1
|
Wang Z, Sun X, Lin Y, Fu Y, Yi Z. Stealth in non-tuberculous mycobacteria: clever challengers to the immune system. Microbiol Res 2025; 292:128039. [PMID: 39752805 DOI: 10.1016/j.micres.2024.128039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/19/2025]
Abstract
Non-tuberculous Mycobacteria (NTM) are found extensively in various environments, yet most are non-pathogenic. Only a limited number of these organisms can cause various infections, including those affecting the lungs, skin, and central nervous system, particularly when the host's autoimmune function is compromised. Among these, Non-tuberculous Mycobacteria Pulmonary Diseases (NTM-PD) are the most prevalent. Currently, there is a lack of effective treatments and preventive measures for NTM infections. This article aims to deepen the comprehension of the pathogenic mechanisms linked to NTM and to formulate new intervention strategies by synthesizing current research and detailing the different tactics used by NTM to avoid elimination by the host's immune response. These intricate mechanisms not only affect the innate immune response but also successfully oppose the adaptive immune response, establishing persistent infections within the host. This includes effects on the functions of macrophages, neutrophils, dendritic cells, and T lymphocytes, as well as modulation of cytokine production. The article particularly emphasizes the survival strategies of NTM within macrophages, such as inhibiting phagosome maturation and acidification, resisting intracellular killing mechanisms, and interfering with autophagy and cell death pathways. This review aims to deepen the understanding of NTM's immune evasion mechanisms, thereby facilitating efforts to inhibit its proliferation and spread within the host, ultimately providing new methods and strategies for NTM-related treatments.
Collapse
Affiliation(s)
- Zhenghao Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Xiurong Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Yuli Lin
- School of Medical Laboratory, Shandong Second Medical University, Weifang 261053, China
| | - Yurong Fu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China.
| | - Zhengjun Yi
- School of Medical Laboratory, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
2
|
Kim SY, Zo S, Kim DH, Shin SJ, Jhun BW. Single-cell transcriptomics by clinical course of Mycobacterium avium complex pulmonary disease. Sci Rep 2024; 14:15663. [PMID: 38977917 PMCID: PMC11231222 DOI: 10.1038/s41598-024-66523-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024] Open
Abstract
Mycobacterium avium complex pulmonary disease (MAC-PD) has a heterogeneous clinical course. However, immune profiles associated with MAC-PD clinical course are limited. We performed single-cell RNA sequencing of peripheral blood mononuclear cells from 21 MAC-PD patients divided into three clinical courses: group A, spontaneous culture conversion; group B, stable disease without antibiotic treatment; and group C, progressive disease with antibiotic treatment. A lower proportion of NK cells and higher proportion of monocytes were noted in group C compared to combined groups A and B. The proportion of classical monocytes was higher in group C compared to groups A and B, while the proportion of non-classical monocytes decreased. EGR1, HSPA1A, HSPA1B, and CD83 were up-regulated in spontaneous culture conversion group A compared to progressive disease group C. Up-regulation of MYOM2 and LILRA4 and down-regulation of MT-ATP8, CD83, and CCL3L1 was found in progressive disease group C. PCBP1, FOS, RGCC, S100B, G0S2, AREG, and LYN were highly expressed in favorable treatment response compared to unfavorable response. Our findings may offer a comprehensive understanding of the host immune profiles that influence a particular MAC-PD clinical course and could suggest an immunological mechanism associated with the disease progression of MAC-PD.
Collapse
Affiliation(s)
- Su-Young Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sungmin Zo
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Dae Hun Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Byung Woo Jhun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
3
|
Rabe H, Lönnermark E, Johansson E, Gilljam M, Jönsson B. In vitro stimulation with nontuberculous mycobacteria induced a stronger cytokine response in leukocytes isolated from individuals with latent tuberculosis compared to those isolated from active tuberculosis or cystic fibrosis patients. Tuberculosis (Edinb) 2024; 147:102504. [PMID: 38522174 DOI: 10.1016/j.tube.2024.102504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/08/2024] [Accepted: 03/17/2024] [Indexed: 03/26/2024]
Abstract
Mycobacterium tuberculosis and opportunistic environmental non-tuberculous mycobacteria (NTM) can cause severe infection. Why latent tuberculosis infection advances to active disease, and why some individuals with cystic fibrosis (CF) develop pulmonary infections with NTM is still poorly understood. The aim of this study was to investigate the effector function of peripheral blood mononuclear cells (PBMC) from individuals with active or latent tuberculosis, individuals with CF with or without pulmonary NTM-infection and healthy controls, by measuring cytokine response to in vitro stimulation with different species of NTMs. The cytokine concentrations of IL-17A, IL-22, IL-23, IL-10, IL12p70 and IFN-γ were measured in PBMC-culture supernatants after stimulation with NTMs. PBMCs from individuals with latent tuberculosis infection showed strong IL-17A, IL-22, and IFN-γ responses compared to individuals with active tuberculosis or CF. IL-10 production was low in both tuberculosis groups compared to the CF groups and controls. This study suggests that IL-17A and IL-22 might be important to keep tuberculosis in a latent phase and that individuals with CF with an ongoing NTM infection seem to have a low cytokine response.
Collapse
Affiliation(s)
- Hardis Rabe
- Unit of Biological Function, Research Institutes of Sweden (RISE AB), Gothenburg, Sweden; Institute of Biomedicine, Department of infectious diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Elisabeth Lönnermark
- Institute of Biomedicine, Department of infectious diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Infectious Diseases, Gothenburg, Sweden.
| | - Ewa Johansson
- Institute of Biomedicine, Department of infectious diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Microbiology, Gothenburg, Sweden.
| | - Marita Gilljam
- Region Västra Götaland, Sahlgrenska University Hospital, Respiratory Medicine, Department of Internal Medicine and Clinical Nutrition, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg CF Centre, Adult Clinic, Gothenburg, Sweden.
| | - Bodil Jönsson
- Institute of Biomedicine, Department of infectious diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Microbiology, Gothenburg, Sweden.
| |
Collapse
|
4
|
Lin TL, Kuo YL, Lai JH, Lu CC, Yuan CT, Hsu CY, Yan BS, Wu LSH, Wu TS, Wang JY, Yu CJ, Lai HC, Shu JC, Shu CC. Gut microbiota dysbiosis-related susceptibility to nontuberculous mycobacterial lung disease. Gut Microbes 2024; 16:2361490. [PMID: 38860456 PMCID: PMC11174134 DOI: 10.1080/19490976.2024.2361490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 05/24/2024] [Indexed: 06/12/2024] Open
Abstract
The role of gut microbiota in host defense against nontuberculous mycobacterial lung disease (NTM-LD) was poorly understood. Here, we showed significant gut microbiota dysbiosis in patients with NTM-LD. Reduced abundance of Prevotella copri was significantly associated with NTM-LD and its disease severity. Compromised TLR2 activation activity in feces and plasma in the NTM-LD patients was highlighted. In the antibiotics-treated mice as a study model, gut microbiota dysbiosis with reduction of TLR2 activation activity in feces, sera, and lung tissue occurred. Transcriptomic analysis demonstrated immunocompromised in lung which were closely associated with increased NTM-LD susceptibility. Oral administration of P. copri or its capsular polysaccharides enhanced TLR2 signaling, restored immune response, and ameliorated NTM-LD susceptibility. Our data highlighted the association of gut microbiota dysbiosis, systematically compromised immunity and NTM-LD development. TLR2 activation by P. copri or its capsular polysaccharides might help prevent NTM-LD.
Collapse
Affiliation(s)
- Tzu-Lung Lin
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Microbiota Research Center and Emerging Viral Infections Research Center, Chang Gung University, Taoyuan, Taiwan
- REVIVEBIO CO, Taipei city, Taiwan
| | - Yen-Liang Kuo
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, Fu Jen Catholic University Hospital, New Taipei City, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Juo-Hsin Lai
- Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Chen Lu
- REVIVEBIO CO, Taipei city, Taiwan
- Department of Respiratory Therapy, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chang-Tsu Yuan
- Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pathology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chi-Yu Hsu
- Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Bo-Shiun Yan
- Institute of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Lawrence Shih-Hsin Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City, Taiwan
| | - Ting-Shu Wu
- Division of Infectious Diseases, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Jann-Yuan Wang
- Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chong-Jen Yu
- Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Hsin-Chih Lai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Microbiota Research Center and Emerging Viral Infections Research Center, Chang Gung University, Taoyuan, Taiwan
- REVIVEBIO CO, Taipei city, Taiwan
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jwu-Ching Shu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chin-Chung Shu
- Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
5
|
Matsuyama M, Matsumura S, Nonaka M, Nakajima M, Sakai C, Arai N, Ueda K, Hizawa N. Pathophysiology of pulmonary nontuberculous mycobacterial (NTM) disease. Respir Investig 2023; 61:135-148. [PMID: 36640546 DOI: 10.1016/j.resinv.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/25/2022] [Accepted: 12/08/2022] [Indexed: 01/15/2023]
Abstract
In recent years, the incidence and prevalence of pulmonary nontuberculous mycobacterial (NTM) disease have increased worldwide. Although the reasons for this increase are unclear, dealing with this disease is essential. Pulmonary NTM disease is a chronic pulmonary infection caused by NTM bacteria, which are ubiquitous in various environments. In Japan, Mycobacterium avium-intracellulare complex (MAC) accounts for approximately 90% of the causative organisms of pulmonary NTM disease, which is also called pulmonary MAC disease or pulmonary MAI disease. It is important to elucidate the pathophysiology of this disease, which occurs frequently in postmenopausal women despite the absence of obvious immunodeficiency. The pathophysiology of this disease has not been fully elucidated; however, it can largely be divided into bacterial (environmental) and host-side problems. The host factors can be further divided into immune and airway problems. The authors suggest that the triangular relationship between bacteria, immunity, and the airway is important in the pathophysiology of this disease. The latest findings on the pathophysiology of pulmonary NTM disease are reviewed.
Collapse
Affiliation(s)
- Masashi Matsuyama
- Department of Respiratory Medicine, Institute of Medicine, University of Tsukuba, Japan.
| | - Sosuke Matsumura
- Department of Respiratory Medicine, Institute of Medicine, University of Tsukuba, Japan
| | - Mizu Nonaka
- Department of Respiratory Medicine, Institute of Medicine, University of Tsukuba, Japan
| | - Masayuki Nakajima
- Department of Respiratory Medicine, Institute of Medicine, University of Tsukuba, Japan
| | - Chio Sakai
- Department of Respiratory Medicine, Institute of Medicine, University of Tsukuba, Japan
| | - Naoki Arai
- Department of Respiratory Medicine, Institute of Medicine, University of Tsukuba, Japan
| | - Kodai Ueda
- Department of Respiratory Medicine, Institute of Medicine, University of Tsukuba, Japan
| | - Nobuyuki Hizawa
- Department of Respiratory Medicine, Institute of Medicine, University of Tsukuba, Japan
| |
Collapse
|
6
|
Rais M, Abdelaal H, Reese VA, Ferede D, Larsen SE, Pecor T, Erasmus JH, Archer J, Khandhar AP, Cooper SK, Podell BK, Reed SG, Coler RN, Baldwin SL. Immunogenicity and protection against Mycobacterium avium with a heterologous RNA prime and protein boost vaccine regimen. Tuberculosis (Edinb) 2023; 138:102302. [PMID: 36586154 PMCID: PMC10361416 DOI: 10.1016/j.tube.2022.102302] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Prophylactic efficacy of two different delivery platforms for vaccination against Mycobacterium avium (M. avium) were tested in this study; a subunit and an RNA-based vaccine. The vaccine antigen, ID91, includes four mycobacterial antigens: Rv3619, Rv2389, Rv3478, and Rv1886. We have shown that ID91+GLA-SE is effective against a clinical NTM isolate, M. avium 2-151 smt. Here, we extend these results and show that a heterologous prime/boost strategy with a repRNA-ID91 (replicon RNA) followed by protein ID91+GLA-SE boost is superior to the subunit protein vaccine given as a homologous prime/boost regimen. The repRNA-ID91/ID91+GLA-SE heterologous regimen elicited a higher polyfunctional CD4+ TH1 immune response when compared to the homologous protein prime/boost regimen. More significantly, among all the vaccine regimens tested only repRNA-ID91/ID91+GLA-SE induced IFN-γ and TNF-secreting CD8+ T cells. Furthermore, the repRNA-ID91/ID91+GLA-SE vaccine strategy elicited high systemic proinflammatory cytokine responses and induced strong ID91 and an Ag85B-specific humoral antibody response a pre- and post-challenge with M. avium 2-151 smt. Finally, while all prophylactic prime/boost vaccine regimens elicited a degree of protection in beige mice, the heterologous repRNA-ID91/ID91+GLA-SE vaccine regimen provided greater pulmonary protection than the homologous protein prime/boost regimen. These data indicate that a prophylactic heterologous repRNA-ID91/ID91+GLA-SE vaccine regimen augments immunogenicity and confers protection against M. avium.
Collapse
Affiliation(s)
- Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Hazem Abdelaal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Valerie A Reese
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Debora Ferede
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Sasha E Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Tiffany Pecor
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | | | | | | | - Sarah K Cooper
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Brendan K Podell
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | | | - Rhea N Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA; Department of Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Susan L Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA.
| |
Collapse
|
7
|
Jung BG, Dean K, Wadle C, Samten B, Tripathi D, Wallace RJ, Brown-Elliott BA, Tucker T, Idell S, Philley JV, Vankayalapati R. Decreased Interleukin-1 Family Cytokine Production in Patients with Nontuberculous Mycobacterial Lung Disease. Microbiol Spectr 2022; 10:e0311022. [PMID: 36255321 PMCID: PMC9769609 DOI: 10.1128/spectrum.03110-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/01/2022] [Indexed: 01/09/2023] Open
Abstract
Nontuberculous mycobacteria (NTM) cause pulmonary disease in individuals without obvious immunodeficiency. This study was initiated to gain insight into the immunological factors that predispose persons to NTM pulmonary disease (NTMPD). Blood was obtained from 15 pairs of NTMPD patients and their healthy household contacts. Peripheral blood mononuclear cells (PBMCs) were stimulated with the Mycobacterium avium complex (MAC). A total of 34 cytokines and chemokines were evaluated in plasma and PBMC culture supernatants using multiplex immunoassays, and gene expression in the PBMCs was determined using real-time PCR. PBMCs from NTMPD patients produced significantly less interleukin-1β (IL-1β), IL-18, IL-1α, and IL-10 than PBMCs from their healthy household contacts in response to MAC. Although plasma RANTES levels were high in NTMPD patients, they had no effect on IL-1β production by macrophages infected with MAC. Toll-like receptor 2 (TLR2) and TWIK2 (a two-pore domain K+ channel) were impaired in response to MAC in PBMCs of NTMPD patients. A TLR2 inhibitor decreased all four cytokines, whereas a two-pore domain K+ channel inhibitor decreased the production of IL-1β, IL-18, and IL-1α, but not IL-10, by MAC-stimulated PBMCs and monocytes. The ratio of monocytes was reduced in whole blood of NTMPD patients compared with that of healthy household contacts. A reduced monocyte ratio might contribute to the attenuated production of IL-1 family cytokines by PBMCs of NTMPD patients in response to MAC stimulations. Collectively, our findings suggest that the attenuated IL-1 response may increase susceptibility to NTM pulmonary infection through multiple factors, including impaired expression of the TLR2 and TWIK2 and reduced monocyte ratio. IMPORTANCE Upon MAC stimulation, the production of IL-1 family cytokines and IL-10 by PBMCs of NTMPD patients was attenuated compared with that of healthy household contacts. Upon MAC stimulation, the expression of TLR2 and TWIK2 (one of the two-pore domain K+ channels) was attenuated in PBMCs of NTMPD patients compared with that of healthy household contacts. The production of IL-1 family cytokines by MAC-stimulated PBMCs and MAC-infected monocytes of healthy donors was reduced by a TLR2 inhibitor and two-pore domain K+ channel inhibitor. The ratio of monocytes was reduced in whole blood of NTMPD patients compared with that of healthy household contacts. Collectively, our data suggest that defects in the expression of TLR2 and TWIK2 in human PBMCs or monocytes and reduced monocyte ratio are involved in the reduced production of IL-1 family cytokines, and it may increase susceptibility to NTM pulmonary infection.
Collapse
Affiliation(s)
- Bock-Gie Jung
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Kristin Dean
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Carly Wadle
- Department of Medicine, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Buka Samten
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Deepak Tripathi
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Richard J. Wallace
- Department of Microbiology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Barbara A. Brown-Elliott
- Department of Microbiology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Torry Tucker
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
- The Texas Lung Injury Institute, Tyler, Texas, USA
| | - Julie V. Philley
- Department of Medicine, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Ramakrishna Vankayalapati
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| |
Collapse
|
8
|
Lee JM, Park J, Reed SG, Coler RN, Hong JJ, Kim LH, Lee W, Kwon KW, Shin SJ. Vaccination inducing durable and robust antigen-specific Th1/Th17 immune responses contributes to prophylactic protection against Mycobacterium avium infection but is ineffective as an adjunct to antibiotic treatment in chronic disease. Virulence 2022; 13:808-832. [PMID: 35499090 PMCID: PMC9067471 DOI: 10.1080/21505594.2022.2068489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/01/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
Mycobacterium avium complex (MAC) causing pulmonary disease in humanshas emerged worldwide. Thus, effective strategies simultaneously aiming to prevent MAC infection and accelerate therapeutic efficacy are required. To this end, subunit vaccine-induced protection against a well-defined virulent Mycobacterium avium (Mav) isolate was assessed as a preventative and therapeutic modality in murine models. Mav-derived culture filtrate antigen (CFA) was used as a vaccine antigen with glucopyranosyl lipid A stable emulsion (GLA-SE) or GLA-SE plus cyclic-di-GMP (GLA-SE/CDG), and we compared the immunogenicities, protective efficacies and immune correlates. Interestingly, CFA+GLA-SE/CDG immunization induced greater CFA-specific Th1/Th17 responses in both the lung and spleen than among the tested groups. Consequently, protective efficacy was optimally achieved with CFA+GLA-SE/CDG by significantly reducing bacterial loads along with long-lasting maintenance of antigen-specific Th1/Th17 cytokine-producing multifunctional T cell responses and relevant cytokine productions. Thus, we employed this subunit vaccine as an adjunct to antibiotic treatment. However, this vaccine was ineffective in further reducing bacterial loads. Collectively, our study demonstrates that strong Mav CFA-specific Th1/Th17 responses are critical for preventative protection against Mav infection but may be ineffective or even detrimental in an established and progressive chronic disease, indicating that different approaches to combating Mav infection are necessary according to vaccination purposes.
Collapse
Affiliation(s)
- Ju Mi Lee
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Jiyun Park
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Rhea N Coler
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Jung Joo Hong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Lee-Han Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
9
|
Jung BG, Samten B, Dean K, Wallace RJ, Brown-Elliott BA, Tucker T, Idell S, Philley JV, Vankayalapati R. Early IL-17A production helps establish Mycobacterium intracellulare infection in mice. PLoS Pathog 2022; 18:e1010454. [PMID: 35363832 PMCID: PMC9007361 DOI: 10.1371/journal.ppat.1010454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 04/13/2022] [Accepted: 03/17/2022] [Indexed: 12/24/2022] Open
Abstract
Nontuberculous mycobacteria (NTM) infection is common in patients with structural lung damage. To address how NTM infection is established and causes lung damage, we established an NTM mouse model by intranasal inoculation of clinical isolates of M. intracellulare. During the 39-week course of infection, the bacteria persistently grew in the lung and caused progressive granulomatous and fibrotic lung damage with mortality exceeding 50%. Lung neutrophils were significantly increased at 1 week postinfection, reduced at 2 weeks postinfection and increased again at 39 weeks postinfection. IL-17A was increased in the lungs at 1-2 weeks of infection and reduced at 3 weeks postinfection. Depletion of neutrophils during early (0-2 weeks) and late (32-34 weeks) infection had no effect on mortality or lung damage in chronically infected mice. However, neutralization of IL-17A during early infection significantly reduced bacterial burden, fibrotic lung damage, and mortality in chronically infected mice. Since it is known that IL-17A regulates matrix metalloproteinases (MMPs) and that MMPs contribute to the pathogenesis of pulmonary fibrosis, we determined the levels of MMPs in the lungs of M. intracellulare-infected mice. Interestingly, MMP-3 was significantly reduced by anti-IL-17A neutralizing antibody. Moreover, in vitro data showed that exogenous IL-17A exaggerated the production of MMP-3 by lung epithelial cells upon M. intracellulare infection. Collectively, our findings suggest that early IL-17A production precedes and promotes organized pulmonary M. intracellulare infection in mice, at least in part through MMP-3 production.
Collapse
Affiliation(s)
- Bock-Gie Jung
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| | - Buka Samten
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| | - Kristin Dean
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| | - Richard J. Wallace
- Department of Microbiology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| | - Barbara A. Brown-Elliott
- Department of Microbiology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| | - Torry Tucker
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
- The Texas Lung Injury Institute, Tyler, Texas, United States of America
| | - Julie V. Philley
- Department of Medicine, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| | - Ramakrishna Vankayalapati
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| |
Collapse
|
10
|
Park EJ, Silwal P, Jo EK. Host-Pathogen Interactions Operative during Mycobacteroides abscessus Infection. Immune Netw 2022; 21:e40. [PMID: 35036027 PMCID: PMC8733189 DOI: 10.4110/in.2021.21.e40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/01/2021] [Accepted: 12/09/2021] [Indexed: 12/01/2022] Open
Abstract
Mycobacteroides abscessus (previously Mycobacterium abscessus; Mabc), one of rapidly growing nontuberculous mycobacteria (NTM), is an important pathogen of NTM pulmonary diseases (NTM-PDs) in both immunocompetent and immunocompromised individuals. Mabc infection is chronic and often challenging to treat due to drug resistance, motivating the development of new therapeutics. Despite this, there is a lack of understanding of the relationship between Mabc and the immune system. This review highlights recent progress in the molecular architecture of Mabc and host interactions. We discuss several microbial components that take advantage of host immune defenses, host defense pathways that can overcome Mabc pathogenesis, and how host-pathogen interactions determine the outcomes of Mabc infection. Understanding the molecular mechanisms underlying host-pathogen interactions during Mabc infection will enable the identification of biomarkers and/or drugs to control immune pathogenesis and protect against NTM infection.
Collapse
Affiliation(s)
- Eun-Jin Park
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon 35015, Korea.,Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - Prashanta Silwal
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon 35015, Korea.,Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon 35015, Korea.,Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon 35015, Korea
| |
Collapse
|
11
|
Kumar K, Kon OM. Personalised Medicine for Tuberculosis and Non-Tuberculous Mycobacterial Pulmonary Disease. Microorganisms 2021; 9:2220. [PMID: 34835346 PMCID: PMC8624359 DOI: 10.3390/microorganisms9112220] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 12/29/2022] Open
Abstract
Personalised medicine, in which clinical management is individualised to the genotypic and phenotypic data of patients, offers a promising means by which to enhance outcomes in the management of mycobacterial pulmonary infections. In this review, we provide an overview of how personalised medicine approaches may be utilised to identify patients at risk of developing tuberculosis (TB) or non-tuberculous mycobacterial pulmonary disease (NTM-PD), diagnose these conditions and guide effective treatment strategies. Despite recent technological and therapeutic advances, TB and NTM-PD remain challenging conditions to diagnose and treat. Studies have identified a range of genetic and immune factors that predispose patients to pulmonary mycobacterial infections. Molecular tests such as nucleic acid amplification assays and next generation sequencing provide a rapid means by which to identify mycobacterial isolates and their antibiotic resistance profiles, thus guiding selection of appropriate antimicrobials. Host-directed therapies and therapeutic drug monitoring offer ways of tailoring management to the clinical needs of patients at an individualised level. Biomarkers may hold promise in differentiating between latent and active TB, as well as in predicting mycobacterial disease progression and response to treatment.
Collapse
Affiliation(s)
- Kartik Kumar
- National Heart and Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK;
- Department of Respiratory Medicine, St Mary’s Hospital, Imperial College Healthcare NHS Trust, Praed Street, London W2 1NY, UK
| | - Onn Min Kon
- National Heart and Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK;
- Department of Respiratory Medicine, St Mary’s Hospital, Imperial College Healthcare NHS Trust, Praed Street, London W2 1NY, UK
| |
Collapse
|
12
|
Shu CC, Wu MF, Pan SW, Wu TS, Lai HC, Lin MC. Host immune response against environmental nontuberculous mycobacteria and the risk populations of nontuberculous mycobacterial lung disease. J Formos Med Assoc 2020; 119 Suppl 1:S13-S22. [PMID: 32451216 DOI: 10.1016/j.jfma.2020.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/21/2020] [Accepted: 05/04/2020] [Indexed: 12/16/2022] Open
Abstract
Nontuberculous mycobacterial lung disease (NTM-LD) prevalence has been increasing over the recent decades. Numerous host factors are associated with NTM-LD development, including susceptible phenotypes such as ciliary defect and lung structural change, pulmonary clearance defect with poor clearance of secretions, and immune suppression. Specifically, regarding the susceptible host phenotypes without clear pathogenesis, a slender body, pectus excavatum, and postmenopausal female status are common. Also, decreased host immunity to NTM, especially T helper 1 cell responses is frequently observed. Even so, the underlying mechanisms remain unclear and relevant large-scale studies are lacking. Infections due to host genetics associated defects are mostly untreatable but rare in Asia, particularly Taiwan. Nevertheless, some risks for NTM-LD are controllable over disease progression. We suggest that clinicians first manage host factors and deal with the controllable characteristics of NTM-LD, followed by optimizing anti-NTM treatment. Further researches focusing on NTM-LD pathogenesis, especially the host-NTM interaction may advance understanding the nature of the disease and develop efficient therapeutic regimens.
Collapse
Affiliation(s)
- Chin-Chung Shu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Fang Wu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan; Institute of Statistical Sciences, Academia Sinica, Taipei, Taiwan
| | - Sheng-Wei Pan
- Department of Chest Medicine, Taipei Veterans General Hospital, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ting-Shu Wu
- Division of Infectious Diseases, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fuxing St., Guishan Dist., Taoyuan 33305, Taiwan.
| | - Hsin-Chih Lai
- Central Research Laboratory, Xiamen Chang Gung Hospital, Xiamen, Fujian, China; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Meng-Chih Lin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| |
Collapse
|
13
|
Han SA, Ko Y, Shin SJ, Jhun BW. Characteristics of Circulating CD4 + T Cell Subsets in Patients with Mycobacterium avium Complex Pulmonary Disease. J Clin Med 2020; 9:jcm9051331. [PMID: 32375214 PMCID: PMC7290757 DOI: 10.3390/jcm9051331] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
Although prevalence of Mycobacterium avium complex pulmonary disease (MAC-PD) is increasing, limited data are available regarding vulnerability to Mycobacterium avium complex (MAC) infections. To understand the pathobiology of interaction between MAC and host-immunity, it is important to understand the characteristics for circulating T cells in terms of the immunological phenotype and functional correlates in MAC-PD. We aimed to characterize immunophenotype, cytokine profile, and immune inhibitory receptors of circulating CD4+ T cells in MAC-PD patients. We enrolled 71 MAC-PD and 20 control individuals. Flow cytometric analysis was performed to determine T cell subsets and immune checkpoint markers. Ex vivo cytokine productions in response to MAC were determined using enzyme-linked immunosorbent assay. The frequencies of CD4+ T cells and CD4+IL-17+ T cells decreased, while CD4+IL-4+ T cells and CD4+CD25+Foxp3+ T cells increased in peripheral blood mononuclear cells (PBMCs) of MAC-PD individuals upon MAC stimulation compared with those cells in healthy donor-PBMCs. Additionally, we found increased PD-1, CTLA-4, and TIM-3-expressing T cells in MAC- PD individuals in response to MAC-stimulation, indicating that suppressed T cell-mediated response is associated with the susceptibility to MAC infection. These results may help to explain impaired T cell-mediated responses and pave the way for better strategies to achieve protective immunity against MAC infection.
Collapse
Affiliation(s)
- Sun Ae Han
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
| | - Yousang Ko
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul 05355, Korea;
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
- Correspondence: (S.J.S.); (B.W.J.)
| | - Byung Woo Jhun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
- Correspondence: (S.J.S.); (B.W.J.)
| |
Collapse
|
14
|
|
15
|
Ratnatunga CN, Lutzky VP, Kupz A, Doolan DL, Reid DW, Field M, Bell SC, Thomson RM, Miles JJ. The Rise of Non-Tuberculosis Mycobacterial Lung Disease. Front Immunol 2020; 11:303. [PMID: 32194556 PMCID: PMC7062685 DOI: 10.3389/fimmu.2020.00303] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 02/06/2020] [Indexed: 12/21/2022] Open
Abstract
The incidence and number of deaths from non-tuberculous mycobacterial (NTM) disease have been steadily increasing globally. These lesser known “cousins” of Mycobacterium tuberculosis (TB) were once thought to be harmless environmental saprophytics and only dangerous to individuals with defective lung structure or the immunosuppressed. However, NTM are now commonly infecting seemingly immune competent children and adults at increasing rates through pulmonary infection. This is of concern as the pathology of NTM is difficult to treat. Indeed, NTM have become extremely antibiotic resistant, and now have been found to be internationally dispersed through person-to-person contact. The reasons behind this NTM increase are only beginning to be elucidated. Solutions to the problem are needed given NTM disease is more common in the tropics. Importantly, 40% of the world's population live in the tropics and due to climate change, the Tropics are expanding which will increase NTM infection regions. This review catalogs the global and economic disease burden, at risk populations, treatment options, host-bacterial interaction, immune dynamics, recent developments and research priorities for NTM disease.
Collapse
Affiliation(s)
- Champa N. Ratnatunga
- The Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- *Correspondence: Champa N. Ratnatunga
| | - Viviana P. Lutzky
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Andreas Kupz
- The Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - Denise L. Doolan
- The Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - David W. Reid
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Matthew Field
- The Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
| | - Scott C. Bell
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Rachel M. Thomson
- Immunology Department, Gallipoli Medical Research Institute, Brisbane, QLD, Australia
| | - John J. Miles
- The Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
- John J. Miles
| |
Collapse
|
16
|
Wu UI, Olivier KN, Kuhns DB, Fink DL, Sampaio EP, Zelazny AM, Shallom SJ, Marciano BE, Lionakis MS, Holland SM. Patients with Idiopathic Pulmonary Nontuberculous Mycobacterial Disease Have Normal Th1/Th2 Cytokine Responses but Diminished Th17 Cytokine and Enhanced Granulocyte-Macrophage Colony-Stimulating Factor Production. Open Forum Infect Dis 2019; 6:ofz484. [PMID: 31807607 DOI: 10.1093/ofid/ofz484] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/08/2019] [Indexed: 11/13/2022] Open
Abstract
Objective Although disseminated nontuberculous mycobacterial infection is attributed to defects in the interleukin (IL)-12/interferon-γ circuit, the immunophenotype of idiopathic pulmonary nontuberculous mycobacterial (PNTM) disease is not well defined. Method We phenotyped Th1, Th2, Th17, and Treg cytokines and colony-stimulating factor production from patients with idiopathic PNTM disease. Data were compared with healthy donors, cystic fibrosis (CF), and primary ciliary dyskinesia (PCD) patients with PNTM disease. Both supernatant cytokine production and intracellular cytokines expressed by various leukocyte subpopulations following mitogen and antigen stimulation were assayed by electrochemiluminescence-based multiplex immunoassay and flow cytometry, respectively. Results Regardless of antigen or mitogen stimulation, neither intracellular nor extracellular Th1, Th2, and Treg cytokine levels differed between patients and controls. Th17 cells and IL-17A levels were lower in idiopathic PNTM patients, whereas monocyte granulocyte-macrophage colony-stimulating factor (GM-CSF) expression in response to NTM stimulation was higher compared with healthy donors. Besides, distinct cytokine responses following stimulation by Mycobacterium abscessus and Mycobacterium avium were observed consistently within each group. Conclusions The IL-12/IFN-γ circuit appeared intact in patients with idiopathic PNTM disease. However, idiopathic PNTM patients had reduced Th17 response and higher mycobacteria-induced monocyte GM-CSF expression.
Collapse
Affiliation(s)
- Un-In Wu
- Division of Infectious Diseases, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kenneth N Olivier
- Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, the National Institutes of Health, Bethesda, Maryland, USA
| | - Douglas B Kuhns
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Danielle L Fink
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Elizabeth P Sampaio
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, the National Institutes of Health, Bethesda, Maryland, USA
| | - Adrian M Zelazny
- Microbiology Service, Department of Laboratory Medicine, Clinical Center, the National Institutes of Health, Bethesda, Maryland, USA
| | - Shamira J Shallom
- Microbiology Service, Department of Laboratory Medicine, Clinical Center, the National Institutes of Health, Bethesda, Maryland, USA
| | - Beatriz E Marciano
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, the National Institutes of Health, Bethesda, Maryland, USA
| | - Michail S Lionakis
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, the National Institutes of Health, Bethesda, Maryland, USA
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, the National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
17
|
Chat VS, Kearns DG, Uppal SK, Wu JJ. Risk of atypical mycobacterial infections in psoriasis patients during IL-17 inhibitor therapy. J DERMATOL TREAT 2019; 32:495-496. [PMID: 31689137 DOI: 10.1080/09546634.2019.1687812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Vipawee S Chat
- Medical College of Georgia at Augusta University, Augusta, GA, USA
| | | | | | - Jashin J Wu
- Dermatology Research, Education Foundation, Irvine, CA, USA
| |
Collapse
|
18
|
Cowman S, van Ingen J, Griffith DE, Loebinger MR. Non-tuberculous mycobacterial pulmonary disease. Eur Respir J 2019; 54:13993003.00250-2019. [PMID: 31221809 DOI: 10.1183/13993003.00250-2019] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/31/2019] [Indexed: 02/03/2023]
Abstract
Nontuberculous mycobacterial pulmonary disease (NTM-PD) is a challenging infection which is becoming increasingly prevalent, particularly in the elderly, for reasons which are unknown. While underlying lung disease is a well-established risk factor for NTM-PD, it may also occur in apparently healthy individuals. No single common genetic or immunological defect has been identified in this group, and it is likely that multiple pathways contribute towards host susceptibility to NTM-PD which further interact with environmental and microbiological factors leading to the development of disease.The diagnosis of NTM-PD relies on the integration of clinical, radiological and microbiological results. The clinical course of NTM-PD is heterogeneous, with some patients remaining stable without the need for treatment and others developing refractory disease associated with considerable mortality and morbidity. Treatment regimens are based on the identity of the isolated species, drug sensitivity testing (for some agents) and the severity of disease. Multiple antibiotics are typically required for prolonged periods of time and treatment is frequently poorly tolerated. Surgery may be beneficial in selected cases. In some circumstances cure may not be attainable and there is a pressing need for better regimens to treat refractory and drug-resistant NTM-PD.This review summarises current knowledge on the epidemiology, aetiology and diagnosis of NTM-PD and discusses the treatment of two of the most clinically significant species, the M. avium and M. abscessus complexes, with a focus on refractory disease and novel therapies.
Collapse
Affiliation(s)
- Steven Cowman
- Host Defence Unit, Royal Brompton Hospital, London, UK.,Imperial College, London, UK
| | - Jakko van Ingen
- Dept of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - David E Griffith
- Dept of Medicine, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Michael R Loebinger
- Host Defence Unit, Royal Brompton Hospital, London, UK .,Imperial College, London, UK
| |
Collapse
|
19
|
Cowman SA, Jacob J, Hansell DM, Kelleher P, Wilson R, Cookson WOC, Moffatt MF, Loebinger MR. Whole-Blood Gene Expression in Pulmonary Nontuberculous Mycobacterial Infection. Am J Respir Cell Mol Biol 2019; 58:510-518. [PMID: 29206475 DOI: 10.1165/rcmb.2017-0230oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The factors predisposing toward the development of pulmonary nontuberculous mycobacterial (pNTM) disease and influencing disease progression remain unclear. Impaired immune responses have been reported in individuals with pNTM disease, but data are limited and inconsistent. In this study, we sought to use gene expression profiling to examine the host response to pNTM disease. Microarray analysis of whole-blood gene expression was performed on 25 subjects with pNTM disease and 27 uninfected control subjects with respiratory disease. Gene expression results were compared with phenotypic variables and survival data. Compared with uninfected control subjects, pNTM disease was associated with downregulation of 213 transcripts enriched for terms related to T cell signaling, including IFNG. Reduced IFNG expression was associated with more severe computed tomography changes and impaired lung function. Mortality was associated with the expression of transcripts related to the innate immune response and inflammation, whereas transcripts related to T and B cell function were associated with improved survival. These findings suggest that pNTM disease is associated with an aberrant immune response, which may reflect an underlying propensity to infection or result from NTM infection itself. There were important differences in the immune response associated with survival and mortality in pNTM disease.
Collapse
Affiliation(s)
- Steven A Cowman
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,2 Host Defence Unit and
| | - Joseph Jacob
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,3 Department of Radiology, Royal Brompton Hospital, London, United Kingdom
| | - David M Hansell
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,3 Department of Radiology, Royal Brompton Hospital, London, United Kingdom
| | - Peter Kelleher
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,2 Host Defence Unit and
| | - Robert Wilson
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,2 Host Defence Unit and
| | - William O C Cookson
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Miriam F Moffatt
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Michael R Loebinger
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,2 Host Defence Unit and
| |
Collapse
|
20
|
Shu CC, Wu LSH, Wu MF, Lai HC, Wang PH, Cheng SL, Wang JY, Yu CJ. Mono- and poly-functional T cells in nontuberculous mycobacteria lung disease patients: Implications in analyzing risk of disease progression. Cytokine 2019; 120:176-185. [PMID: 31085455 DOI: 10.1016/j.cyto.2019.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 12/21/2022]
Abstract
AT A GLANCE The diagnosis and progression of nontuberculous mycobacteria lung disease (NTN-LD) are important for clinical judgement but cannot easily be predicted. The immunological response of mono- and poly-functional T cells, a representative of host reactivity to NTM, could be a surrogate biomarker for disease and progression prediction. BACKGROUND Mycobacterium avium complex (MAC) and M. abscessus (MAB) induced lung disease (LD) have become a clinical concern. Predicting clinical disease relevance and progression is important, but suitable biomarkers are lacking. The host immune response of mono- and poly-functional T cells might aid in clinical judgement. METHODS We enrolled 140 participants, including 42 MAC-LD, 25 MAB-LD, 31 MAC airway colonization (MAC-Co), 15 MAB-Co patients, and 27 healthy controls. Their blood mono- and poly-functional T cells were measured and analyzed after in-vitro stimulation. RESULTS Patients with MAC-LD generally had lower total IFN-γ+, total TNF-α+ and triple-positive T cells but higher mono-IL-2+ expression than the controls and MAC-Co group. The MAB-LD group had lower total IL-2 and triple positive cells than the controls and colonization group. Multivariate analysis revealed that body mass index (BMI), mono-IL2+ CD4+ and triple positive-CD8+ cells (PMA stimulation) significantly predicted MAC-LD from the controls. By contrast, male gender and triple positive-CD4+ cells predicted MAC-LD from colonization. On the other hand, the triple positive-CD4+ cells (PMA stimulation) alone or together with the mock/MAB ratio of IL-2+/TNF-α+ CD4 cells could predict MAB-LD in the MAB-Co group or the controls. Among MAC/MAB-LD patients without anti-mycobacterial treatment, MAC-specific mono-IFN-γ+ CD4+ cells and PMA-induced triple positive-CD4+ cells were correlated with progression, with an area under the ROC curve of 0.875. CONCLUSIONS The patients with MAC/MAB-LD had attenuated poly-functional T cells. The triple-positive CD4+ cells could be useful in diagnosing disease from colonization. MAC-specific mono-IFN-γ+ CD4+ cells and triple positive-CD4+ might predict radiographic progression, which could be useful in making treatment decisions.
Collapse
Affiliation(s)
- Chin-Chung Shu
- Department of Internal Medicine, National Taiwan University Hospital, Taiwan
| | | | - Ming-Fang Wu
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan; Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Chih Lai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan; Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan; Department of Laboratory Medicine, Xiamen Chang Gung Memorial Hospital, Xiamen, Fujian, China
| | - Ping-Huai Wang
- Division of Chest Medicine, Department of Internal Medicine, Far Eastern Memorial Hospital, Taiwan; Department of Nursing, Oriental Institute of Technology, Taiwan.
| | - Shih-Lung Cheng
- Division of Chest Medicine, Department of Internal Medicine, Far Eastern Memorial Hospital, Taiwan; Department of Chemical Engineering and Materials Science, Yuan-Ze University, Taiwan
| | - Jann-Yuan Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taiwan
| | - Chong-Jen Yu
- Department of Internal Medicine, National Taiwan University Hospital, Taiwan
| |
Collapse
|
21
|
Huiberts A, Zweijpfenning SMH, Pennings LJ, Boeree MJ, van Ingen J, Magis-Escurra C, Hoefsloot W. Outcomes of hypertonic saline inhalation as a treatment modality in nontuberculous mycobacterial pulmonary disease. Eur Respir J 2019; 54:13993003.02143-2018. [PMID: 31000680 DOI: 10.1183/13993003.02143-2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/07/2019] [Indexed: 01/23/2023]
Affiliation(s)
- Anne Huiberts
- Dept of Pulmonary Diseases, Radboud Center for Infectious diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sanne M H Zweijpfenning
- Dept of Pulmonary Diseases, Radboud Center for Infectious diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lian J Pennings
- Dept of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin J Boeree
- Dept of Pulmonary Diseases, Radboud Center for Infectious diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jakko van Ingen
- Dept of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cecile Magis-Escurra
- Dept of Pulmonary Diseases, Radboud Center for Infectious diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wouter Hoefsloot
- Dept of Pulmonary Diseases, Radboud Center for Infectious diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
22
|
Bamba Y, Moro H, Aoki N, Koizumi T, Ohshima Y, Watanabe S, Sakagami T, Koya T, Takada T, Kikuchi T. Multiplex cytokine analysis in Mycobacterium avium complex lung disease: relationship between CXCL10 and poor prognostic factors. BMC Infect Dis 2019; 19:263. [PMID: 30885152 PMCID: PMC6423821 DOI: 10.1186/s12879-019-3888-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/07/2019] [Indexed: 11/26/2022] Open
Abstract
Background Mycobacterium avium complex lung disease (MAC-LD) can deteriorate rapidly to become fatal. Reported poor prognostic factors include radiographic findings, undernutrition, anemia and high inflammation test values. However, the association of these prognostic factors with the pathophysiology of the disease remains unknown. We aimed to clarify the pathophysiology of MAC-LD and develop a new biomarker that reflects the immune response to the disease. Methods We performed the cytokine panel analyses of serum from patients with MAC-LD and compared each cytokine level with clinically negative prognostic factors (radiographic disease type, body mass index, albumin, C-reactive protein and hemoglobin) and high-resolution CT scores. Results We analyzed 27 patients with MAC-LD, 6 with the fibrocavitary form and 21 with the nodular bronchiectatic form on high-resolution CT. Serum CXC motif ligand 10 (CXCL10) concentration was significantly elevated in patients with the fibrocavitary form (p = 0.008). CXCL10 levels correlated with body mass index (r = − 0.60, p = 0.0008), serum albumin concentration (r = − 0.45, p = 0.016) and high-resolution CT scores (r = 0.61, p = 0.0006). Among 14 patients initially untreated, antibiotic therapy was initiated for five during the study period. CXCL10 concentration was significantly higher in these patients (p = 0.046), and receiver operating characteristic analysis for CXCL10 concentration on treatment initiation produced an area under the curve of 0.844, with a sensitivity of 100%, specificity of 66.7%, and cut-off value of 366.5 pg/mL. Conclusion We revealed cytokine profiles in patients with MAC-LD. Serum CXCL10 levels probably reflect the severity of MAC-LD. Our findings suggest that CXCL10 concentration may be a promising biomarker for managing treatment for patients with MAC disease of the lung. Electronic supplementary material The online version of this article (10.1186/s12879-019-3888-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuuki Bamba
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757 Ichibancho, Asahimachi-dori, Chuo Ward, Niigata City, 951-8510, Japan
| | - Hiroshi Moro
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757 Ichibancho, Asahimachi-dori, Chuo Ward, Niigata City, 951-8510, Japan.
| | - Nobumasa Aoki
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757 Ichibancho, Asahimachi-dori, Chuo Ward, Niigata City, 951-8510, Japan
| | - Takeshi Koizumi
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757 Ichibancho, Asahimachi-dori, Chuo Ward, Niigata City, 951-8510, Japan
| | - Yasuyoshi Ohshima
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757 Ichibancho, Asahimachi-dori, Chuo Ward, Niigata City, 951-8510, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757 Ichibancho, Asahimachi-dori, Chuo Ward, Niigata City, 951-8510, Japan
| | - Takuro Sakagami
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757 Ichibancho, Asahimachi-dori, Chuo Ward, Niigata City, 951-8510, Japan
| | - Toshiyuki Koya
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757 Ichibancho, Asahimachi-dori, Chuo Ward, Niigata City, 951-8510, Japan
| | - Toshinori Takada
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757 Ichibancho, Asahimachi-dori, Chuo Ward, Niigata City, 951-8510, Japan
| | - Toshiaki Kikuchi
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757 Ichibancho, Asahimachi-dori, Chuo Ward, Niigata City, 951-8510, Japan
| |
Collapse
|
23
|
Vinnard C, Mezochow A, Oakland H, Klingsberg R, Hansen-Flaschen J, Hamilton K. Assessing Response to Therapy for Nontuberculous Mycobacterial Lung Disease: Quo Vadis? Front Microbiol 2018; 9:2813. [PMID: 30524407 PMCID: PMC6256187 DOI: 10.3389/fmicb.2018.02813] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 11/01/2018] [Indexed: 12/26/2022] Open
Abstract
Assessing progression of disease or response to treatment remains a major challenge in the clinical management of nontuberculous mycobacterial (NTM) infections of the lungs. Serial assessments of validated measures of treatment response address whether the current therapeutic approach is on track toward clinical cure, which remains a fundamental question for clinicians and patients during the course of NTM disease treatment. The 2015 NTM Research Consortium Workshop, which included a patient advisory panel, identified treatment response biomarkers as a priority area for investigation. Limited progress in addressing this challenge also hampers drug development efforts. The Biomarker Qualification Program at the FDA supports the use of a validated treatment response biomarker across multiple drug development programs. Current approaches in clinical practice include microbiologic and radiographic monitoring, along with symptomatic and quality-of-life assessments. Blood-based monitoring, including assessments of humoral and cell-mediated NTM-driven immune responses, remain under investigation. Alignment of data collection schemes in prospective multicenter studies, including the support of biosample repositories, will support identification of treatment response biomarkers under standard-of-care and investigational therapeutic strategies. In this review, we outline the role of treatment monitoring biomarkers in both clinical practice and drug development frameworks.
Collapse
Affiliation(s)
- Christopher Vinnard
- Public Health Research Institute, New Jersey Medical School, Newark, NJ, United States
| | - Alyssa Mezochow
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hannah Oakland
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Ross Klingsberg
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - John Hansen-Flaschen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Keith Hamilton
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
24
|
Addy C, Doran G, Jones AL, Wright G, Caskey S, Downey DG. Microscopic polyangiitis secondary to Mycobacterium abscessus in a patient with bronchiectasis: a case report. BMC Pulm Med 2018; 18:170. [PMID: 30453935 PMCID: PMC6245610 DOI: 10.1186/s12890-018-0732-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 10/29/2018] [Indexed: 11/30/2022] Open
Abstract
Background Non-Tuberculous Mycobacterial–pulmonary disease (NTM-PD) is increasing in incidence and prevalence. Mycobacterium abscessus (M.abscessus) is a rapid growing multi-resistant NTM associated with severe NTM-PD requiring prolonged antibiotic therapy. Complications of therapy are common but reports on direct complications of active NTM-PD are rare. Vasculitis has been described as a rare complication of NTM-PD, most often in individuals with inherited immune defects. This case is the first to describe an ANCA positive vasculitide (Microscopic Polyangiitis) secondary to M.abscessus pulmonary disease. Case presentation A 70 year old female with bronchiectasis underwent a clinical decline associated with the growth of M.abscessus and was diagnosed with NTM-PD. Before treatment could be initiated she developed small joint arthralgia and a glove and stocking axonal loss sensorimotor neuropathy. Positive Perinuclear Anti-Neutrophil Cytoplasmic Antibodies (P-ANCA) and Myeloperoxidase-ANCA (MPO-ANCA) titres led to a diagnosis of microscopic polyangiitis. Further investigation revealed reduced interferon-gamma production but no other significant immune dysfunction. Dual treatment with immunosuppressive therapy (Corticosteroids/Cyclophosphamide) for vasculitis and antimicrobial therapy for M.abscessus NTM-PD was initiated. Clinical stability was difficult to achieve with reductions in immunosuppression triggering vasculitic flares. One flare led to retinal vein occlusion with impending visual loss requiring escalation in immunosuppression to Rituximab infusions. An increase in immunosuppression led to a deterioration in NTM-PD necessitating alterations to antibiotic regimes. Adverse effects including alopecia and Achilles tendonitis have further limited antibiotic choices resulting in a strategy of pulsed intra-venous therapy to stabilise NTM-PD. Conclusions This is the first reported case of an ANCA positive vasculitis secondary to M.abscessus pulmonary disease. This rare but important complication had a significant impact on the patient adding to the complexity of an already significant disease and treatment burden. The potential role of reduced interferon-gamma production in this case highlights the importance of investigating immune function in those with mycobacterial infection and the intricate relationship between mycobacterial infection and immune dysfunction. Immune dysfunction caused by genetic defects or immunosuppressive therapy is a known risk factor for NTM-PD. Balancing immunosuppressive therapy with prolonged antimicrobial treatment is challenging and likely to become more common as the number of individuals being treated with biologics and immunosuppressive agents increases.
Collapse
Affiliation(s)
- C Addy
- Centre for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland. .,Regional Respiratory Centre, Belfast City Hospital, 51 Lisburn Road, Belfast, BT9 7AB, Northern Ireland.
| | - G Doran
- Regional Respiratory Centre, Belfast City Hospital, 51 Lisburn Road, Belfast, BT9 7AB, Northern Ireland
| | - A L Jones
- Department of Respiratory Medicine, Royal Brompton Hospitals, Sydney Street, London, SW3 6NP, England
| | - G Wright
- Department of Rheumatology, Musgrave Park Hospital, Stockmans Ln, Belfast, BT9 7JB, Northern Ireland
| | - S Caskey
- Regional Respiratory Centre, Belfast City Hospital, 51 Lisburn Road, Belfast, BT9 7AB, Northern Ireland
| | - D G Downey
- Centre for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland.,Regional Respiratory Centre, Belfast City Hospital, 51 Lisburn Road, Belfast, BT9 7AB, Northern Ireland
| |
Collapse
|
25
|
Lutzky VP, Ratnatunga CN, Smith DJ, Kupz A, Doolan DL, Reid DW, Thomson RM, Bell SC, Miles JJ. Anomalies in T Cell Function Are Associated With Individuals at Risk of Mycobacterium abscessus Complex Infection. Front Immunol 2018; 9:1319. [PMID: 29942313 PMCID: PMC6004551 DOI: 10.3389/fimmu.2018.01319] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 05/28/2018] [Indexed: 12/13/2022] Open
Abstract
The increasing global incidence and prevalence of non-tuberculous mycobacteria (NTM) infection is of growing concern. New evidence of person-to-person transmission of multidrug-resistant NTM adds to the global concern. The reason why certain individuals are at risk of NTM infections is unknown. Using high definition flow cytometry, we studied the immune profiles of two groups that are at risk of Mycobacterium abscessus complex infection and matched controls. The first group was cystic fibrosis (CF) patients and the second group was elderly individuals. CF individuals with active M. abscessus complex infection or a history of M. abscessus complex infection exhibited a unique surface T cell phenotype with a marked global deficiency in TNFα production during mitogen stimulation. Importantly, immune-based signatures were identified that appeared to predict at baseline the subset of CF individuals who were at risk of M. abscessus complex infection. In contrast, elderly individuals with M. abscessus complex infection exhibited a separate T cell phenotype underlined by the presence of exhaustion markers and dysregulation in type 1 cytokine release during mitogen stimulation. Collectively, these data suggest an association between T cell signatures and individuals at risk of M. abscessus complex infection, however, validation of these immune anomalies as robust biomarkers will require analysis on larger patient cohorts.
Collapse
Affiliation(s)
- Viviana P Lutzky
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Champa N Ratnatunga
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Centre for Biodiscovery and Molecular Development of Therapeutics, Centre for Biosecurity and Tropical Infectious Diseases, AITHM, James Cook University, Cairns, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Daniel J Smith
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Andreas Kupz
- Centre for Biodiscovery and Molecular Development of Therapeutics, Centre for Biosecurity and Tropical Infectious Diseases, AITHM, James Cook University, Cairns, QLD, Australia
| | - Denise L Doolan
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Centre for Biodiscovery and Molecular Development of Therapeutics, Centre for Biosecurity and Tropical Infectious Diseases, AITHM, James Cook University, Cairns, QLD, Australia
| | - David W Reid
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Rachel M Thomson
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia.,Gallipoli Medical Research Institute, Brisbane, QLD, Australia
| | - Scott C Bell
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - John J Miles
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Centre for Biodiscovery and Molecular Development of Therapeutics, Centre for Biosecurity and Tropical Infectious Diseases, AITHM, James Cook University, Cairns, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| |
Collapse
|
26
|
Matsuyama M, Martins AJ, Shallom S, Kamenyeva O, Kashyap A, Sampaio EP, Kabat J, Olivier KN, Zelazny AM, Tsang JS, Holland SM. Transcriptional Response of Respiratory Epithelium to Nontuberculous Mycobacteria. Am J Respir Cell Mol Biol 2018; 58:241-252. [PMID: 28915071 DOI: 10.1165/rcmb.2017-0218oc] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The incidence of pulmonary nontuberculous mycobacteria (NTM) disease is increasing, but host responses in respiratory epithelium infected with NTM are not fully understood. In this work, we aimed to identify infection-relevant gene expression signatures of NTM infection of the respiratory epithelium. We infected air-liquid interface (ALI) primary respiratory epithelial cell cultures with Mycobacterium avium subsp. avium (MAC) or Mycobacterium abscessus subsp. abscessus (MAB). We used cells from four different donors to obtain generalizable data. Differentiated respiratory epithelial cells at the ALI were infected with MAC or MAB at a multiplicity of infection of 100:1 or 1,000:1, and RNA sequencing was performed at Days 1 and 3 after infection. In response to infection, we found down-regulation of ciliary genes but upregulation of genes associated with cytokines/chemokines, such as IL-32, and cholesterol biosynthesis. Inflammatory response genes tended to be more upregulated by MAB than by MAC infection. Primary respiratory epithelial cell infection with NTM at the ALI identified ciliary function, cholesterol biosynthesis, and cytokine/chemokine production as major host responses to infection. Some of these pathways may be amenable to therapeutic manipulation.
Collapse
Affiliation(s)
| | - Andrew J Martins
- 2 Systems Genomics and Bioinformatics Unit, Laboratory of Systems Biology, and
| | - Shamira Shallom
- 3 Microbiology Service, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland; and
| | - Olena Kamenyeva
- 4 Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | | | | | - Juraj Kabat
- 4 Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Kenneth N Olivier
- 5 Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Adrian M Zelazny
- 3 Microbiology Service, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland; and
| | - John S Tsang
- 2 Systems Genomics and Bioinformatics Unit, Laboratory of Systems Biology, and
| | | |
Collapse
|
27
|
Susceptibility to non-tuberculous mycobacterial disease is influenced by rs1518111 in IL10. Hum Immunol 2017; 78:391-393. [DOI: 10.1016/j.humimm.2017.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 02/01/2017] [Accepted: 02/01/2017] [Indexed: 01/15/2023]
|
28
|
Bruffaerts N, Vluggen C, Roupie V, Duytschaever L, Van den Poel C, Denoël J, Wattiez R, Letesson JJ, Fretin D, Rigouts L, Chapeira O, Mathys V, Saegerman C, Huygen K. Virulence and immunogenicity of genetically defined human and porcine isolates of M. avium subsp. hominissuis in an experimental mouse infection. PLoS One 2017; 12:e0171895. [PMID: 28182785 PMCID: PMC5300754 DOI: 10.1371/journal.pone.0171895] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/27/2017] [Indexed: 12/02/2022] Open
Abstract
Mycobacterium avium subsp. hominissuis (Mah) represents a health concern for humans and to a lesser extent for pigs, but its zoonotic potential remains elusive. Using multispacer sequence typing (MST) we previously identified 49 different genotypes of Mah among Belgian clinical and porcine isolates, with 5 MSTs shared by both hosts. Using experimental intranasal infection of BALB/c mice, we compared the virulence and immunogenicity of porcine and clinical human isolates with shared genotype or with a genotype only found in humans or pigs. Bacterial replication was monitored for 20 weeks in lungs, spleen and liver and mycobacteria specific spleen cell IFN-γ, IL-10 and IL-17 production as well as serum antibody responses were analyzed. Isolates varied in virulence, with human and porcine isolates sharing MST22 genotype showing a thousand fold higher bacterial replication in lungs and more dissemination to spleen and liver than the human and porcine MST91 isolates. Virulent MST22 type was also associated with progressive suppression of IFN-γ and IL-17 responses, and increased IL-10 production. Whole genome sequencing of the two virulent isolates with MST22 genotype and two avirulent isolates of genotype MST91 and comparison with two well-studied M. avium subsp. hominissuis reference strains i.e. Mah 104 and Mah TH135, identified in the two MST22 isolates nine specific virulence factors of the mammalian cell entry family, that were identical with Mah 104 strain. Despite the obvious limitations of the mouse model, a striking link of virulence and identity at the genome level of porcine and human isolates with the same multisequence type, for which no correlation of place of residence (humans) or farm of origin (pigs) was observed, seems to point to the existence in the environment of certain genotypes of Mah which may be more infectious both for humans and pigs than other genotypes.
Collapse
Affiliation(s)
- Nicolas Bruffaerts
- Service Immunology, Operational Direction Communicable and infectious Diseases, Scientific Institute of Public Health, Brussels, Belgium
- * E-mail: (NB); (KH)
| | - Christelle Vluggen
- Service Bacterial diseases, Operational Direction Communicable and infectious Diseases, Scientific Institute of Public Health, Brussels, Belgium
| | - Virginie Roupie
- Unit Bacterial Zoonoses of livestock, Operational Direction Bacterial Diseases, Veterinary and Agrochemical Research Centre, Brussels, Belgium
| | - Lucille Duytschaever
- Unit Bacterial Zoonoses of livestock, Operational Direction Bacterial Diseases, Veterinary and Agrochemical Research Centre, Brussels, Belgium
- Research Unit in Epidemiology and Risk Analysis applied to Veterinary Sciences, Fundamental and Applied Research for Animal and Health, Université of Liège, Liège, Belgium
| | - Christophe Van den Poel
- Service Immunology, Operational Direction Communicable and infectious Diseases, Scientific Institute of Public Health, Brussels, Belgium
| | - Joseph Denoël
- Research Unit in Epidemiology and Risk Analysis applied to Veterinary Sciences, Fundamental and Applied Research for Animal and Health, Université of Liège, Liège, Belgium
| | - Ruddy Wattiez
- Service Protéomique et Microbiologie, Université de Mons, Mons, Belgium
| | - Jean-Jacques Letesson
- Unité de Recherche en Biologie des Microorganismes, Université de Namur, Namur, Belgium
| | - David Fretin
- Unit Bacterial Zoonoses of livestock, Operational Direction Bacterial Diseases, Veterinary and Agrochemical Research Centre, Brussels, Belgium
| | - Leen Rigouts
- Department Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Unit Mycobacteriology, Institute of Tropical Medicine, Antwerp, Belgium
| | | | - Vanessa Mathys
- Service Bacterial diseases, Operational Direction Communicable and infectious Diseases, Scientific Institute of Public Health, Brussels, Belgium
| | - Claude Saegerman
- Research Unit in Epidemiology and Risk Analysis applied to Veterinary Sciences, Fundamental and Applied Research for Animal and Health, Université of Liège, Liège, Belgium
| | - Kris Huygen
- Service Immunology, Operational Direction Communicable and infectious Diseases, Scientific Institute of Public Health, Brussels, Belgium
- * E-mail: (NB); (KH)
| |
Collapse
|
29
|
Lake MA, Ambrose LR, Lipman MCI, Lowe DM. '"Why me, why now?" Using clinical immunology and epidemiology to explain who gets nontuberculous mycobacterial infection. BMC Med 2016; 14:54. [PMID: 27007918 PMCID: PMC4806462 DOI: 10.1186/s12916-016-0606-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 03/18/2016] [Indexed: 02/01/2023] Open
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. 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.
Collapse
Affiliation(s)
- M Alexandra Lake
- Royal Free London NHS Foundation Trust, London, UK.,Division of Infection and Immunity, University College London, London, UK
| | - Lyn R Ambrose
- Institute of Immunity and Transplantation, University College London, Royal Free Campus, Pond Street, London, NW3 2QG, UK
| | - Marc C I Lipman
- Royal Free London NHS Foundation Trust, London, UK.,UCL Respiratory, Division of Medicine, Faculty of Medical Sciences, University College London, Royal Free Campus, London, UK
| | - David M Lowe
- Royal Free London NHS Foundation Trust, London, UK. .,Institute of Immunity and Transplantation, University College London, Royal Free Campus, Pond Street, London, NW3 2QG, UK.
| |
Collapse
|
30
|
Is Pulmonary non-Tuberculous Mycobacterial Disease Linked with a High Burden of Latent Cytomegalovirus? J Clin Immunol 2016; 36:113-6. [PMID: 26759253 DOI: 10.1007/s10875-016-0233-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 01/04/2016] [Indexed: 01/13/2023]
Abstract
Cytomegalovirus (CMV) establishes lifelong infections with episodes of active replication. We hypothesized that recurrent CMV replication in older individuals may suppress protective immune responses to non-tuberculous mycobacteria (NTM) and so potentiate pulmonary disease. Accordingly, levels of antibodies to three CMV antigen preparations were higher in NTM patients than in age-matched controls. This did not reflect broad-spectrum B cell activation as total immunoglobulin levels were not equivalently increased.
Collapse
|
31
|
Becker KL, van Ingen J, Ten Oever J, Merkus PJ, Ferwerda G, Netea MG, Magis-Escurra C, Reijers MH, van de Veerdonk FL. Deficient interleukin-17 production in response to Mycobacterium abscessus in cystic fibrosis. Eur Respir J 2016; 47:990-3. [PMID: 26743483 DOI: 10.1183/13993003.00446-2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 10/20/2015] [Indexed: 11/05/2022]
Affiliation(s)
- Katharina L Becker
- Dept of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jakko van Ingen
- Dept of Medical Microbiology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jaap Ten Oever
- Dept of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Peter J Merkus
- Dept of Pediatrics, Division of Respiratory Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Gerben Ferwerda
- Dept of Pediatrics, Laboratory of Pediatric Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Mihai G Netea
- Dept of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Cecile Magis-Escurra
- Dept of Pulmonology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Monique H Reijers
- Dept of Pulmonology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | |
Collapse
|
32
|
Host susceptibility to non-tuberculous mycobacterial infections. THE LANCET. INFECTIOUS DISEASES 2015; 15:968-80. [PMID: 26049967 DOI: 10.1016/s1473-3099(15)00089-4] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Revised: 10/29/2014] [Accepted: 11/11/2014] [Indexed: 11/22/2022]
Abstract
Non-tuberculous mycobacteria cause a broad range of clinical disorders, from cutaneous infections, such as cervical or intrathoracic lymphadenitis in children, to disseminated infections at all ages. Recognition of the underlying immune defect is crucial for rational treatment, preventive care, family screening, and, in some cases, transplantation. So far, at least seven autosomal mutations (in IL12B, IL12RB1, ISG15, IFNGR1, IFNGR2, STAT1, and IRF8) and two X-linked mutations (in IKBKG and CYBB), mostly presenting in childhood, have been reported to confer susceptibility to disseminated non-tuberculous mycobacterial infection. GATA2 deficiency and anti-interferon γ autoantibodies also give rise to disseminated infection, typically in late childhood or adulthood. Furthermore, isolated pulmonary non-tuberculous mycobacterial infection has been increasing in prevalence in people without recognised immune dysfunction. In this Review, we discuss how to detect and differentiate host susceptibility factors underlying localised and systemic non-tuberculous mycobacterial infections.
Collapse
|
33
|
Attia EAS, Abdallah M, El-Khateeb E, Saad AA, Lotfi RA, Abdallah M, El-Shennawy D. Serum Th17 cytokines in leprosy: correlation with circulating CD4(+) CD25 (high)FoxP3 (+) T-regs cells, as well as down regulatory cytokines. Arch Dermatol Res 2014; 306:793-801. [PMID: 25018055 DOI: 10.1007/s00403-014-1486-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 06/26/2014] [Accepted: 07/08/2014] [Indexed: 12/16/2022]
Abstract
Leprosy is not only a bacteriological disease but also an immunological disease, in which T helper17 and CD4(+) CD25(high)FoxP3(+) regulatory T cells (T-regs), among others, may play a role. We aimed to evaluate serum levels of interleukin (IL)-17, IL-22 (Th17 cytokines), IL-10 and transforming growth factor (TGF)-β (down regulatory cytokines) in 43 untreated leprosy patients and 40 controls by enzyme-linked immunosorbent assay, and to assess circulating CD4(+) CD25(high)FoxP3(+)T-regs in patients using flow cytometry. Patients were grouped into tuberculoid, pure neural, borderline, lepromatous, type 1 reactional leprosy, and erythema nodosum leprosum. IL-10 and TGF-β were significantly higher in patients as compared to controls (p < 0.001), while IL-17, but not IL-22, was significantly lower (p < 0.001), with no significant difference comparing patients' subgroups. Significantly higher CD4(+) CD25(high)FoxP3(+)T-regs levels was detected in tuberculoid, type 1 reaction and pure neural leprosy, while the lowest levels in erythema nodosum leprosum (p < 0.001). TregsFoxP3 expression% was significantly lower in pure neural leprosy than other patients' subgroups (p < 0.05). T-regs/T-effs was lowest in erythema nodosum leprosum (p < 0.05). TGF-β correlated negatively with TregsFoxP3 expression% and T-effs% (p = 0.009 and 0.018 respectively). Leprosy is associated with defective IL-17 and overproduction of IL-10 and TGF-β. Tuberculoid, type 1 reaction and pure neural leprosy express significantly higher circulating T-regs, consistent with effector immune mechanisms activation, but with lower TregsFoxP3 expression (in pure neural leprosy). Erythema nodosum leprosum is characterized by deficient T-regs and increased TregsFoxP3 expression%. The present study pinpointed a potential role of Th17, CD4(+) CD25(high)FoxP3(+)T-regs, and probably CD4(+) CD25(+)IL-10(+) T regulatory cells 1 (Tr1), and Th3 in leprosy.
Collapse
Affiliation(s)
- E A S Attia
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Ain Shams University, Cairo, 11381, Egypt,
| | | | | | | | | | | | | |
Collapse
|
34
|
Kim SY, Koh WJ, Kim YH, Jeong BH, Park HY, Jeon K, Kim JS, Cho SN, Shin SJ. Importance of reciprocal balance of T cell immunity in Mycobacterium abscessus complex lung disease. PLoS One 2014; 9:e109941. [PMID: 25295870 PMCID: PMC4190320 DOI: 10.1371/journal.pone.0109941] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 09/05/2014] [Indexed: 11/18/2022] Open
Abstract
Background Little is known about the nature of the host immune response to Mycobacterium abscessus complex (MABC) infection. The aim of the present study was to investigate whether alterations in serum immunomolecule levels after treating MABC lung disease patients with antibiotics can reflect the disease-associated characteristics. Methods A total of 22 immunomolecules in 24 MABC lung disease patients before and after antibiotic therapy were quantitatively analyzed using a multiplex bead-based system. Results In general, the pre-treatment levels of T helper type 1 (Th1)-related cytokines, i.e., interferon (IFN)-γ and interleukin (IL)-12, and Th2-related cytokines, i.e., IL-4 and IL-13, were significantly decreased in patients compared with control subjects. In contrast, the pre-treatment levels of Th17-related cytokines, i.e., IL-17 and IL-23, were significantly increased in MABC patients. Interestingly, significantly higher levels of IFN-γ-induced protein (IP)-10 and monokine induced by IFN-γprotein (MIG) were detected in patients with failure of sputum conversion at post-treatment compared to patients with successful sputum conversion. Conclusion Reduced Th1 and Th2 responses and enhanced Th17 responses in patients may perpetuate MABC lung disease, and the immunomolecules IP-10 and MIG, induced through IFN-γ, may serve as key markers for indicating the treatment outcome.
Collapse
Affiliation(s)
- Su-Young Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Won-Jung Koh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yee Hyung Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Byeong-Ho Jeong
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hye Yun Park
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyeongman Jeon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jong-Seok Kim
- Department of Microbiology, Institute of Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang-Nae Cho
- Department of Microbiology, Institute of Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute of Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- * E-mail:
| |
Collapse
|
35
|
Kim SY, Koh WJ, Park HY, Jeon K, Kwon OJ, Cho SN, Shin SJ. Changes in serum immunomolecules during antibiotic therapy for Mycobacterium avium complex lung disease. Clin Exp Immunol 2014; 176:93-101. [PMID: 24354934 DOI: 10.1111/cei.12253] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2013] [Indexed: 01/09/2023] Open
Abstract
Little information is available regarding changes in immune status for patients with Mycobacterium avium complex (MAC) lung disease during antibiotic therapy. Serum immunomolecules from 42 patients with MAC lung disease were assayed comparatively using an array-based system according to (i) patients with MAC lung disease at the time of diagnosis versus healthy controls and (ii) alterations after 12 months of antibiotic therapy in the MAC lung disease group. In addition, cytokine analyses were performed to determine whether cytokine responses were associated specifically with the disease phenotype, treatment outcome and aetiological agent. Notably, the serum concentrations of type 1 cytokine-associated molecules, such as CD40L, interferon (IFN)-γ, interleukin (IL)-8 and IL-23, were decreased significantly in patients at the time of diagnosis, suggesting that these molecules may serve as indicators of host susceptibility to MAC disease. Although the overall serum level of T helper type 1 (Th1)-related molecules, such as CD40L and IFN-γ, was restored after treatment, Th17-related cytokines, such as IL-17 and IL-23, were down-regulated significantly at 12 months post-treatment compared to pretreatment. Furthermore, these cytokine patterns differed among patient subgroups. Decreased serum concentrations of IL-17 and/or IL-23 were associated with failure of sputum conversion, the fibrocavitary disease phenotype and M. intracellulare lung disease. Thus, the reciprocal balance between Th1 and Th17 immunity during antibiotic therapy for MAC lung disease is critical for dictating the treatment response. In conclusion, a low level of Th1-related immunomolecules may perpetuate MAC lung disease, and the serum concentrations of Th17-related cytokines can reflect the treatment outcome, disease phenotype and aetiological agent.
Collapse
Affiliation(s)
- S-Y Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | | | | | | | | |
Collapse
|
36
|
Searching for an immunogenetic factor that will illuminate susceptibility to non-tuberculous mycobacterial disease. Hum Immunol 2013; 74:1382-5. [DOI: 10.1016/j.humimm.2013.06.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/24/2013] [Accepted: 06/07/2013] [Indexed: 11/24/2022]
|
37
|
Kartalija M, Ovrutsky AR, Bryan CL, Pott GB, Fantuzzi G, Thomas J, Strand MJ, Bai X, Ramamoorthy P, Rothman MS, Nagabhushanam V, McDermott M, Levin AR, Frazer-Abel A, Giclas PC, Korner J, Iseman MD, Shapiro L, Chan ED. Patients with nontuberculous mycobacterial lung disease exhibit unique body and immune phenotypes. Am J Respir Crit Care Med 2013; 187:197-205. [PMID: 23144328 PMCID: PMC5446199 DOI: 10.1164/rccm.201206-1035oc] [Citation(s) in RCA: 165] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 10/10/2012] [Indexed: 01/15/2023] Open
Abstract
RATIONALE Among patients with nontuberculous mycobacterial lung disease is a subset of previously healthy women with a slender body morphotype, often with scoliosis and/or pectus excavatum. We hypothesize that unidentified factors predispose these individuals to pulmonary nontuberculous mycobacterial disease. OBJECTIVES To compare body morphotype, serum adipokine levels, and whole-blood cytokine responses of patients with pulmonary nontuberculous mycobacteria (pNTM) with contemporary control subjects who are well matched demographically. METHODS We enrolled 103 patients with pNTM and 101 uninfected control subjects of similar demographics. Body mass index and body fat were quantified. All patients with pNTM and a subset of control subjects were evaluated for scoliosis and pectus excavatum. Serum leptin and adiponectin were measured. Specific cytokines important to host-defense against mycobacteria were measured in whole blood before and after stimulation. MEASUREMENTS AND MAIN RESULTS Patients with pNTM and control subjects were well matched for age, gender, and race. Patients with pNTM had significantly lower body mass index and body fat and were significantly taller than control subjects. Scoliosis and pectus excavatum were significantly more prevalent in patients with pNTM. The normal relationships between the adipokines and body fat were lost in the patients with pNTM, a novel finding. IFN-γ and IL-10 levels were significantly suppressed in stimulated whole blood of patients with pNTM. CONCLUSIONS This is the first study to comprehensively compare body morphotype, adipokines, and cytokine responses between patients with NTM lung disease and demographically matched controls. Our findings suggest a novel, predisposing immunophenotype that should be mechanistically defined.
Collapse
Affiliation(s)
| | | | - Courtney L. Bryan
- Division of Infectious Diseases
- Denver Veterans Affairs Medical Center, Denver, Colorado
| | - Gregory B. Pott
- Division of Infectious Diseases
- Denver Veterans Affairs Medical Center, Denver, Colorado
| | - Giamila Fantuzzi
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois; and
| | | | | | - Xiyuan Bai
- Division of Pulmonary Sciences and Critical Care Medicine, and
- Departments of Medicine and Academic Affairs
| | | | - Micol S. Rothman
- Division of Endocrinology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | | | - Michael McDermott
- Division of Endocrinology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | | | | | | | - Judith Korner
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Michael D. Iseman
- Division of Infectious Diseases
- Division of Pulmonary Sciences and Critical Care Medicine, and
- Departments of Medicine and Academic Affairs
| | - Leland Shapiro
- Division of Infectious Diseases
- Denver Veterans Affairs Medical Center, Denver, Colorado
| | - Edward D. Chan
- Division of Pulmonary Sciences and Critical Care Medicine, and
- Departments of Medicine and Academic Affairs
- Denver Veterans Affairs Medical Center, Denver, Colorado
| |
Collapse
|
38
|
Monocyte-derived macrophages do not explain susceptibility to pulmonary non-tuberculous mycobacterial disease. Clin Transl Immunology 2012; 1:e2. [PMID: 25505947 PMCID: PMC4256549 DOI: 10.1038/cti.2012.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 09/04/2012] [Indexed: 12/24/2022] Open
Abstract
Pulmonary infections with non-tuberculous mycobacteria (NTM) affect a subset of older individuals (mostly women) with no known immunological defects. As NTMs are intracellular pathogens, it is important to establish whether NTM disease is associated with defective production of Th1 cytokines or poor responses by host macrophage/monocytes. We have shown that patients display vigorous production of interferon gamma (IFNγ) when CD4 T cells are stimulated with mycobacterial antigens. This implicated the macrophage response to IFNγ. Blood monocytes are poorly representative of lung macrophages, so monocyte-derived macrophages (MDMs) were created and then stimulated with lipomannan (a Toll-like receptor (TLR)2 agonist), lipopolysaccharide (LPS; a TLR4 agonist) or recombinant human IFNγ. MDMs from NTM patients, their offspring and healthy donors expressed similar amounts of IFNγR1, and cellular responses to IFNγ were similar, so there is no evidence of a genetic defect in this pathway. MDMs from NTM patients produced less interleukin-6 in response to LPS (P<0.01) than cells from controls, but other cytokine responses were normal. This warrants further study.
Collapse
|
39
|
Jönsson B, Ridell M, Wold AE. Non-tuberculous mycobacteria and their surface lipids efficiently induced IL-17 production in human T cells. Microbes Infect 2012; 14:1186-95. [DOI: 10.1016/j.micinf.2012.07.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 07/25/2012] [Accepted: 07/26/2012] [Indexed: 01/09/2023]
|
40
|
Cochrane M, Armitage CW, O’Meara CP, Beagley KW. Towards a Chlamydia trachomatis vaccine: how close are we? Future Microbiol 2010; 5:1833-56. [DOI: 10.2217/fmb.10.148] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infections and preventable blindness worldwide. The incidence of chlamydial sexually transmitted infections has increased rapidly and current antibiotic therapy has failed as an intervention strategy. The most accepted strategy for protection and/or control of chlamydial infections is a vaccine that induces both local neutralizing antibodies to prevent infections by the extracellular elementary bodies and a cell-mediated immune response to target the intracellular infection. This article will discuss the challenges in vaccine design for the prevention of chlamydial urogenital infection and/or disease, including selection of target antigens, discussion of effective delivery systems, immunization routes and adjuvants for induction of protective immunity at the targeted mucosal surface whilst minimizing severe inflammatory disease sequelae.
Collapse
Affiliation(s)
- Melanie Cochrane
- Institute of Health & Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Charles W Armitage
- Institute of Health & Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Connor P O’Meara
- Institute of Health & Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | | |
Collapse
|