndrome. Am J Respir Crit Care Med. 2008;178:1066–74.PubMedPubMedCentralCrossRef
60.
go back to reference Lim A, Allison C, Price P, Waterer G. Susceptibility to pulmonary disease due to Mycobacterium avium-intracellulare complex may reflect low IL-17 and high IL-10 responses rather than Th1 deficiency. Clin Immunol. 2010;137:296–302.PubMedCrossRef Lim A, Allison C, Price P, Waterer G. Susceptibility to pulmonary disease due to Mycobacterium avium-intracellulare complex may reflect low IL-17 and high IL-10 responses rather than Th1 deficiency. Clin Immunol. 2010;137:296–302.PubMedCrossRef
61.
go back to reference Lim A, Allison C, Tan DBA, Oliver B, Price P, Waterer G. Immunological markers of lung disease due to non-tuberculous mycobacteria. Dis Markers. 2010;29:103–9.PubMedPubMedCentralCrossRef Lim A, Allison C, Tan DBA, Oliver B, Price P, Waterer G. Immunological markers of lung disease due to non-tuberculous mycobacteria. Dis Markers. 2010;29:103–9.PubMedPubMedCentralCrossRef
62.
go back to reference Szymanski EP, Leung JM, Fowler CJ, Haney C, Hsu AP, Chen F, et al. Pulmonary Nontuberculous Mycobacterial Infection. A Multisystem, Multigenic Disease. Am J Respir Crit Care Med. 2015;192:618–28.PubMedCrossRef Szymanski EP, Leung JM, Fowler CJ, Haney C, Hsu AP, Chen F, et al. Pulmonary Nontuberculous Mycobacterial Infection. A Multisystem, Multigenic Disease. Am J Respir Crit Care Med. 2015;192:618–28.PubMedCrossRef
63.
go back to reference Koh W-J, Kwon OJ, Kim EJ, Lee KS, Ki C-S, Kim JW. NRAMP1 gene polymorphism and susceptibility to nontuberculous mycobacterial lung diseases. Chest. 2005;128:94–101.PubMedCrossRef Koh W-J, Kwon OJ, Kim EJ, Lee KS, Ki C-S, Kim JW. NRAMP1 gene polymorphism and susceptibility to nontuberculous mycobacterial lung diseases. Chest. 2005;128:94–101.PubMedCrossRef
64.
go back to reference Horsburgh CR. Mycobacterium avium complex infection in the acquired immunodeficiency syndrome. N Engl J Med. 1991;324:1332–8.PubMedCrossRef Horsburgh CR. Mycobacterium avium complex infection in the acquired immunodeficiency syndrome. N Engl J Med. 1991;324:1332–8.PubMedCrossRef
65.
go back to reference Bloch KC, Zwerling L, Pletcher MJ, Hahn JA, Gerberding JL, Ostroff SM, et al. Incidence and clinical implications of isolation of Mycobacterium kansasii: results of a 5-year, population-based study. Ann Intern Med. 1998;129:698–704.PubMedCrossRef Bloch KC, Zwerling L, Pletcher MJ, Hahn JA, Gerberding JL, Ostroff SM, et al. Incidence and clinical implications of isolation of Mycobacterium kansasii: results of a 5-year, population-based study. Ann Intern Med. 1998;129:698–704.PubMedCrossRef
66.
go back to reference Campo RE, Campo CE. Mycobacterium kansasii disease in patients infected with human immunodeficiency virus. Clin Infect Dis. 1997;24:1233–8.PubMedCrossRef Campo RE, Campo CE. Mycobacterium kansasii disease in patients infected with human immunodeficiency virus. Clin Infect Dis. 1997;24:1233–8.PubMedCrossRef
67.
go back to reference Meije Y, Piersimoni C, Torre-Cisneros J, Dilektasli AG, Aguado JM. Mycobacterial infections in solid organ transplant recipients. Clin Microbiol Infect. 2014;20 Suppl 7:89–101.PubMedCrossRef Meije Y, Piersimoni C, Torre-Cisneros J, Dilektasli AG, Aguado JM. Mycobacterial infections in solid organ transplant recipients. Clin Microbiol Infect. 2014;20 Suppl 7:89–101.PubMedCrossRef
68.
go back to reference Piersimoni C. Nontuberculous mycobacteria infection in solid organ transplant recipients. Eur J Clin Microbiol Infect Dis. 2012;31:397–403.PubMedCrossRef Piersimoni C. Nontuberculous mycobacteria infection in solid organ transplant recipients. Eur J Clin Microbiol Infect Dis. 2012;31:397–403.PubMedCrossRef
69.
go back to reference Wu U-I, Holland SM. Host susceptibility to non-tuberculous mycobacterial infections. Lancet Infect Dis. 2015;15. Wu U-I, Holland SM. Host susceptibility to non-tuberculous mycobacterial infections. Lancet Infect Dis. 2015;15.
70.
go back to reference Ho TA, Rommelaere M, Coche E, Yombi J-C, Kanaan N. Nontuberculous mycobacterial pulmonary infection in renal transplant recipients. Transpl Infect Dis. 2010;12:138–42.PubMedCrossRef Ho TA, Rommelaere M, Coche E, Yombi J-C, Kanaan N. Nontuberculous mycobacterial pulmonary infection in renal transplant recipients. Transpl Infect Dis. 2010;12:138–42.PubMedCrossRef
71.
go back to reference Al-Anazi KA, Al-Jasser AM, Al-Anazi WK. Infections caused by non-tuberculous mycobacteria in recipients of hematopoietic stem cell transplantation. Front Oncol. 2014;4:311.PubMedPubMedCentral Al-Anazi KA, Al-Jasser AM, Al-Anazi WK. Infections caused by non-tuberculous mycobacteria in recipients of hematopoietic stem cell transplantation. Front Oncol. 2014;4:311.PubMedPubMedCentral
72.
go back to reference Gaviria JM, Garcia PJ, Garrido SM, Corey L, Boeckh M. Nontuberculous mycobacterial infections in hematopoietic stem cell transplant recipients: characteristics of respiratory and catheter-related infections. Biol Blood Marrow Transplant. 2000;6:361–9.PubMedCrossRef Gaviria JM, Garcia PJ, Garrido SM, Corey L, Boeckh M. Nontuberculous mycobacterial infections in hematopoietic stem cell transplant recipients: characteristics of respiratory and catheter-related infections. Biol Blood Marrow Transplant. 2000;6:361–9.PubMedCrossRef
73.
go back to reference Koetz K, Bryl E, Spickschen K, O’Fallon WM, Goronzy JJ, Weyand CM. T cell homeostasis in patients with rheumatoid arthritis. Proc Natl Acad Sci. 2000;97:9203–8.PubMedPubMedCentralCrossRef Koetz K, Bryl E, Spickschen K, O’Fallon WM, Goronzy JJ, Weyand CM. T cell homeostasis in patients with rheumatoid arthritis. Proc Natl Acad Sci. 2000;97:9203–8.PubMedPubMedCentralCrossRef
74.
go back to reference Hodgson K, Morris J, Bridson T, Govan B, Rush C, Ketheesan N. Immunological mechanisms contributing to the double burden of diabetes and intracellular bacterial infections. Immunology. 2015;144:171–85.PubMedPubMedCentralCrossRef Hodgson K, Morris J, Bridson T, Govan B, Rush C, Ketheesan N. Immunological mechanisms contributing to the double burden of diabetes and intracellular bacterial infections. Immunology. 2015;144:171–85.PubMedPubMedCentralCrossRef
75.
go back to reference Winthrop KL, Baxter R, Liu L, Varley CD, Curtis JR, Baddley JW, et al. Mycobacterial diseases and antitumour necrosis factor therapy in USA. Ann Rheum Dis. 2013;72:37–42.PubMedCrossRef Winthrop KL, Baxter R, Liu L, Varley CD, Curtis JR, Baddley JW, et al. Mycobacterial diseases and antitumour necrosis factor therapy in USA. Ann Rheum Dis. 2013;72:37–42.PubMedCrossRef
76.
go back to reference Winthrop KL, Chang E, Yamashita S, Iademarco MF, LoBue PA. Nontuberculous Mycobacteria Infections and Anti–Tumor Necrosis Factor-α Therapy. Emerg Infect Dis. 2009;15:1556–61.PubMedPubMedCentralCrossRef Winthrop KL, Chang E, Yamashita S, Iademarco MF, LoBue PA. Nontuberculous Mycobacteria Infections and Anti–Tumor Necrosis Factor-α Therapy. Emerg Infect Dis. 2009;15:1556–61.PubMedPubMedCentralCrossRef
77.
go back to reference Andrejak C, Nielsen R, Thomsen VO, Duhaut P, Sorensen HT, Thomsen RW. Chronic respiratory disease, inhaled corticosteroids and risk of non-tuberculous mycobacteriosis. Thorax. 2012;68:256–62.PubMedCrossRef Andrejak C, Nielsen R, Thomsen VO, Duhaut P, Sorensen HT, Thomsen RW. Chronic respiratory disease, inhaled corticosteroids and risk of non-tuberculous mycobacteriosis. Thorax. 2012;68:256–62.PubMedCrossRef
78.
go back to reference Brode SK, Jamieson FB, Ng R, Campitelli MA, Kwong JC, Paterson JM, Li P, et al. Increased risk of mycobacterial infections associated with anti-rheumatic medications. Thorax. 2015;70:677–82.PubMedCrossRef Brode SK, Jamieson FB, Ng R, Campitelli MA, Kwong JC, Paterson JM, Li P, et al. Increased risk of mycobacterial infections associated with anti-rheumatic medications. Thorax. 2015;70:677–82.PubMedCrossRef
79.
go back to reference Hojo M, Iikura M, Hirano S, Sugiyama H, Kobayashi N, Kudo K. Increased risk of nontuberculous mycobacterial infection in asthmatic patients using long-term inhaled corticosteroid therapy. Respirology. 2012;17:185–90.PubMedCrossRef Hojo M, Iikura M, Hirano S, Sugiyama H, Kobayashi N, Kudo K. Increased risk of nontuberculous mycobacterial infection in asthmatic patients using long-term inhaled corticosteroid therapy. Respirology. 2012;17:185–90.PubMedCrossRef
80.
go back to reference Mirsaeidi M, Hadid W, Ericsoussi B, Rodgers D, Sadikot RT. Non-tuberculous mycobacterial disease is common in patients with non-cystic fibrosis bronchiectasis. Int J Infect Dis. 2013;17:e1000–4.PubMedPubMedCentralCrossRef Mirsaeidi M, Hadid W, Ericsoussi B, Rodgers D, Sadikot RT. Non-tuberculous mycobacterial disease is common in patients with non-cystic fibrosis bronchiectasis. Int J Infect Dis. 2013;17:e1000–4.PubMedPubMedCentralCrossRef
81.
go back to reference Chu H, Zhao L, Xiao H, Zhang Z, Zhang J, Gui T, et al. Prevalence of nontuberculous mycobacteria in patients with bronchiectasis: a meta-analysis. Arch Med Sci. 2014;10:661–8.PubMedPubMedCentralCrossRef Chu H, Zhao L, Xiao H, Zhang Z, Zhang J, Gui T, et al. Prevalence of nontuberculous mycobacteria in patients with bronchiectasis: a meta-analysis. Arch Med Sci. 2014;10:661–8.PubMedPubMedCentralCrossRef
82.
go back to reference Cole PJ. Inflammation: a two-edged sword--the model of bronchiectasis. Eur J Respir Dis Suppl. 1986;147:6–15.PubMed Cole PJ. Inflammation: a two-edged sword--the model of bronchiectasis. Eur J Respir Dis Suppl. 1986;147:6–15.PubMed
83.
go back to reference Ojo O, Lagan AL, Rajendran V, Spanjer A, Chen L, Sohal SS, et al. Pathological changes in the COPD lung mesenchyme - Novel lessons learned from in??vitro and in??vivo studies. Pulm Pharmacol Ther. 2014;29(April):1–8. Ojo O, Lagan AL, Rajendran V, Spanjer A, Chen L, Sohal SS, et al. Pathological changes in the COPD lung mesenchyme - Novel lessons learned from in??vitro and in??vivo studies. Pulm Pharmacol Ther. 2014;29(April):1–8.
84.
go back to reference Hoefsloot W, van Ingen J, Magis-Escurra C, Reijers MH, van Soolingen D, Dekhuijzen RPN, et al. Prevalence of nontuberculous mycobacteria in COPD patients with exacerbations. J Infect. 2013;66:542–5.PubMedCrossRef Hoefsloot W, van Ingen J, Magis-Escurra C, Reijers MH, van Soolingen D, Dekhuijzen RPN, et al. Prevalence of nontuberculous mycobacteria in COPD patients with exacerbations. J Infect. 2013;66:542–5.PubMedCrossRef
85.
go back to reference Yeh J-J, Wang Y-C, Sung F-C, Chou CY-T, Kao C-H. Nontuberculosis Mycobacterium Disease is a Risk Factor for Chronic Obstructive Pulmonary Disease: A Nationwide Cohort Study. Lung. 2014;192:403–11.PubMedCrossRef Yeh J-J, Wang Y-C, Sung F-C, Chou CY-T, Kao C-H. Nontuberculosis Mycobacterium Disease is a Risk Factor for Chronic Obstructive Pulmonary Disease: A Nationwide Cohort Study. Lung. 2014;192:403–11.PubMedCrossRef
86.
go back to reference McDonnell MJ, Ward C, Lordan JL, Rutherford RM. Non-cystic fibrosis bronchiectasis. QJM. 2013;106:709–15.PubMedCrossRef McDonnell MJ, Ward C, Lordan JL, Rutherford RM. Non-cystic fibrosis bronchiectasis. QJM. 2013;106:709–15.PubMedCrossRef
87.
go back to reference Hassett DJ, Borchers MT, Panos RJ. Chronic obstructive pulmonary disease (COPD): Evaluation from clinical, immunological and bacterial pathogenesis perspectives. J Microbiol. 2014;52:211–26.PubMedCrossRef Hassett DJ, Borchers MT, Panos RJ. Chronic obstructive pulmonary disease (COPD): Evaluation from clinical, immunological and bacterial pathogenesis perspectives. J Microbiol. 2014;52:211–26.PubMedCrossRef
88.
go back to reference Tang AC, Turvey SE, Alves MP, Regamey N, Tümmler B, Hartl D. Current concepts: host-pathogen interactions in cystic fibrosis airways disease. Eur Respir Rev. 2014;23:320–32.PubMedCrossRef Tang AC, Turvey SE, Alves MP, Regamey N, Tümmler B, Hartl D. Current concepts: host-pathogen interactions in cystic fibrosis airways disease. Eur Respir Rev. 2014;23:320–32.PubMedCrossRef
89.
go back to reference Martiniano SL, Nick JA. Nontuberculous mycobacterial infections in cystic fibrosis. Clin Chest Med. 2015;36:101–15.PubMedCrossRef Martiniano SL, Nick JA. Nontuberculous mycobacterial infections in cystic fibrosis. Clin Chest Med. 2015;36:101–15.PubMedCrossRef
90.
go back to reference Bryant JM, Grogono DM, Greaves D, Foweraker J, Roddick I, Inns T, et al. Whole-genome sequencing to identify transmission of Mycobacterium abscessus between patients with cystic fibrosis: a retrospective cohort study. Lancet (London, England). 2013;381:1551–60.CrossRef Bryant JM, Grogono DM, Greaves D, Foweraker J, Roddick I, Inns T, et al. Whole-genome sequencing to identify transmission of Mycobacterium abscessus between patients with cystic fibrosis: a retrospective cohort study. Lancet (London, England). 2013;381:1551–60.CrossRef
91.
go back to reference Ziedalski TM. Prospective Analysis of Cystic Fibrosis Transmembrane Regulator Mutations in Adults With Bronchiectasis or Pulmonary Nontuberculous Mycobacterial Infection. CHEST J. 2006;130:995.CrossRef Ziedalski TM. Prospective Analysis of Cystic Fibrosis Transmembrane Regulator Mutations in Adults With Bronchiectasis or Pulmonary Nontuberculous Mycobacterial Infection. CHEST J. 2006;130:995.CrossRef
92.
go back to reference Fowler CJ, Olivier KN, Leung JM, Smith CC, Huth AG, Root H, et al. Abnormal nasal nitric oxide production, ciliary beat frequency, and Toll-like receptor response in pulmonary nontuberculous mycobacterial disease epithelium. Am J Respir Crit Care Med. 2013;187:1374–81.PubMedPubMedCentralCrossRef Fowler CJ, Olivier KN, Leung JM, Smith CC, Huth AG, Root H, et al. Abnormal nasal nitric oxide production, ciliary beat frequency, and Toll-like receptor response in pulmonary nontuberculous mycobacterial disease epithelium. Am J Respir Crit Care Med. 2013;187:1374–81.PubMedPubMedCentralCrossRef
93.
go back to reference Jang M-A, Kim S-Y, Jeong B-H, Park HY, Jeon K, Kim J-W, et al. Association of CFTR gene variants with nontuberculous mycobacterial lung disease in a Korean population with a low prevalence of cystic fibrosis. J Hum Genet. 2013;58:298–303.PubMedCrossRef Jang M-A, Kim S-Y, Jeong B-H, Park HY, Jeon K, Kim J-W, et al. Association of CFTR gene variants with nontuberculous mycobacterial lung disease in a Korean population with a low prevalence of cystic fibrosis. J Hum Genet. 2013;58:298–303.PubMedCrossRef
94.
95.
go back to reference Andréjak C, Thomsen VØ, Johansen IS, Riis A, Benfield TL, Duhaut P, et al. Nontuberculous pulmonary mycobacteriosis in Denmark: incidence and prognostic factors. Am J Respir Crit Care Med. 2010;181:514–21.PubMedCrossRef Andréjak C, Thomsen VØ, Johansen IS, Riis A, Benfield TL, Duhaut P, et al. Nontuberculous pulmonary mycobacteriosis in Denmark: incidence and prognostic factors. Am J Respir Crit Care Med. 2010;181:514–21.PubMedCrossRef
96.
go back to reference Ye P, Kirschner DE. Measuring emigration of human thymocytes by T-cell receptor excision circles. Crit Rev Immunol. 2002;22:483–97.PubMedCrossRef Ye P, Kirschner DE. Measuring emigration of human thymocytes by T-cell receptor excision circles. Crit Rev Immunol. 2002;22:483–97.PubMedCrossRef
97.
go back to reference Cicin-Sain L, Messaoudi I, Park B, Currier N, Planer S, Fischer M, et al. Dramatic increase in naive T cell turnover is linked to loss of naive T cells from old primates. Proc Natl Acad Sci U S A. 2007;104:19960–5.PubMedPubMedCentralCrossRef Cicin-Sain L, Messaoudi I, Park B, Currier N, Planer S, Fischer M, et al. Dramatic increase in naive T cell turnover is linked to loss of naive T cells from old primates. Proc Natl Acad Sci U S A. 2007;104:19960–5.PubMedPubMedCentralCrossRef
98.
go back to reference Naylor K, Li G, Vallejo AN, Lee W-W, Koetz K, Bryl E, Witkowski J, et al. The influence of age on T cell generation and TCR diversity. J Immunol. 2005;174:7446–52.PubMedCrossRef Naylor K, Li G, Vallejo AN, Lee W-W, Koetz K, Bryl E, Witkowski J, et al. The influence of age on T cell generation and TCR diversity. J Immunol. 2005;174:7446–52.PubMedCrossRef
99.
go back to reference Nikolich-Zugich J. Ageing and life-long maintenance of T-cell subsets in the face of latent persistent infections. Nat Rev Immunol. 2008;8:512–22.PubMedCrossRef Nikolich-Zugich J. Ageing and life-long maintenance of T-cell subsets in the face of latent persistent infections. Nat Rev Immunol. 2008;8:512–22.PubMedCrossRef
100.
go back to reference Griffith DE, Girard WM, Wallace RJ. Clinical features of pulmonary disease caused by rapidly growing mycobacteria. An analysis of 154 patients. Am Rev Respir Dis. 1993;147:1271–8.PubMedCrossRef Griffith DE, Girard WM, Wallace RJ. Clinical features of pulmonary disease caused by rapidly growing mycobacteria. An analysis of 154 patients. Am Rev Respir Dis. 1993;147:1271–8.PubMedCrossRef
101.
go back to reference Tsuyuguchi K, Suzuki K, Matsumoto H, Tanaka E, Amitani R, Kuze F. Effect of oestrogen on Mycobacterium avium complex pulmonary infection in mice. Clin Exp Immunol. 2001;123:428–34.PubMedPubMedCentralCrossRef Tsuyuguchi K, Suzuki K, Matsumoto H, Tanaka E, Amitani R, Kuze F. Effect of oestrogen on Mycobacterium avium complex pulmonary infection in mice. Clin Exp Immunol. 2001;123:428–34.PubMedPubMedCentralCrossRef
102.
go back to reference Danley J, Kwait R, Peterson DD, Sendecki J, Vaughn B, Nakisbendi K, et al. Normal estrogen, but low dehydroepiandrosterone levels, in women with pulmonary Mycobacterium avium complex. A preliminary study. Ann Am Thorac Soc. 2014;11:908–14.PubMedCrossRef Danley J, Kwait R, Peterson DD, Sendecki J, Vaughn B, Nakisbendi K, et al. Normal estrogen, but low dehydroepiandrosterone levels, in women with pulmonary Mycobacterium avium complex. A preliminary study. Ann Am Thorac Soc. 2014;11:908–14.PubMedCrossRef
103.
go back to reference Mirsaeidi M, Sadikot RT. Gender susceptibility to mycobacterial infections in patients with non-CF bronchiectasis. Int J Mycobacteriology. 2015;4:92–6.CrossRef Mirsaeidi M, Sadikot RT. Gender susceptibility to mycobacterial infections in patients with non-CF bronchiectasis. Int J Mycobacteriology. 2015;4:92–6.CrossRef
104.
go back to reference Chan ED, Iseman MD. Slender, older women appear to be more susceptible to nontuberculous mycobacterial lung disease. Gend Med. 2010;7:5–18.PubMedCrossRef Chan ED, Iseman MD. Slender, older women appear to be more susceptible to nontuberculous mycobacterial lung disease. Gend Med. 2010;7:5–18.PubMedCrossRef
105.
go back to reference Dirac MA, Horan KL, Doody DR, Meschke JS, Park DR, Jackson LA, et al. Environment or host?: A case–control study of risk factors for Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2012;186:684–91.PubMedCrossRef Dirac MA, Horan KL, Doody DR, Meschke JS, Park DR, Jackson LA, et al. Environment or host?: A case–control study of risk factors for Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2012;186:684–91.PubMedCrossRef
106.
go back to reference Lee SJ, Ryu YJ, Lee JH, Chang JH, Shim SS. The impact of low subcutaneous fat in patients with nontuberculous mycobacterial lung disease. Lung. 2014;192:395–401.PubMedCrossRef Lee SJ, Ryu YJ, Lee JH, Chang JH, Shim SS. The impact of low subcutaneous fat in patients with nontuberculous mycobacterial lung disease. Lung. 2014;192:395–401.PubMedCrossRef
107.
go back to reference Ordway D, Henao-Tamayo M, Smith E, Shanley C, Harton M, Troudt J, et al. Animal model of Mycobacterium abscessus lung infection. J Leukoc Biol. 2008;83:1502–11.PubMedCrossRef Ordway D, Henao-Tamayo M, Smith E, Shanley C, Harton M, Troudt J, et al. Animal model of Mycobacterium abscessus lung infection. J Leukoc Biol. 2008;83:1502–11.PubMedCrossRef
108.
go back to reference Leung JM, Fowler C, Smith C, Adjemian J, Frein C, Claypool RJ, et al. A familial syndrome of pulmonary nontuberculous mycobacteria infections. Am J Respir Crit Care Med. 2013;188:1373–6.PubMedPubMedCentralCrossRef Leung JM, Fowler C, Smith C, Adjemian J, Frein C, Claypool RJ, et al. A familial syndrome of pulmonary nontuberculous mycobacteria infections. Am J Respir Crit Care Med. 2013;188:1373–6.PubMedPubMedCentralCrossRef
109.
go back to reference Iseman MD, Buschman DL, Ackerson LM. Pectus excavatum and scoliosis. Thoracic anomalies associated with pulmonary disease caused by Mycobacterium avium complex. Am Rev Respir Dis. 1991;144:914–6.PubMedCrossRef Iseman MD, Buschman DL, Ackerson LM. Pectus excavatum and scoliosis. Thoracic anomalies associated with pulmonary disease caused by Mycobacterium avium complex. Am Rev Respir Dis. 1991;144:914–6.PubMedCrossRef
110.
go back to reference Jeon K, Kim S-Y, Jeong B-H, Chang B, Shin SJ, Koh W-J. Severe vitamin D deficiency is associated with non-tuberculous mycobacterial lung disease: a case–control study. Respirology. 2013;18:983–8.PubMedCrossRef Jeon K, Kim S-Y, Jeong B-H, Chang B, Shin SJ, Koh W-J. Severe vitamin D deficiency is associated with non-tuberculous mycobacterial lung disease: a case–control study. Respirology. 2013;18:983–8.PubMedCrossRef
112.
go back to reference Casanova J-L, Abel L. The human model: a genetic dissection of immunity to infection in natural conditions. Nat Rev Immunol. 2004;4:55–66.PubMedCrossRef Casanova J-L, Abel L. The human model: a genetic dissection of immunity to infection in natural conditions. Nat Rev Immunol. 2004;4:55–66.PubMedCrossRef
113.
go back to reference Tsai T-F, Chiu H-Y, Song M, Chan D. A case of latent tuberculosis reactivation in a patient treated with ustekinumab without concomitant isoniazid chemoprophylaxis in the PEARL trial. Br J Dermatol. 2013;168:444–6.PubMedCrossRef Tsai T-F, Chiu H-Y, Song M, Chan D. A case of latent tuberculosis reactivation in a patient treated with ustekinumab without concomitant isoniazid chemoprophylaxis in the PEARL trial. Br J Dermatol. 2013;168:444–6.PubMedCrossRef
114.
go back to reference Souto A, Maneiro JR, Salgado E, Carmona L, Gomez-Reino JJ. Risk of tuberculosis in patients with chronic immune-mediated inflammatory diseases treated with biologics and tofacitinib: a systematic review and meta-analysis of randomized controlled trials and long-term extension studies. Rheumatology (Oxford). 2014;53:1872–85.CrossRef Souto A, Maneiro JR, Salgado E, Carmona L, Gomez-Reino JJ. Risk of tuberculosis in patients with chronic immune-mediated inflammatory diseases treated with biologics and tofacitinib: a systematic review and meta-analysis of randomized controlled trials and long-term extension studies. Rheumatology (Oxford). 2014;53:1872–85.CrossRef
115.
go back to reference Hopman RK, Lawrence SJ, Oh ST. Disseminated tuberculosis associated with ruxolitinib. Leukemia. 2014;28:1750–1.PubMedCrossRef Hopman RK, Lawrence SJ, Oh ST. Disseminated tuberculosis associated with ruxolitinib. Leukemia. 2014;28:1750–1.PubMedCrossRef
116.
go back to reference Winthrop KL, Iseman M. Bedfellows: mycobacteria and rheumatoid arthritis in the era of biologic therapy. Nat Rev Rheumatol. 2013;9:524–31.PubMedCrossRef Winthrop KL, Iseman M. Bedfellows: mycobacteria and rheumatoid arthritis in the era of biologic therapy. Nat Rev Rheumatol. 2013;9:524–31.PubMedCrossRef
117.
go back to reference Novosad SA, Winthrop KL. Beyond tumor necrosis factor inhibition: the expanding pipeline of biologic therapies for inflammatory diseases and their associated infectious sequelae. Clin Infect Dis. 2014;58:1587–98.PubMedCrossRef Novosad SA, Winthrop KL. Beyond tumor necrosis factor inhibition: the expanding pipeline of biologic therapies for inflammatory diseases and their associated infectious sequelae. Clin Infect Dis. 2014;58:1587–98.PubMedCrossRef
118.
go back to reference de Masson A, Maillart E, Veziris N, Meyssonnier V, Papeix C, Caumes E. Cavitary pulmonary disease in a patient treated with natalizumab. Presse Med. 2014;43:1009–12.PubMedCrossRef de Masson A, Maillart E, Veziris N, Meyssonnier V, Papeix C, Caumes E. Cavitary pulmonary disease in a patient treated with natalizumab. Presse Med. 2014;43:1009–12.PubMedCrossRef
119.
go back to reference Chan ED, Kaminska AM, Gill W, Chmura K, Feldman NE, Bai X, et al. Alpha-1-antitrypsin (AAT) anomalies are associated with lung disease due to rapidly growing mycobacteria and AAT inhibits Mycobacterium abscessus infection of macrophages. Scand J Infect Dis. 2007;39:690–6.PubMedCrossRef Chan ED, Kaminska AM, Gill W, Chmura K, Feldman NE, Bai X, et al. Alpha-1-antitrypsin (AAT) anomalies are associated with lung disease due to rapidly growing mycobacteria and AAT inhibits Mycobacterium abscessus infection of macrophages. Scand J Infect Dis. 2007;39:690–6.PubMedCrossRef
120.
go back to reference McGrath EE, Bardsley P. An association between Mycobacterium malmoense and coal workers’ pneumoconiosis. Lung. 2009;187:51–4. McGrath EE, Bardsley P. An association between Mycobacterium malmoense and coal workers’ pneumoconiosis. Lung. 2009;187:51–4.
121.
go back to reference Kim YM, Kim M, Kim SK, Park K, Jin S-H, Lee US, et al. Mycobacterial infections in coal workers’ pneumoconiosis patients in South Korea. Scand J Infect Dis. 2009;41:656–62.PubMedCrossRef Kim YM, Kim M, Kim SK, Park K, Jin S-H, Lee US, et al. Mycobacterial infections in coal workers’ pneumoconiosis patients in South Korea. Scand J Infect Dis. 2009;41:656–62.PubMedCrossRef
122.
go back to reference Witty LA, Tapson VF, Piantadosi CA. Isolation of mycobacteria in patients with pulmonary alveolar proteinosis. Medicine (Baltimore). 1994;73:103–9.CrossRef Witty LA, Tapson VF, Piantadosi CA. Isolation of mycobacteria in patients with pulmonary alveolar proteinosis. Medicine (Baltimore). 1994;73:103–9.CrossRef
Metadata
Title
‘”Why me, why now?” Using clinical immunology and epidemiology to explain who gets nontuberculous mycobacterial infection
Authors
M. Alexandra Lake
Lyn R. Ambrose
Marc C. I. Lipman
David M. Lowe
Publication date
01-12-2016
Publisher
BioMed Central
Published in
BMC Medicine / Issue 1/2016
Electronic ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-016-0606-6

Other articles of this Issue 1/2016

BMC Medicine 1/2016 Go to the issue