51
|
Shah P, Rebick G, Bajaj S, Desvignes L, Lo Sicco K. Evidenced-based guidelines for tuberculosis screening before biologic treatment initiation. J Am Acad Dermatol 2020; 83:e25-e26. [DOI: 10.1016/j.jaad.2020.02.084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 11/25/2022]
|
52
|
Mortazavi Moghaddam SG, Namaei MH, Eslami Manoochehri R, Zardast M. The sequential assay of interleukin-10 and 13 serum levels in relation to radiographic changes during pulmonary tuberculosis treatment. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2020; 25:63. [PMID: 33088300 PMCID: PMC7554419 DOI: 10.4103/jrms.jrms_116_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 11/15/2019] [Accepted: 02/26/2020] [Indexed: 11/22/2022]
Abstract
Background: We evaluated the sequential changes of interleukin (IL)-10 and IL-13 serum levels with tuberculosis (TB)-related radiographic changes during pulmonary TB (PTB) treatment. Materials and Methods: In this cross-sectional study during two consecutive years, forty cases with PTB were recorded, and finally, 24 cases were completed the study. Serum levels of IL-10 and IL-13 were measured on admission time, and 6 months later. Furthermore, chest radiography was performed on admission and 6 months later in the treatment course. Results: Radiography at the baseline indicated pulmonary infiltration in all patients (n = 24). Fifteen (62.5%) cases had abnormal and 9 (37.5%) cases had normal radiography at the end of 6 months treatment course. IL-10 and IL-13 upregulated during the treatment time course, and their relationship with radiographic changes shifted from negative (r = −0.14 and P = 0.71) on admission to positive (r = 0.80 and P < 0.001) at the end of 6 months treatment course in normal radiography group. IL-10 level at the start of the treatment was 121.90 ± 88.81 in patients with normal and 82.68 ± 41.50 in patients with abnormal radiography (P = 0.31). Conclusion: Sequential increase in IL-10 and IL-13 during PTB treatment course may have a role in clearing the TB-related radiographic infiltration and preventing scar formation.
Collapse
Affiliation(s)
- Sayyed Gholamreza Mortazavi Moghaddam
- Department of Internal Medicine, Division of Pulmonary, School of Medicine, Vali-e-Asr Hospital, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Hasan Namaei
- Infectius Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Reza Eslami Manoochehri
- Department of Internal Medicine, School of Medicine, Vali-e-Asr Hospital, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahmood Zardast
- Department of Pathology, School of Medicine, Vali-e-Asr Hospital, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
53
|
Sun W, Jiao L, Liu T, Song J, Wang M, Liang L, Wen C, Hu L, Qu W, Ying B. No Significant Effects of IL-6 and IL-13 Gene Variants on Tuberculosis Susceptibility in the Chinese Population. DNA Cell Biol 2020; 39:1356-1367. [PMID: 32522041 DOI: 10.1089/dna.2020.5404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Tuberculosis (TB) is an intricate infectious disease that causes a large number of deaths in the population. Interleukin (IL)-6 and IL-13 play functional roles in host resistance to Mycobacterium tuberculosis infection. Our aim in this study was to explore the association of IL-6 and IL-13 polymorphisms with TB susceptibility in the Western Chinese Han population. The case and control groups comprised 900 TB patients and 1534 healthy controls, respectively, and four single-nucleotide polymorphisms (SNPs) were genotyped in IL-6 and five SNPs in IL-13 through the improved multiplex ligation detection reaction method. We found no genetic variants in the IL-6 or IL-13 genes that were related to TB susceptibility in the analysis of alleles, genotypes, genetic models, and TB clinical subtypes, except for a trend toward low pulmonary tuberculosis and extrapulmonary tuberculosis susceptibility for the SNPs rs1295686 and rs20541. Our study did not find a link between IL-6 and IL-13 polymorphisms and TB susceptibility in the Western Chinese Han population. Therefore, our present data revealed the challenge of applying IL-6 and IL-13 SNPs as genetic markers for TB and that increased sample sizes and additional races are needed for further studies.
Collapse
Affiliation(s)
- Wei Sun
- Department of Laboratory Medicine, Guiyang Children's Hospital, Guiyang, China
| | - Lin Jiao
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Tangyuheng Liu
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Jiajia Song
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Minjin Wang
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Lu Liang
- Department of Laboratory Medicine, Guiyang Children's Hospital, Guiyang, China
| | - Chunrong Wen
- Department of Laboratory Medicine, Guiyang Children's Hospital, Guiyang, China
| | - Lei Hu
- Department of Laboratory Medicine, Guiyang Children's Hospital, Guiyang, China
| | - Wei Qu
- Department of Laboratory Medicine, Guiyang Children's Hospital, Guiyang, China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
54
|
Moideen K, Kumar NP, Bethunaickan R, Banurekha VV, Nair D, Babu S. Heightened systemic levels of anti-inflammatory cytokines in pulmonary tuberculosis and alterations following anti-tuberculosis treatment. Cytokine 2020; 127:154929. [DOI: 10.1016/j.cyto.2019.154929] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/21/2019] [Accepted: 11/13/2019] [Indexed: 01/29/2023]
|
55
|
Vavougios GD. Mycobacterial immunomodulation and viral manipulation of neuronal copper efflux in the setting of sporadic Parkinson's disease: A multi - hit, outside - in hypothesis of its pathogenesis. Med Hypotheses 2019; 136:109505. [PMID: 31765844 DOI: 10.1016/j.mehy.2019.109505] [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: 09/09/2019] [Revised: 11/07/2019] [Accepted: 11/16/2019] [Indexed: 12/24/2022]
Abstract
Following Braak's hypothesis on the infectious pathogenesis of sporadic Parkinson's disease (sPD), several bacteria and viruses have been investigated as likely culprits. Recent research has focused on neuroinvasive influenza A viruses (IAV), whereas a genetic link between sPD and tuberculosis has arisen in LRRK2 - dependent maturation of the phagosome. An integrative, outside - in, multi - hit hypothesis is presented here, where (a) mycobacterial immunomodulation creates a phagocyte niche along with cytokine mediated, site specific (i.e. the gut) alterations of both immunity and the microbiome, (b) copper modulating IAVs gain latency in and control over phagocytes and their phenotypes, (c) gain access to the central nervous system (CNS) via the olfactory and vagus nerves in subsequent infection cycles, (d) induce indolent neuroinflammation characterized by perturbed intraneuronal copper compartmentalization and (e) produce α - synuclein (aSyn) pathology at least in part via copper - induced aggregation and misfolding as well as potential synergy with other underlying, corroborating factors (either genetic or acquired) contributing to dopaminergic neurodegeneration. This hypothesis explores recently arisen evidence for each step of this process, as well as pre-existing, yet unexplored overlapping pathophysiological characteristics of sPD with mycobacterial and IAV infections. The implications of this proposed pathogenic model extend both in sPD research (i.e. determining non - tuberculous mycobacteria as the first hit organism, inactivating IAV - induced copper hijacking), as well as therapeutics.
Collapse
Affiliation(s)
- George D Vavougios
- Athens Naval Hospital, Department of Neurology, Deinokratous 70, Athens 11521, Greece; University of Thessaly Department of Electrical and Computer Engineering, University of Thessaly, Volos 38221, Greece.
| |
Collapse
|
56
|
Pooran A, Davids M, Nel A, Shoko A, Blackburn J, Dheda K. IL-4 subverts mycobacterial containment in Mycobacterium tuberculosis-infected human macrophages. Eur Respir J 2019; 54:13993003.02242-2018. [PMID: 31097521 DOI: 10.1183/13993003.02242-2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 05/07/2019] [Indexed: 12/17/2022]
Abstract
Protective immunity against Mycobacterium tuberculosis is poorly understood. The role of interleukin (IL)-4, the archetypal T-helper type 2 (Th2) cytokine, in the immunopathogenesis of human tuberculosis remains unclear.Blood and/or bronchoalveolar lavage fluid (BAL) were obtained from participants with pulmonary tuberculosis (TB) (n=23) and presumed latent TB infection (LTBI) (n=22). Messenger RNA expression levels of interferon (IFN)-γ, IL-4 and its splice variant IL-4δ2 were determined by real-time PCR. The effect of human recombinant (hr)IL-4 on mycobacterial survival/containment (CFU·mL-1) was evaluated in M. tuberculosis-infected macrophages co-cultured with mycobacterial antigen-primed effector T-cells. Regulatory T-cell (Treg) and Th1 cytokine levels were evaluated using flow cytometry.In blood, but not BAL, IL-4 mRNA levels (p=0.02) and the IL-4/IFN-γ ratio (p=0.01) was higher in TB versus LTBI. hrIL-4 reduced mycobacterial containment in infected macrophages (p<0.008) in a dose-dependent manner and was associated with an increase in Tregs (p<0.001), but decreased CD4+Th1 cytokine levels (CD4+IFN-γ+ p<0.001; CD4+TNFα+ p=0.01). Blocking IL-4 significantly neutralised mycobacterial containment (p=0.03), CD4+IFNγ+ levels (p=0.03) and Treg expression (p=0.03).IL-4 can subvert mycobacterial containment in human macrophages, probably via perturbations in Treg and Th1-linked pathways. These data may have implications for the design of effective TB vaccines and host-directed therapies.
Collapse
Affiliation(s)
- Anil Pooran
- Centre for Lung Infection and Immunity, Division of Pulmonology, Dept of Medicine and UCT Lung Institute & South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
| | - Malika Davids
- Centre for Lung Infection and Immunity, Division of Pulmonology, Dept of Medicine and UCT Lung Institute & South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
| | - Andrew Nel
- Dept of Integrative Biomedical Sciences, Institute for Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Aubrey Shoko
- Centre for Proteomics and Genomics Research, Cape Town, South Africa
| | - Jonathan Blackburn
- Dept of Integrative Biomedical Sciences, Institute for Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Keertan Dheda
- Centre for Lung Infection and Immunity, Division of Pulmonology, Dept of Medicine and UCT Lung Institute & South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa .,Faculty of Infectious and Tropical Diseases, Dept of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
57
|
Bahr NC, Meintjes G, Boulware DR. Inadequate diagnostics: the case to move beyond the bacilli for detection of meningitis due to Mycobacterium tuberculosis. J Med Microbiol 2019; 68:755-760. [PMID: 30994435 PMCID: PMC7176281 DOI: 10.1099/jmm.0.000975] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Tuberculosis (TB) meningitis is extremely difficult to diagnose due to its pauci-bacillary disease nature and new techniques are needed. Improved test sensitivity would allow for greater clinician confidence in diagnostic testing and has the potential to improve patient outcomes. Traditional microbiologic and molecular tests for TB meningitis focus on detection of TB bacilli and are inadequate. Smear microscopy is rapid but only ~10-15 % sensitive. Culture has 50-60 % sensitivity but is slow. Xpert MTB/Rif Ultra is a rapid, automated PCR-based assay with ~70 % sensitivity versus clinical case definition. Thus, even the best current testing may miss up to 30 % of cases. Clinicians are often left to treat empirically with prolonged regimens with significant side effects or risk a missed case that would result in death. Rather than relying strictly on microbiologic or molecular testing to diagnose TB meningitis, we propose that testing of CSF for biomarkers of host response may have an adjunctive role to play in improving the diagnosis of TB meningitis.
Collapse
Affiliation(s)
- Nathan C. Bahr
- University of Kansas, Kansas City, KS, USA
- *Correspondence: Nathan C. Bahr,
| | | | | |
Collapse
|
58
|
Arrey F, Löwe D, Kuhlmann S, Kaiser P, Moura-Alves P, Krishnamoorthy G, Lozza L, Maertzdorf J, Skrahina T, Skrahina A, Gengenbacher M, Nouailles G, Kaufmann SHE. Humanized Mouse Model Mimicking Pathology of Human Tuberculosis for in vivo Evaluation of Drug Regimens. Front Immunol 2019; 10:89. [PMID: 30766535 PMCID: PMC6365439 DOI: 10.3389/fimmu.2019.00089] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/14/2019] [Indexed: 11/13/2022] Open
Abstract
Human immune system mice are highly valuable for in vivo dissection of human immune responses. Although they were employed for analyzing tuberculosis (TB) disease, there is little data on the spatial organization and cellular composition of human immune cells in TB granuloma pathology in this model. We demonstrate that human immune system mice, generated by transplanted human fetal liver derived hematopoietic stem cells develop a continuum of pulmonary lesions upon Mycobacterium tuberculosis aerosol infection. In particular, caseous necrotic granulomas, which contribute to prolonged TB treatment time, developed, and had cellular phenotypic spatial-organization similar to TB patients. By comparing two recommended drug regimens, we confirmed observations made in clinical settings: Adding Moxifloxacin to a classical chemotherapy regimen had no beneficial effects on bacterial eradication. We consider this model instrumental for deeper understanding of human specific features of TB pathogenesis and of particular value for the pre-clinical drug development pipeline.
Collapse
Affiliation(s)
- Frida Arrey
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Delia Löwe
- Department of Molecular Pharmacology and Cell Biology, Leibniz Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Stefanie Kuhlmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Peggy Kaiser
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Pedro Moura-Alves
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | | | - Laura Lozza
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Jeroen Maertzdorf
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Tatsiana Skrahina
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Alena Skrahina
- Republican Scientific and Practical Centre for Pulmonology and Tuberculosis, Minsk, Belarus
| | - Martin Gengenbacher
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Geraldine Nouailles
- Division of Pulmonary Inflammation, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan H. E. Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
59
|
Mycobacterium tuberculosis antigens repress Th1 immune response suppression and promotes lung cancer metastasis through PD-1/PDl-1 signaling pathway. Cell Death Dis 2019; 10:44. [PMID: 30718463 PMCID: PMC6362089 DOI: 10.1038/s41419-018-1237-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/11/2018] [Accepted: 11/19/2018] [Indexed: 12/14/2022]
Abstract
Given one-third of the world's population is infected with Mycobacterium tuberculosis (MTB), it is important to identify the underling molecular mechanism between development of TB and lung cancer. This study investigated the immune response to MTB infection on lung metastasis in lung cancer cells via T cell-mediated immune response. To clarify this problem, we analyzed the expression levels of PD-1, PD-L1, and PD-L2 and immune function in antigen-specific T cell as derived from MTB patients or spleen lymphocytes derived from wild-type and PD-1 knockout mice with MTB antigen stimulation and Lewis lung cancer cells injection. Our data indicate that the expression levels of PD-1, PD-L1, and PD-L2 were elevated in active pulmonary TB patients, as well as in mice received MTB and lung cancer cells treatment. We also observed the T cell-mediated cellular immune response were inhibited by MTB while MTB significantly promote tumor metastasis in lung. In conclusion, the PD-1/PD-L pathway is required MTB repressed T-cell immune response and promotes tumor metastasis. This study provides evidence that blockade of PD-1/PD-L1 signaling pathway may benefit patients with MTB or other chronic infection and even prevent them from development of cancer.
Collapse
|
60
|
Jung J, Zeng H, Horng T. Metabolism as a guiding force for immunity. Nat Cell Biol 2019; 21:85-93. [PMID: 30602764 DOI: 10.1038/s41556-018-0217-x] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022]
Abstract
Recent studies indicate that cellular metabolism plays a key role in supporting immune cell maintenance and development. Here, we review how metabolism guides immune cell activation and differentiation to distinct cellular states, and how differential regulation of metabolism allows for context-dependent support during activation and lineage commitment. We discuss emerging principles of metabolic support of immune cell function in physiology and disease, as well as their general relevance to the field of cell biology.
Collapse
Affiliation(s)
- Jonathan Jung
- Department of Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,School of Medicine, University of Glasgow, Glasgow, UK
| | - Hu Zeng
- Division of Rheumatology, Mayo Clinic, Rochester, MN, USA. .,Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| | - Tiffany Horng
- Department of Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA. .,ShanghaiTech University, Shanghai, China.
| |
Collapse
|
61
|
Morgan DJ, Casulli J, Chew C, Connolly E, Lui S, Brand OJ, Rahman R, Jagger C, Hussell T. Innate Immune Cell Suppression and the Link With Secondary Lung Bacterial Pneumonia. Front Immunol 2018; 9:2943. [PMID: 30619303 PMCID: PMC6302086 DOI: 10.3389/fimmu.2018.02943] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022] Open
Abstract
Secondary infections arise as a consequence of previous or concurrent conditions and occur in the community or in the hospital setting. The events allowing secondary infections to gain a foothold have been studied for many years and include poor nutrition, anxiety, mental health issues, underlying chronic diseases, resolution of acute inflammation, primary immune deficiencies, and immune suppression by infection or medication. Children, the elderly and the ill are particularly susceptible. This review is concerned with secondary bacterial infections of the lung that occur following viral infection. Using influenza virus infection as an example, with comparisons to rhinovirus and respiratory syncytial virus infection, we will update and review defective bacterial innate immunity and also highlight areas for potential new investigation. It is currently estimated that one in 16 National Health Service (NHS) hospital patients develop an infection, the most common being pneumonia, lower respiratory tract infections, urinary tract infections and infection of surgical sites. The continued drive to understand the mechanisms of why secondary infections arise is therefore of key importance.
Collapse
Affiliation(s)
- David J Morgan
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Joshua Casulli
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Christine Chew
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Emma Connolly
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Sylvia Lui
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Oliver J Brand
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Rizwana Rahman
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Christopher Jagger
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Tracy Hussell
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
62
|
Abad Dar M, Hölscher C. Arginase-1 Is Responsible for IL-13-Mediated Susceptibility to Trypanosoma cruzi Infection. Front Immunol 2018; 9:2790. [PMID: 30555475 PMCID: PMC6281981 DOI: 10.3389/fimmu.2018.02790] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 11/13/2018] [Indexed: 01/17/2023] Open
Abstract
Arginase-1 (Arg-1) is a marker for alternatively activated macrophages (AAM) and is mainly induced by the type 2 cytokines interleukin (IL)-4 and IL-13 through the common IL-4 receptor-alpha (Rα) subunit. Both, Arg-1 and AAM undermine macrophage effector functions against intracellular parasites and are therefore implicated in the susceptibility to infection with Trypanosoma cruzi, the causative agent of Chagas' disease. However, the involvement of Arg-1 in promoting intracellular replication of T. cruzi in AAM has not been proven so far in vivo. Because Arg-1 is only moderately expressed in T. cruzi-infected wildtype mice, we elucidated the role of Arg-1 and AAM during infection in IL-13-overexpressing (IL-13tg) mice, which are characterized by an inflammation-induced development of AAM and an accompanied elevated expression of Arg-1. In comparison to wildtype littermates, IL-13tg mice were highly susceptible to T. cruzi infection with enhanced parasitemia and impaired survival. Importantly, T. cruzi-infected IL-13tg mice developed an elevated alternative macrophage activation with increased arginase activity. To proof the hypothesis, that Arg-1 accounts for the increased susceptibility of IL-13tg mice, we blocked arginase activity in infected IL-13tg mice. Because this arginase inhibition resulted in a decreased susceptibility to experimental Chagas disease our study supports in summary the conclusion that IL-13/IL-4Rα-driven Arg-1 expression contributes to the permissiveness of the host to T. cruzi infection.
Collapse
Affiliation(s)
- Mahin Abad Dar
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | | |
Collapse
|
63
|
Marakalala MJ, Martinez FO, Plüddemann A, Gordon S. Macrophage Heterogeneity in the Immunopathogenesis of Tuberculosis. Front Microbiol 2018; 9:1028. [PMID: 29875747 PMCID: PMC5974223 DOI: 10.3389/fmicb.2018.01028] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/01/2018] [Indexed: 12/13/2022] Open
Abstract
Macrophages play a central role in tuberculosis, as the site of primary infection, inducers and effectors of inflammation, innate and adaptive immunity, as well as mediators of tissue destruction and repair. Early descriptions by pathologists have emphasized their morphological heterogeneity in granulomas, followed by delineation of T lymphocyte-dependent activation of anti-mycobacterial resistance. More recently, powerful genetic and molecular tools have become available to describe macrophage cellular properties and their role in host-pathogen interactions. In this review we discuss aspects of macrophage heterogeneity relevant to the pathogenesis of tuberculosis and, conversely, lessons that can be learnt from mycobacterial infection, with regard to the immunobiological functions of macrophages in homeostasis and disease.
Collapse
Affiliation(s)
- Mohlopheni J. Marakalala
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Fernando O. Martinez
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
- Botnar Research Centre, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Annette Plüddemann
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Siamon Gordon
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
64
|
Zhao Y, Yang X, Zhang X, Yu Q, Zhao P, Wang J, Duan C, Li J, Johnson H, Feng X, Zhang H. IP-10 and RANTES as biomarkers for pulmonary tuberculosis diagnosis and monitoring. Tuberculosis (Edinb) 2018; 111:45-53. [PMID: 30029914 DOI: 10.1016/j.tube.2018.05.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 04/20/2018] [Accepted: 05/12/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE We aimed to determine whether IP-10 and RANTES plasma levels can be used in diagnosis and monitoring of pulmonary tuberculosis (PTB). METHODS Plasma levels of cytokines/chemokines were measured using a Bio-Plex® multiplex cytokine assay system in a cohort containing 457 clinically suspected PTB patients including a training set (n = 41)and two independent test sets A (n = 242) and B (n = 174). RESULTS Plasma levels of IP-10 and RANTES were significantly higher in PTB patients than healthy controls' in both training and independent test sets (P < 0.05). Compared with other combinations, the combination of IP-10 and RANTES had the best performance with an AUC of 1.0 in training set. The performance characteristic of this model was successfully validated in independent test set A although this combination only resulted in a slightly improvement of AUC value in independent test set B. Plasma IP-10 and RANTES levels were weakly and positively correlated with blood glucose concentrations. Moreover, IP-10 levels were positively correlated with CRP and ESR in PTB patients. Furthermore, in response to therapy, both IP-10 and RANTES levels significantly decreased over the period of 6 months (P < 0.001). CONCLUSIONS Taken together, combination of IP-10 and RANTES could be potentially used as diagnostic and monitoring biomarker in PTB management.
Collapse
Affiliation(s)
- Yanfeng Zhao
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China; Beijing Research Institute for Tuberculosis Control, No 5, Dongguang Hutong, Xinjiekou, Beijing, 100035, China
| | - Xiqin Yang
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Xuhui Zhang
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Qin Yu
- Chaoyang District Center for Disease Control and Prevention, 25 Panjiayuan, Huaweili, Beijing, 100029, China
| | - Ping Zhao
- Chaoyang District Center for Disease Control and Prevention, 25 Panjiayuan, Huaweili, Beijing, 100029, China
| | - Jianxia Wang
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Cuimi Duan
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Jiangxue Li
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | | | - Xiaoyan Feng
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China.
| | - Heqiu Zhang
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China.
| |
Collapse
|
65
|
Stüve P, Minarrieta L, Erdmann H, Arnold-Schrauf C, Swallow M, Guderian M, Krull F, Hölscher A, Ghorbani P, Behrends J, Abraham WR, Hölscher C, Sparwasser TD, Berod L. De Novo Fatty Acid Synthesis During Mycobacterial Infection Is a Prerequisite for the Function of Highly Proliferative T Cells, But Not for Dendritic Cells or Macrophages. Front Immunol 2018; 9:495. [PMID: 29675017 PMCID: PMC5895737 DOI: 10.3389/fimmu.2018.00495] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 02/26/2018] [Indexed: 12/21/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of human tuberculosis, is able to efficiently manipulate the host immune system establishing chronic infection, yet the underlying mechanisms of immune evasion are not fully understood. Evidence suggests that this pathogen interferes with host cell lipid metabolism to ensure its persistence. Fatty acid metabolism is regulated by acetyl-CoA carboxylase (ACC) 1 and 2; both isoforms catalyze the conversion of acetyl-CoA into malonyl-CoA, but have distinct roles. ACC1 is located in the cytosol, where it regulates de novo fatty acid synthesis (FAS), while ACC2 is associated with the outer mitochondrial membrane, regulating fatty acid oxidation (FAO). In macrophages, mycobacteria induce metabolic changes that lead to the cytosolic accumulation of lipids. This reprogramming impairs macrophage activation and contributes to chronic infection. In dendritic cells (DCs), FAS has been suggested to underlie optimal cytokine production and antigen presentation, but little is known about the metabolic changes occurring in DCs upon mycobacterial infection and how they affect the outcome of the immune response. We therefore determined the role of fatty acid metabolism in myeloid cells and T cells during Mycobacterium bovis BCG or Mtb infection, using novel genetic mouse models that allow cell-specific deletion of ACC1 and ACC2 in DCs, macrophages, or T cells. Our results demonstrate that de novo FAS is induced in DCs and macrophages upon M. bovis BCG infection. However, ACC1 expression in DCs and macrophages is not required to control mycobacteria. Similarly, absence of ACC2 did not influence the ability of DCs and macrophages to cope with infection. Furthermore, deletion of ACC1 in DCs or macrophages had no effect on systemic pro-inflammatory cytokine production or T cell priming, suggesting that FAS is dispensable for an intact innate response against mycobacteria. In contrast, mice with a deletion of ACC1 specifically in T cells fail to generate efficient T helper 1 responses and succumb early to Mtb infection. In summary, our results reveal ACC1-dependent FAS as a crucial mechanism in T cells, but not DCs or macrophages, to fight against mycobacterial infection.
Collapse
Affiliation(s)
- Philipp Stüve
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Lucía Minarrieta
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Hanna Erdmann
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Catharina Arnold-Schrauf
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Maxine Swallow
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Melanie Guderian
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Freyja Krull
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | | | - Peyman Ghorbani
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Jochen Behrends
- Core Facility Fluorescence Cytometry, Research Center Borstel, Borstel, Germany
| | - Wolf-Rainer Abraham
- Department of Chemical Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Tim D Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Luciana Berod
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| |
Collapse
|
66
|
Halwani R, Vazquez-Tello A, Kenana R, Al-Otaibi M, Alhasan KA, Shakoor Z, Al-Muhsen S. Association of IL-13 rs20541 and rs1295686 variants with symptomatic asthma in a Saudi Arabian population. J Asthma 2017; 55:1157-1165. [PMID: 29211635 DOI: 10.1080/02770903.2017.1400047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Interleukin 13 (IL-13) plays a critical pro-inflammatory role in asthma. Several single nucleotide polymorphisms (SNPs) are associated with asthma susceptibility in specific populations; however, further replicative studies in other ethnic groups are mandatory. METHODS The association between IL-13 SNPs rs762534, rs20541, rs1295686, and rs1800925 (risk alleles A, A, T, and A, respectively) and asthma predisposition in a Saudi Arabian cohort was examined via a case-control cross-sectional study. RESULTS The frequencies of alleles between asthmatics and control populations were significantly different for rs20541 and rs1295686 SNPs (p < 0.001), whereas the frequencies of genotypes between asthmatics and controls were significantly different only for rs20541. The association of the risk (minor) alleles with asthma was examined using the dominant genetic model. Individuals with at least one copy of the risk alleles A (for rs20541) and T (for rs1295686) had significantly greater odds of being asthmatic (OR = 2.13, 95% CI = 1.39-3.26, p < 0.0001; OR = 1.69, 95% CI = 1.12-2.54, p = 0.008) relative to their most common homozygous genotypes. On the other hand, the minor A alleles for rs762534 and rs1800925 were not significantly associated with asthma risk. Regarding haplotype association analysis, individuals with at least one copy of the minor "risk" allele for both rs20541 and rs1295686 (CATG and CATA, respectively) had greater odds of being asthmatic relative to CGCG haplotype; however, this trend was not statistically significant (p > 0.3). CONCLUSIONS IL-13 minor T and A alleles for rs1295686 and rs20541, respectively, were associated with significantly higher risk of asthma in the Saudi Arabian population.
Collapse
Affiliation(s)
- Rabih Halwani
- a Immunology Research Laboratory and Asthma Research Chair, College of Medicine , King Saud University , Riyadh , Saudi Arabia.,b Department of Pediatrics , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Alejandro Vazquez-Tello
- a Immunology Research Laboratory and Asthma Research Chair, College of Medicine , King Saud University , Riyadh , Saudi Arabia
| | - Rosan Kenana
- a Immunology Research Laboratory and Asthma Research Chair, College of Medicine , King Saud University , Riyadh , Saudi Arabia
| | - Maram Al-Otaibi
- c Department of Pathology , King Khalid University Hospital, College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Khalid A Alhasan
- b Department of Pediatrics , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Zahid Shakoor
- c Department of Pathology , King Khalid University Hospital, College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Saleh Al-Muhsen
- a Immunology Research Laboratory and Asthma Research Chair, College of Medicine , King Saud University , Riyadh , Saudi Arabia.,b Department of Pediatrics , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| |
Collapse
|
67
|
Horn V, Triantafyllopoulou A. DNA damage signaling and polyploid macrophages in chronic inflammation. Curr Opin Immunol 2017; 50:55-63. [PMID: 29202328 DOI: 10.1016/j.coi.2017.11.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 10/30/2017] [Accepted: 11/17/2017] [Indexed: 01/01/2023]
Abstract
Whole genome duplications, an important step in cancer development, also occur in the macrophage lineage in disease: large multinucleated macrophages found within compact, ordered aggregates of immune cells, called granulomas, are a well-known histologic entity. Very recent work suggests that granuloma macrophages remarkably acquire epithelial cell features and the genotoxic stress response instructs granuloma macrophage genome duplications, suggesting that granuloma macrophages and pre-malignant epithelial cells may share common mechanisms of adaptation to chronic genotoxic stress. Exploring these mechanisms is key for a better understanding of the pathogenesis of chronic inflammatory diseases. Here we review the mechanisms of macrophage polyploidization, the role of DNA damage signaling in this process and the function of polyploid macrophages, with a focus on chronic inflammation.
Collapse
Affiliation(s)
- Veronika Horn
- Department of Rheumatology and Clinical Immunology, Charité University Medical Center, D-10117 Berlin, Germany; Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Antigoni Triantafyllopoulou
- Department of Rheumatology and Clinical Immunology, Charité University Medical Center, D-10117 Berlin, Germany; Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, D-79106 Freiburg, Germany; German Rheumatism Research Center, A Leibniz Institute, D-10117 Berlin, Germany; Institute of Microbiology, Charité University Medical Center, D-12203 Berlin, Germany.
| |
Collapse
|
68
|
Lange SM, McKell MC, Schmidt SM, Hossfeld AP, Chaturvedi V, Kinder JM, McAlees JW, Lewkowich IP, Way SS, Turner J, Qualls JE. l-Citrulline Metabolism in Mice Augments CD4 + T Cell Proliferation and Cytokine Production In Vitro, and Accumulation in the Mycobacteria-Infected Lung. Front Immunol 2017; 8:1561. [PMID: 29201027 PMCID: PMC5696333 DOI: 10.3389/fimmu.2017.01561] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/31/2017] [Indexed: 12/25/2022] Open
Abstract
Activation, recruitment, and effector function of T lymphocytes are essential for control of mycobacterial infection. These processes are tightly regulated in T cells by the availability of l-arginine within the microenvironment. In turn, mycobacterial infection dampens T cell responsiveness through arginase induction in myeloid cells, promoting sequestration of l-arginine within the local milieu. Here, we show T cells can replenish intracellular l-arginine through metabolism of l-citrulline to mediate inflammatory function, allowing anti-mycobacterial T cells to overcome arginase-mediated suppression. Furthermore, T cell l-citrulline metabolism is necessary for accumulation of CD4+ T cells at the site of infection, suggesting this metabolic pathway is involved during anti-mycobacterial T cell immunity in vivo. Together, these findings establish a contribution for l-arginine synthesis by T cells during mycobacterial infection, and implicate l-citrulline as a potential immuno-nutrient to modulate host immunity.
Collapse
Affiliation(s)
- Shannon M Lange
- Laboratory of Dr. Joseph E. Qualls, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Immunology Graduate Program, University of Cincinnati/Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Melanie C McKell
- Laboratory of Dr. Joseph E. Qualls, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Immunology Graduate Program, University of Cincinnati/Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Stephanie M Schmidt
- Laboratory of Dr. Joseph E. Qualls, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Austin P Hossfeld
- Laboratory of Dr. Joanne Turner, Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Vandana Chaturvedi
- Laboratory of Dr. Sing Sing Way, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Jeremy M Kinder
- Laboratory of Dr. Sing Sing Way, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Jaclyn W McAlees
- Laboratory of Dr. Ian P. Lewkowich, Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Ian P Lewkowich
- Laboratory of Dr. Ian P. Lewkowich, Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Sing Sing Way
- Laboratory of Dr. Sing Sing Way, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Joanne Turner
- Laboratory of Dr. Joanne Turner, Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States.,Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Joseph E Qualls
- Laboratory of Dr. Joseph E. Qualls, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
69
|
Schmok E, Abad Dar M, Behrends J, Erdmann H, Rückerl D, Endermann T, Heitmann L, Hessmann M, Yoshimura A, Rose-John S, Scheller J, Schaible UE, Ehlers S, Lang R, Hölscher C. Suppressor of Cytokine Signaling 3 in Macrophages Prevents Exacerbated Interleukin-6-Dependent Arginase-1 Activity and Early Permissiveness to Experimental Tuberculosis. Front Immunol 2017; 8:1537. [PMID: 29176982 PMCID: PMC5686055 DOI: 10.3389/fimmu.2017.01537] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/27/2017] [Indexed: 01/16/2023] Open
Abstract
Suppressor of cytokine signaling 3 (SOCS3) is a feedback inhibitor of interleukin (IL)-6 signaling in macrophages. In the absence of this molecule, macrophages become extremely prone to an IL-6-dependent expression of arginase-1 (Arg1) and nitric oxide synthase (NOS)2, the prototype markers for alternative or classical macrophage activation, respectively. Because both enzymes are antipodean macrophage effector molecules in Mycobacterium tuberculosis (Mtb) infection, we assessed the relevance of SOCS3 for macrophage activation during experimental tuberculosis using macrophage-specific SOCS3-deficient (LysMcreSOCS3loxP/loxP) mice. Aerosol infection of LysMcreSOCS3loxP/loxP mice resulted in remarkably higher bacterial loads in infected lungs and exacerbated pulmonary inflammation. This increased susceptibility to Mtb infection was accompanied by enhanced levels of both classical and alternative macrophage activation. However, high Arg1 expression preceded the increased induction of NOS2 and at early time points of infection mycobacteria were mostly found in cells positive for Arg1. This sequential activation of Arg1 and NOS2 expression in LysMcreSOCS3loxP/loxP mice appears to favor the initial replication of Mtb particularly in Arg1-positive cells. Neutralization of IL-6 in Mtb-infected LysMcreSOCS3loxP/loxP mice reduced arginase activity and restored control of mycobacterial replication in LysMcreSOCS3loxP/loxP mice. Our data reveal an unexpected role of SOCS3 during experimental TB: macrophage SOCS3 restrains early expression of Arg1 and helps limit Mtb replication in resident lung macrophages, thereby limiting the growth of mycobacteria. Together, SOCS3 keeps IL-6-dependent divergent macrophage responses such as Nos2 and Arg1 expression under control and safeguard protective macrophage effector mechanisms.
Collapse
Affiliation(s)
- Erik Schmok
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Mahin Abad Dar
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Jochen Behrends
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Hanna Erdmann
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Dominik Rückerl
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Tanja Endermann
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Lisa Heitmann
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | | | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Graduate School of Medicine, Keio University, Tokyo, Japan
| | - Stefan Rose-John
- Department of Biochemistry, Christian-Albrechts-University, Kiel, Germany.,Cluster of Excellence Inflammation-at-Interfaces (Borstel-Kiel-Lübeck-Plön), Kiel, Germany
| | - Jürgen Scheller
- Medical Faculty, Institute of Biochemistry and Molecular Biology II, Heinrich-Heine-University, Düsseldorf, Germany
| | | | - Stefan Ehlers
- Cluster of Excellence Inflammation-at-Interfaces (Borstel-Kiel-Lübeck-Plön), Kiel, Germany.,Microbial Inflammation Research, Research Center Borstel, Borstel, Germany.,Molecular Inflammation Medicine, Christian-Albrechts-University, Kiel, Germany
| | - Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Christoph Hölscher
- Infection Immunology, Research Center Borstel, Borstel, Germany.,Cluster of Excellence Inflammation-at-Interfaces (Borstel-Kiel-Lübeck-Plön), Kiel, Germany
| |
Collapse
|
70
|
Shaping the niche in macrophages: Genetic diversity of the M. tuberculosis complex and its consequences for the infected host. Int J Med Microbiol 2017; 308:118-128. [PMID: 28969988 DOI: 10.1016/j.ijmm.2017.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/09/2017] [Accepted: 09/11/2017] [Indexed: 12/12/2022] Open
Abstract
Pathogenic mycobacteria of the Mycobacterium tuberculosis complex (MTBC) have co-evolved with their individual hosts and are able to transform the hostile environment of the macrophage into a permissive cellular habitat. The impact of MTBC genetic variability has long been considered largely unimportant in TB pathogenesis. Members of the MTBC can now be distinguished into three major phylogenetic groups consisting of 7 phylogenetic lineages and more than 30 so called sub-lineages/subgroups. MTBC genetic diversity indeed influences the transmissibility and virulence of clinical MTBC isolates as well as the immune response and the clinical outcome. Here we review the genetic diversity and epidemiology of MTBC strains and describe the current knowledge about the host immune response to infection with MTBC clinical isolates using human and murine experimental model systems in vivo and in vitro. We discuss the role of innate cytokines in detail and portray two in our group recently developed approaches to characterize the intracellular niches of MTBC strains. Characterizing the niches and deciphering the strategies of MTBC strains to transform an antibacterial effector cell into a permissive cellular habitat offers the opportunity to identify strain- and lineage-specific key factors which may represent targets for novel antimicrobial or host directed therapies for tuberculosis.
Collapse
|
71
|
Abstract
Type 2 immunity is characterized by the production of IL-4, IL-5, IL-9 and IL-13, and this immune response is commonly observed in tissues during allergic inflammation or infection with helminth parasites. However, many of the key cell types associated with type 2 immune responses - including T helper 2 cells, eosinophils, mast cells, basophils, type 2 innate lymphoid cells and IL-4- and IL-13-activated macrophages - also regulate tissue repair following injury. Indeed, these cell populations engage in crucial protective activity by reducing tissue inflammation and activating important tissue-regenerative mechanisms. Nevertheless, when type 2 cytokine-mediated repair processes become chronic, over-exuberant or dysregulated, they can also contribute to the development of pathological fibrosis in many different organ systems. In this Review, we discuss the mechanisms by which type 2 immunity contributes to tissue regeneration and fibrosis following injury.
Collapse
Affiliation(s)
- Richard L Gieseck
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20852, USA
| | - Mark S Wilson
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Thomas A Wynn
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20852, USA
| |
Collapse
|
72
|
Torrelles JB, Schlesinger LS. Integrating Lung Physiology, Immunology, and Tuberculosis. Trends Microbiol 2017; 25:688-697. [PMID: 28366292 PMCID: PMC5522344 DOI: 10.1016/j.tim.2017.03.007] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/03/2017] [Accepted: 03/10/2017] [Indexed: 11/17/2022]
Abstract
Lungs are directly exposed to the air, have enormous surface area, and enable gas exchange in air-breathing animals. They are constantly 'attacked' by microbes from both outside and inside and thus possess a unique, highly regulated local immune defense system which efficiently allows for microbial clearance while minimizing damaging inflammatory responses. As a prototypic host-adapted airborne pathogen, Mycobacterium tuberculosis traverses the lung and has several 'interaction points' (IPs) which it must overcome to cause infection. These interactions are critical, not only from a pathogenesis perspective but also in considering the effectiveness of therapies and vaccines in the lungs. Here we discuss emerging views on immunologic interactions occurring in the lungs for M. tuberculosis and their impact on infection and persistence.
Collapse
Affiliation(s)
- Jordi B Torrelles
- Department of Microbial Infection and Immunity, College of Medicine, and the Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA.
| | - Larry S Schlesinger
- Department of Microbial Infection and Immunity, College of Medicine, and the Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
73
|
Vyas SP, Goswami R. Striking the right immunological balance prevents progression of tuberculosis. Inflamm Res 2017; 66:1031-1056. [PMID: 28711989 DOI: 10.1007/s00011-017-1081-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/03/2017] [Accepted: 07/07/2017] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Tuberculosis (TB) caused by infection with Mycobacterium tuberculosis (Mtb) is a major burden for human health worldwide. Current standard treatments for TB require prolonged administration of antimycobacterial drugs leading to exaggerated inflammation and tissue damage. This can result in the reactivation of latent TB culminating in TB progression. Thus, there is an unmet need to develop therapies that would shorten the duration of anti-TB treatment and to induce optimal protective immune responses to control the spread of mycobacterial infection with minimal lung pathology. FINDINGS Granulomata is the hallmark structure formed by the organized accumulation of immune cells including macrophages, natural killer cells, dendritic cells, neutrophils, T cells, and B cells to the site of Mtb infection. It safeguards the host by containing Mtb in latent form. However, granulomata can undergo caseation and contribute to the reactivation of latent TB, if the immune responses developed to fight mycobacterial infection are not properly controlled. Thus, an optimal balance between innate and adaptive immune cells might play a vital role in containing mycobacteria in latent form for prolonged periods and prevent the spread of Mtb infection from one individual to another. CONCLUSION Optimal and well-regulated immune responses against Mycobacterium tuberculosis may help to prevent the reactivation of latent TB. Moreover, therapies targeting balanced immune responses could help to improve treatment outcomes among latently infected TB patients and thereby limit the dissemination of mycobacterial infection.
Collapse
Affiliation(s)
| | - Ritobrata Goswami
- School of Bio Science, IIT Kharagpur, Kharagpur, West Bengal, 721302, India.
| |
Collapse
|
74
|
Abstract
Tuberculosis remains one of the greatest threats to human health. The causative bacterium, Mycobacterium tuberculosis, is acquired by the respiratory route. It is exquisitely adapted to humans and is a prototypic intracellular pathogen of macrophages, with alveolar macrophages being the primary conduit of infection and disease. However, M. tuberculosis bacilli interact with and are affected by several soluble and cellular components of the innate immune system which dictate the outcome of primary infection, most commonly a latently infected healthy human host, in whom the bacteria are held in check by the host immune response within the confines of tissue granuloma, the host histopathologic hallmark. Such individuals can develop active TB later in life with impairment in the immune system. In contrast, in a minority of infected individuals, the early host immune response fails to control bacterial growth, and progressive granulomatous disease develops, facilitating spread of the bacilli via infectious aerosols. The molecular details of the M. tuberculosis-host innate immune system interaction continue to be elucidated, particularly those occurring within the lung. However, it is clear that a number of complex processes are involved at the different stages of infection that may benefit either the bacterium or the host. In this article, we describe a contemporary view of the molecular events underlying the interaction between M. tuberculosis and a variety of cellular and soluble components and processes of the innate immune system.
Collapse
|
75
|
Meunier I, Kaufmann E, Downey J, Divangahi M. Unravelling the networks dictating host resistance versus tolerance during pulmonary infections. Cell Tissue Res 2017; 367:525-536. [PMID: 28168323 PMCID: PMC7088083 DOI: 10.1007/s00441-017-2572-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 01/05/2017] [Indexed: 12/19/2022]
Abstract
The appearance of single cell microorganisms on earth dates back to more than 3.5 billion years ago, ultimately leading to the development of multicellular organisms approximately 3 billion years later. The evolutionary burst of species diversity and the “struggle for existence”, as proposed by Darwin, generated a complex host defense system. Host survival during infection in vital organs, such as the lung, requires a delicate balance between host defense, which is essential for the detection and elimination of pathogens and host tolerance, which is critical for minimizing collateral tissue damage. Whereas the cellular and molecular mechanisms of host defense against many invading pathogens have been extensively studied, our understanding of host tolerance as a key mechanism in maintaining host fitness is extremely limited. This may also explain why current therapeutic and preventive approaches targeting only host defense mechanisms have failed to provide full protection against severe infectious diseases, including pulmonary influenza virus and Mycobacterium tuberculosis infections. In this review, we aim to outline various host strategies of resistance and tolerance for effective protection against acute or chronic pulmonary infections.
Collapse
Affiliation(s)
- Isabelle Meunier
- Department of Medicine, Department of Microbiology & Immunology, and Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada
| | - Eva Kaufmann
- Department of Medicine, Department of Microbiology & Immunology, and Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada
| | - Jeffrey Downey
- Department of Medicine, Department of Microbiology & Immunology, and Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada
| | - Maziar Divangahi
- Department of Medicine, Department of Microbiology & Immunology, and Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada. .,RI-MUHC, Centre for Translational Biology, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Block E (EM3.2248), Montreal, Quebec, H4A 3J1, Canada.
| |
Collapse
|
76
|
Moreira-Teixeira L, Sousa J, McNab FW, Torrado E, Cardoso F, Machado H, Castro F, Cardoso V, Gaifem J, Wu X, Appelberg R, Castro AG, O'Garra A, Saraiva M. Type I IFN Inhibits Alternative Macrophage Activation during Mycobacterium tuberculosis Infection and Leads to Enhanced Protection in the Absence of IFN-γ Signaling. THE JOURNAL OF IMMUNOLOGY 2016; 197:4714-4726. [PMID: 27849167 PMCID: PMC5133670 DOI: 10.4049/jimmunol.1600584] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 10/17/2016] [Indexed: 02/01/2023]
Abstract
Tuberculosis causes ∼1.5 million deaths every year, thus remaining a leading cause of death from infectious diseases in the world. A growing body of evidence demonstrates that type I IFN plays a detrimental role in tuberculosis pathogenesis, likely by interfering with IFN-γ–dependent immunity. In this article, we reveal a novel mechanism by which type I IFN may confer protection against Mycobacterium tuberculosis infection in the absence of IFN-γ signaling. We show that production of type I IFN by M. tuberculosis–infected macrophages induced NO synthase 2 and inhibited arginase 1 gene expression. In vivo, absence of both type I and type II IFN receptors led to strikingly increased levels of arginase 1 gene expression and protein activity in infected lungs, characteristic of alternatively activated macrophages. This correlated with increased lung bacterial burden and pathology and decreased survival compared with mice deficient in either receptor. Increased expression of other genes associated with alternatively activated macrophages, as well as increased expression of Th2-associated cytokines and decreased TNF expression, were also observed. Thus, in the absence of IFN-γ signaling, type I IFN suppressed the switching of macrophages from a more protective classically activated phenotype to a more permissive alternatively activated phenotype. Together, our data support a model in which suppression of alternative macrophage activation by type I IFN during M. tuberculosis infection, in the absence of IFN-γ signaling, contributes to host protection.
Collapse
Affiliation(s)
- Lúcia Moreira-Teixeira
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, and Life and Health Sciences Research Institute/3B's PT Government Associate Laboratory, 4710 Braga/Guimarães, Portugal; .,Laboratory of Immunoregulation and Infection, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Jeremy Sousa
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, and Life and Health Sciences Research Institute/3B's PT Government Associate Laboratory, 4710 Braga/Guimarães, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200 Porto, Portugal
| | - Finlay W McNab
- Laboratory of Immunoregulation and Infection, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Egídio Torrado
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, and Life and Health Sciences Research Institute/3B's PT Government Associate Laboratory, 4710 Braga/Guimarães, Portugal
| | - Filipa Cardoso
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, and Life and Health Sciences Research Institute/3B's PT Government Associate Laboratory, 4710 Braga/Guimarães, Portugal
| | - Henrique Machado
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, and Life and Health Sciences Research Institute/3B's PT Government Associate Laboratory, 4710 Braga/Guimarães, Portugal
| | - Flávia Castro
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, and Life and Health Sciences Research Institute/3B's PT Government Associate Laboratory, 4710 Braga/Guimarães, Portugal
| | - Vânia Cardoso
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, and Life and Health Sciences Research Institute/3B's PT Government Associate Laboratory, 4710 Braga/Guimarães, Portugal
| | - Joana Gaifem
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, and Life and Health Sciences Research Institute/3B's PT Government Associate Laboratory, 4710 Braga/Guimarães, Portugal
| | - Xuemei Wu
- Laboratory of Immunoregulation and Infection, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Rui Appelberg
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200 Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050 Porto, Portugal
| | - António Gil Castro
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, and Life and Health Sciences Research Institute/3B's PT Government Associate Laboratory, 4710 Braga/Guimarães, Portugal
| | - Anne O'Garra
- Laboratory of Immunoregulation and Infection, The Francis Crick Institute, London NW1 1AT, United Kingdom.,National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London SW3 6NP, United Kingdom; and
| | - Margarida Saraiva
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, and Life and Health Sciences Research Institute/3B's PT Government Associate Laboratory, 4710 Braga/Guimarães, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200 Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150 Porto, Portugal
| |
Collapse
|
77
|
Herrtwich L, Nanda I, Evangelou K, Nikolova T, Horn V, Sagar, Erny D, Stefanowski J, Rogell L, Klein C, Gharun K, Follo M, Seidl M, Kremer B, Münke N, Senges J, Fliegauf M, Aschman T, Pfeifer D, Sarrazin S, Sieweke MH, Wagner D, Dierks C, Haaf T, Ness T, Zaiss MM, Voll RE, Deshmukh SD, Prinz M, Goldmann T, Hölscher C, Hauser AE, Lopez-Contreras AJ, Grün D, Gorgoulis V, Diefenbach A, Henneke P, Triantafyllopoulou A. DNA Damage Signaling Instructs Polyploid Macrophage Fate in Granulomas. Cell 2016; 167:1264-1280.e18. [PMID: 28084216 DOI: 10.1016/j.cell.2016.09.054] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 07/26/2016] [Accepted: 09/28/2016] [Indexed: 10/20/2022]
Abstract
Granulomas are immune cell aggregates formed in response to persistent inflammatory stimuli. Granuloma macrophage subsets are diverse and carry varying copy numbers of their genomic information. The molecular programs that control the differentiation of such macrophage populations in response to a chronic stimulus, though critical for disease outcome, have not been defined. Here, we delineate a macrophage differentiation pathway by which a persistent Toll-like receptor (TLR) 2 signal instructs polyploid macrophage fate by inducing replication stress and activating the DNA damage response. Polyploid granuloma-resident macrophages formed via modified cell divisions and mitotic defects and not, as previously thought, by cell-to-cell fusion. TLR2 signaling promoted macrophage polyploidy and suppressed genomic instability by regulating Myc and ATR. We propose that, in the presence of persistent inflammatory stimuli, pathways previously linked to oncogene-initiated carcinogenesis instruct a long-lived granuloma-resident macrophage differentiation program that regulates granulomatous tissue remodeling.
Collapse
Affiliation(s)
- Laura Herrtwich
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Center of Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Indrajit Nanda
- Institute of Human Genetics, Biozentrum, Am Hubland, 97074 Würzburg, Germany
| | - Konstantinos Evangelou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Teodora Nikolova
- Institute of Toxicology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Veronika Horn
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Sagar
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Daniel Erny
- Institute of Neuropathology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Jonathan Stefanowski
- Immune Dynamics, Charité Universitätsmedizin and Deutsches Rheumaforschungszentrum, 10117 Berlin, Germany
| | - Leif Rogell
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; Institute of Medical Microbiology and Hygiene, University of Mainz Medical Center, 55131 Mainz, Germany; Research Center for Immunology and Immunotherapy, University of Mainz Medical Center, 55131 Mainz, Germany
| | - Claudius Klein
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Kourosh Gharun
- Center of Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Marie Follo
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Maximilian Seidl
- Department of Pathology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Bernhard Kremer
- Center of Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Nikolas Münke
- Center of Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Julia Senges
- Center of Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Manfred Fliegauf
- Center of Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Tom Aschman
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | | | - Michael H Sieweke
- Aix-Marseille Univ, CNRS, INSERM, CIML, 13288 Marseille, France; Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft (MDC), 13125 Berlin, Germany
| | - Dirk Wagner
- Center of Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Division of Infectious Diseases, Department of Internal Medicine 2, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Christine Dierks
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Thomas Haaf
- Institute of Human Genetics, Biozentrum, Am Hubland, 97074 Würzburg, Germany
| | - Thomas Ness
- Eye Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Mario M Zaiss
- Department of Internal Medicine 3, Rheumatology and Immunology, University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Sachin D Deshmukh
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79106 Freiburg, Germany
| | - Torsten Goldmann
- Department of Pathology, Schleswig-Holstein University Hospital, Campus Lübeck and Research Center Borstel, 23845 Borstel, Germany
| | - Christoph Hölscher
- Division of Infection Immunology, Research Center Borstel, 23845 Borstel, Germany; Cluster of Excellence, Inflammation at Interfaces (Borstel-Kiel-Lübeck-Plön), 24118 Kiel, Germany; German Centre for Infection Research, 23845 Borstel, Germany
| | - Anja E Hauser
- Immune Dynamics, Charité Universitätsmedizin and Deutsches Rheumaforschungszentrum, 10117 Berlin, Germany
| | - Andres J Lopez-Contreras
- Center for Chromosome Stability, Department of Cellular and Molecular Medicine, Panum Institute, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Dominic Grün
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Vassilis Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 115 27 Athens, Greece; Faculty Institute of Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4QL, UK; Biomedical Research Foundation, Academy of Athens, 115 27 Athens, Greece; Department of Pathophysiology School of Medicine, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Andreas Diefenbach
- Institute of Medical Microbiology and Hygiene, University of Mainz Medical Center, 55131 Mainz, Germany; Research Center for Immunology and Immunotherapy, University of Mainz Medical Center, 55131 Mainz, Germany.
| | - Philipp Henneke
- Center of Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Center for Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Antigoni Triantafyllopoulou
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Center of Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.
| |
Collapse
|
78
|
Prosser G, Brandenburg J, Reiling N, Barry CE, Wilkinson RJ, Wilkinson KA. The bacillary and macrophage response to hypoxia in tuberculosis and the consequences for T cell antigen recognition. Microbes Infect 2016; 19:177-192. [PMID: 27780773 PMCID: PMC5335906 DOI: 10.1016/j.micinf.2016.10.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 10/06/2016] [Indexed: 12/11/2022]
Abstract
Mycobacterium tuberculosis is a facultative anaerobe and its characteristic pathological hallmark, the granuloma, exhibits hypoxia in humans and in most experimental models. Thus the host and bacillary adaptation to hypoxia is of central importance in understanding pathogenesis and thereby to derive new drug treatments and vaccines.
Collapse
Affiliation(s)
- Gareth Prosser
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, 20892, United States
| | - Julius Brandenburg
- Microbial Interface Biology, Priority Research Area Infections, Forschungszentrum Borstel, Leibniz Center for Medicine and Biosciences, Parkallee 1-40, D-23845, Borstel, Germany
| | - Norbert Reiling
- Microbial Interface Biology, Priority Research Area Infections, Forschungszentrum Borstel, Leibniz Center for Medicine and Biosciences, Parkallee 1-40, D-23845, Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck, Borstel, Germany
| | - Clifton Earl Barry
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, 20892, United States; Clinical Infectious Diseases Research Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, 7925, South Africa
| | - Robert J Wilkinson
- Clinical Infectious Diseases Research Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, 7925, South Africa; The Francis Crick Institute, London, NW1 2AT, United Kingdom; Department of Medicine, Imperial College, London, W2 1PG, United Kingdom.
| | - Katalin A Wilkinson
- Clinical Infectious Diseases Research Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, 7925, South Africa; The Francis Crick Institute, London, NW1 2AT, United Kingdom
| |
Collapse
|
79
|
Domingo-Gonzalez R, Prince O, Cooper A, Khader SA. Cytokines and Chemokines in Mycobacterium tuberculosis Infection. Microbiol Spectr 2016; 4:10.1128/microbiolspec.TBTB2-0018-2016. [PMID: 27763255 PMCID: PMC5205539 DOI: 10.1128/microbiolspec.tbtb2-0018-2016] [Citation(s) in RCA: 266] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Indexed: 02/06/2023] Open
Abstract
Chemokines and cytokines are critical for initiating and coordinating the organized and sequential recruitment and activation of cells into Mycobacterium tuberculosis-infected lungs. Correct mononuclear cellular recruitment and localization are essential to ensure control of bacterial growth without the development of diffuse and damaging granulocytic inflammation. An important block to our understanding of TB pathogenesis lies in dissecting the critical aspects of the cytokine/chemokine interplay in light of the conditional role these molecules play throughout infection and disease development. Much of the data highlighted in this review appears at first glance to be contradictory, but it is the balance between the cytokines and chemokines that is critical, and the "goldilocks" (not too much and not too little) phenomenon is paramount in any discussion of the role of these molecules in TB. Determination of how the key chemokines/cytokines and their receptors are balanced and how the loss of that balance can promote disease is vital to understanding TB pathogenesis and to identifying novel therapies for effective eradication of this disease.
Collapse
Affiliation(s)
| | - Oliver Prince
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63130
| | - Andrea Cooper
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester LE1 7RH, United Kingdom
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63130
| |
Collapse
|
80
|
Nam SJ, Jeong JY, Jang TW, Jung MH, Chun BK, Cha HJ, Oak CH. Neuron-specific enolase as a novel biomarker reflecting tuberculosis activity and treatment response. Korean J Intern Med 2016; 31:694-702. [PMID: 27271274 PMCID: PMC4939508 DOI: 10.3904/kjim.2015.407] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/12/2016] [Accepted: 03/21/2016] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/AIMS It is not clear which tests are indicative of the activity and severity of tuberculosis (TB). This study aimed to investigate the predictive value of neuron-specific enolase (NSE) and to determine the origin of NSE in TB patients. METHODS A single-center retrospective analysis was conducted on newly diagnosed TB patients between January and December 2010. Patients were categorized into one of two disease groups (focal segmental or extensive) based on chest X-ray. Pre- and post-treatment NSE concentrations were evaluated. To determine the origin of serum NSE concentration, NSE staining was compared with macrophage-specific CD68 staining in lung tissues and with a tissue microarray using immunohistochemistry and immunofluorescence. RESULTS A total of 60 newly diagnosed TB patients were analyzed. In TB patients, NSE serum concentration was significantly increased and NSE level decreased after treatment (p < 0.001). In proportion to serum high-sensitivity C-reactive protein concentration, the mean serum concentration of NSE in the extensive group (25.12 ng/mL) was significantly higher than that in the focal segmental group (20.23 ng/mL, p = 0.04). Immunohistochemical staining revealed a large number of macrophages that stained positively for both NSE and CD68 in TB tissues. In addition, NSE signals mostly co-localized with CD68 signals in the tissue microarray of TB patients. CONCLUSIONS Our results suggest that NSE may be a practical parameter that can be used to monitor TB activity and treatment response. Elevated serum NSE level originates, at least in part, from macrophages in granulomatous lesions.
Collapse
Affiliation(s)
- Sung-Jin Nam
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Changwon, Korea
| | - Jee-Yeong Jeong
- Department of Biochemistry, Kosin University College of Medicine, Busan, Korea
- Cancer Research Institute, Kosin University College of Medicine, Busan, Korea
| | - Tae-Won Jang
- Department of Internal Medicine, Kosin University College of Medicine, Busan, Korea
| | - Mann-Hong Jung
- Department of Internal Medicine, Kosin University College of Medicine, Busan, Korea
| | - Bong-Kwon Chun
- Department of Pathology, Kosin University College of Medicine, Busan, Korea
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan, Korea
| | - Chul-Ho Oak
- Department of Internal Medicine, Kosin University College of Medicine, Busan, Korea
- Correspondence to Chul-Ho Oak, M.D. Department of Internal Medicine, Kosin University College of Medicine, 262 Gamcheon-ro, Seo-gu, Busan 49267, Korea Tel: +82-51-990-6104 Fax: +82-51-990-3010 E-mail:
| |
Collapse
|
81
|
Zumla A, Rao M, Dodoo E, Maeurer M. Potential of immunomodulatory agents as adjunct host-directed therapies for multidrug-resistant tuberculosis. BMC Med 2016; 14:89. [PMID: 27301245 PMCID: PMC4908783 DOI: 10.1186/s12916-016-0635-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/02/2016] [Indexed: 01/15/2023] Open
Abstract
Treatment of multidrug-resistant tuberculosis (MDR-TB) is extremely challenging due to the virulence of the etiologic strains of Mycobacterium tuberculosis (M. tb), the aberrant host immune responses and the diminishing treatment options with TB drugs. New treatment regimens incorporating therapeutics targeting both M. tb and host factors are urgently needed to improve the clinical management outcomes of MDR-TB. Host-directed therapies (HDT) could avert destructive tuberculous lung pathology, facilitate eradication of M. tb, improve survival and prevent long-term functional disability. In this review we (1) discuss the use of HDT for cancer and other infections, drawing parallels and the precedent they set for MDR-TB treatment, (2) highlight preclinical studies of pharmacological agents commonly used in clinical practice which have HDT potential, and (3) outline developments in cellular therapy to promote clinically beneficial immunomodulation to improve treatment outcomes in patients with pulmonary MDR-TB. The use of HDTs as adjuncts to MDR-TB therapy requires urgent evaluation.
Collapse
Affiliation(s)
- Alimuddin Zumla
- Division of Infection and Immunity, University College London, and NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, UK
| | - Martin Rao
- F79, Therapeutic Immunology (TIM) division, Department of Laboratory Medicine (LABMED), Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
| | - Ernest Dodoo
- F79, Therapeutic Immunology (TIM) division, Department of Laboratory Medicine (LABMED), Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
| | - Markus Maeurer
- F79, Therapeutic Immunology (TIM) division, Department of Laboratory Medicine (LABMED), Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden. .,Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
82
|
Ma J, Jung BG, Yi N, Samten B. Early Secreted Antigenic Target of 6 kDa ofMycobacterium tuberculosisStimulates Macrophage Chemoattractant Protein-1 Production by Macrophages and Its Regulation by p38 Mitogen-Activated Protein Kinases and Interleukin-4. Scand J Immunol 2016; 84:39-48. [DOI: 10.1111/sji.12447] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 05/04/2016] [Indexed: 11/29/2022]
Affiliation(s)
- J. Ma
- Department of Pulmonary Immunology; University of Texas Health Science Center at Tyler; Tyler TX USA
| | - B-G. Jung
- Department of Pulmonary Immunology; University of Texas Health Science Center at Tyler; Tyler TX USA
| | - N. Yi
- Department of Pulmonary Immunology; University of Texas Health Science Center at Tyler; Tyler TX USA
| | - B. Samten
- Department of Pulmonary Immunology; University of Texas Health Science Center at Tyler; Tyler TX USA
| |
Collapse
|
83
|
Chen KY, Feng PH, Chang CC, Chen TT, Chuang HC, Lee CN, Su CL, Lin LY, Lee KY. Novel biomarker analysis of pleural effusion enhances differentiation of tuberculous from malignant pleural effusion. Int J Gen Med 2016; 9:183-9. [PMID: 27354819 PMCID: PMC4910680 DOI: 10.2147/ijgm.s100237] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Lymphocytic pleurisy is commonly observed in tuberculosis and cancer. Noninvasive biomarkers are needed to distinguish tuberculous pleural effusion (TPE) from malignant pleural effusion (MPE) because current clinical diagnostic procedures are often invasive. We identified immune response biomarkers that can discriminate between TPE and MPE. Fourteen pleural effusion biomarkers were compared in 22 MPE patients and five TPE patients. Of the innate immunity biomarkers, the median levels of interleukin (IL)-1β and interferon-induced protein-10 (IP-10) were higher in TPE patients than in MPE patients (P<0.05 and P<0.01, respectively). Of the adaptive immunity biomarkers, the median levels of IL-13 and interferon-γ (IFN-γ) were higher in TPE patients than in MPE patients (P<0.05). In addition, the levels of basic fibroblast growth factor were higher in MPE patients than in TPE patients (P<0.05). Receiver operator characteristic analysis of these biomarkers was performed, resulting in the highest area under the curve (AUC) for IP-10 (AUC =0.95, 95% confidence interval, P<0.01), followed by IL-13 (AUC =0.86, 95% confidence interval, P<0.05). Our study shows that five biomarkers (IL-1β, IP-10, IFN-γ, IL-13, and basic fibroblast growth factor) have a potential diagnostic role in differentiating TPE from MPE, particularly in lung cancer-related MPE.
Collapse
Affiliation(s)
- Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, Republic of China; Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, Republic of China; Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Chih-Cheng Chang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Tzu-Tao Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Hsiao-Chi Chuang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, Republic of China; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Chun-Nin Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, Republic of China; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Chien-Ling Su
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, Republic of China; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Lian-Yu Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, Republic of China
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, Republic of China; Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
| |
Collapse
|
84
|
Muciño G, Castro-Obregón S, Hernandez-Pando R, Del Rio G. Autophagy as a target for therapeutic uses of multifunctional peptides. IUBMB Life 2016; 68:259-67. [PMID: 26968336 DOI: 10.1002/iub.1483] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 01/17/2016] [Indexed: 12/21/2022]
Abstract
The emergence of complex diseases is promoting a change from one-target to multitarget drugs and peptides are ideal molecules to fulfill this polypharmacologic role. Here we review current status in the design of polypharmacological peptides aimed to treat complex diseases, focusing on tuberculosis. In this sense, combining multiple activities in single molecules is a two-sided sword, as both positive and negative side effects might arise. These polypharmacologic compounds may be directed to regulate autophagy, a catabolic process that enables cells to eliminate intracellular microbes (xenophagy), such as Mycobacterium tuberculosis (MBT). Here we review some strategies to control MBT infection and propose that a peptide combining both antimicrobial and pro-autophagic activities would have a greater potential to limit MBT infection. This endeavor may complement the knowledge gained in understanding the mechanism of action of antibiotics and may lead to the design of better polypharmacological peptides to treat complex diseases such as tuberculosis.
Collapse
Affiliation(s)
- Gabriel Muciño
- Department of Neurodevelopment and Physiology, Instituto De Fisiología Celular, UNAM, México, D.F, México
| | - Susana Castro-Obregón
- Department of Neurodevelopment and Physiology, Instituto De Fisiología Celular, UNAM, México, D.F, México
| | - Rogelio Hernandez-Pando
- Experimental Pathology Section, Department of Pathology, National Institute of Medical Sciences and Nutrition "Salvador Zubirán,", Mexico
| | - Gabriel Del Rio
- Department of Biochemistry and Structural Biology, Instituto De Fisiología Celular, UNAM, México, D.F, México
| |
Collapse
|
85
|
Donnachie E, Fedotova EP, Hwang SA. Trehalose 6,6-Dimycolate from Mycobacterium tuberculosis Induces Hypercoagulation. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1221-33. [PMID: 26968340 DOI: 10.1016/j.ajpath.2015.12.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 11/09/2015] [Accepted: 12/18/2015] [Indexed: 01/10/2023]
Abstract
Tuberculosis (TB) remains a global health concern. Trehalose 6'6-dimycolate (TDM) activates innate inflammation and likely also stimulates chronic inflammation observed during disease progression. Noninfectious models using purified TDM oil/water emulsions elicit pathologic findings observed in patients with TB. We introduce a new TDM model that promotes inflammatory lung pathologic findings and vascular occlusion and hemorrhage. C57BL/6 and BALB/c mice were injected with 10 μg of i.p. TDM in light mineral oil (TDM-IP). At day 7, another injection of 10 μg of i.v. TDM in oil/water emulsion was given (TDM-IV). The i.p./i.v. TDM (TDM-IVIP) group was compared with mice injected once with i.v. or i.p. TDM. The responses to TDM-IP, TDM-IV, or TDM-IPIV were consistent between mouse strains. Mice that received TDM-IV and TDM-IPIV had inflammatory pathologic findings with increases in inflammatory and T-cell cytokines, and the TDM-IPIV group had further enhancement of IL-10 and granulocyte-macrophage colony-stimulating factor. The TDM-IPIV group had increased CD4(+) T cells in lung tissue, significantly increased coagulation, decreased clot formation time, and increased maximum clot firmness. Masson's trichrome staining revealed increased deposition of collagen in the occluded vasculature. TDM-IPIV promotes a hypercoagulopathy state, independent of inflammation. This new model argues that TDM is sufficient to generate the hypercoagulopathy observed in patients with TB.
Collapse
Affiliation(s)
- Elizabeth Donnachie
- Gulf States Hemophilia and Thrombophilia Center, Department of Pediatrics, University of Texas Medical School at Houston, Houston, Texas
| | - Elena P Fedotova
- Department of Anatomic Pathology, St. Petersburg State Pediatric Medical University, St. Petersburg, Russia
| | - Shen-An Hwang
- Department of Pathology and Laboratory Medicine, University of Texas Medical School at Houston, Houston, Texas.
| |
Collapse
|
86
|
Qualls JE, Murray PJ. Immunometabolism within the tuberculosis granuloma: amino acids, hypoxia, and cellular respiration. Semin Immunopathol 2016; 38:139-52. [PMID: 26490974 PMCID: PMC4779414 DOI: 10.1007/s00281-015-0534-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/01/2015] [Indexed: 02/04/2023]
Abstract
Tuberculosis (TB) granulomas are compact, organized agglomerations of infected and uninfected macrophages, T cells, neutrophils, and other immune cells. Within the granuloma, several unique metabolic adaptations occur to modify the behavior of immune cells, potentially favoring bacterial persistence balanced with protection against immunopathology. These include the induction of arginase-1 in macrophages to temper nitric oxide (NO) production and block T cell proliferation, inhibition of oxygen-requiring NO production in hypoxic regions, and induction of tryptophan-degrading enzymes that modify T cell proliferation and function. The spatial and time-dependent organization of granulomas further influences immunometabolism, for example through lactate production by activated macrophages, which can induce arginase-1. Although complex, the metabolic changes in and around TB granulomas can be potentially modified by host-directed therapies. While elimination of the TB bacilli is often the goal of any anti-TB therapy, host-directed approaches must also account for the possibility of immunopathologic damage to the lung.
Collapse
Affiliation(s)
- Joseph E Qualls
- Department of Pediatrics, Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Peter J Murray
- Department of Infectious Diseases and Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
87
|
A Mutation in IL4RA Is Associated with the Degree of Pathology in Human TB Patients. Mediators Inflamm 2016; 2016:4245028. [PMID: 26977119 PMCID: PMC4764744 DOI: 10.1155/2016/4245028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/12/2016] [Indexed: 12/16/2022] Open
Abstract
The contribution of interleukin- (IL-) 4 receptor-alpha- (Rα-) dependent events in the pathogenesis of tuberculosis (TB) is controversial. We have recently shown IL-13 overexpression in mice to cause recrudescent Mtb replication and centrally necrotizing granulomas strongly resembling pathology of human TB. A deletion of IL-4Rα completely abrogates TB tissue pathology in these mice. To validate our results in human TB patients, we here determined the association of distinct variants of the IL4, IL13, IL4RA, IL13RA1, and IL13RA2 genes with cavity formation in a large Ghanaian cohort of HIV-negative individuals with newly diagnosed pulmonary TB. In fact, the structural variant of the IL4RA I50V, previously shown to result in enhanced signal transduction, was significantly associated with greater cavity size, and a variant of IL13RA2 was associated with disease in females. To evaluate whether the human-like TB pathology in IL-13-overexpressing mice is specifically mediated through the IL-4Rα subunit, we analyzed IL-13 transgenic mice with a genetic ablation of the IL-4Rα. In these mice, the IL-13-mediated increased susceptibility, human-like pathology of collagen deposition around centrally necrotizing granulomas, and alternative macrophage activation were abolished. Together, our genetic association study in human TB patients further supports the assumption that IL-13/IL-4Rα-dependent mechanisms are involved in mediating tissue pathology of human TB.
Collapse
|
88
|
Mouse models of human TB pathology: roles in the analysis of necrosis and the development of host-directed therapies. Semin Immunopathol 2015; 38:221-37. [PMID: 26542392 PMCID: PMC4779126 DOI: 10.1007/s00281-015-0538-9] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/22/2015] [Indexed: 12/28/2022]
Abstract
A key aspect of TB pathogenesis that maintains Mycobacterium tuberculosis in the human population is the ability to cause necrosis in pulmonary lesions. As co-evolution shaped M. tuberculosis (M.tb) and human responses, the complete TB disease profile and lesion manifestation are not fully reproduced by any animal model. However, animal models are absolutely critical to understand how infection with virulent M.tb generates outcomes necessary for the pathogen transmission and evolutionary success. In humans, a wide spectrum of TB outcomes has been recognized based on clinical and epidemiological data. In mice, there is clear genetic basis for susceptibility. Although the spectra of human and mouse TB do not completely overlap, comparison of human TB with mouse lesions across genetically diverse strains firmly establishes points of convergence. By embracing the genetic heterogeneity of the mouse population, we gain tremendous advantage in the quest for suitable in vivo models. Below, we review genetically defined mouse models that recapitulate a key element of M.tb pathogenesis—induction of necrotic TB lesions in the lungs—and discuss how these models may reflect TB stratification and pathogenesis in humans. The approach ensures that roles that mouse models play in basic and translational TB research will continue to increase allowing researchers to address fundamental questions of TB pathogenesis and bacterial physiology in vivo using this well-defined, reproducible, and cost-efficient system. Combination of the new generation mouse models with advanced imaging technologies will also allow rapid and inexpensive assessment of experimental vaccines and therapies prior to testing in larger animals and clinical trials.
Collapse
|
89
|
Rapovy SM, Zhao J, Bricker RL, Schmidt SM, Setchell KDR, Qualls JE. Differential Requirements for L-Citrulline and L-Arginine during Antimycobacterial Macrophage Activity. THE JOURNAL OF IMMUNOLOGY 2015; 195:3293-300. [PMID: 26311904 PMCID: PMC6432794 DOI: 10.4049/jimmunol.1500800] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 07/28/2015] [Indexed: 12/26/2022]
Abstract
Microbicidal NO production is reliant on inducible NO synthase-mediated L-arginine metabolism in macrophages (MΦs). However, L-arginine supply can be restricted by arginase activity, resulting in inefficient NO output and inhibition of antimicrobial MΦ function. MΦs circumvent this by converting L-citrulline to L-arginine, thereby resupplying substrate for NO production. In this article, we define the metabolic signature of mycobacteria-infected murine MΦs supplied L-arginine, L-citrulline, or both amino acids. Using liquid chromatography-tandem mass spectrometry, we determined that L-arginine synthesized from L-citrulline was less effective as a substrate for arginase-mediated L-ornithine production compared with L-arginine directly imported from the extracellular milieu. Following Mycobacterium bovis bacillus Calmette-Guérin infection and costimulation with IFN-γ, we observed that MΦ arginase activity did not inhibit production of NO derived from L-citrulline, contrary to NO inhibition witnessed when MΦs were cultured in L-arginine. Furthermore, we found that arginase-expressing MΦs preferred L-citrulline over L-arginine for the promotion of antimycobacterial activity. We expect that defining the consequences of L-citrulline metabolism in MΦs will provide novel approaches for enhancing immunity, especially in the context of mycobacterial disease.
Collapse
Affiliation(s)
- Shannon M Rapovy
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; and
| | - Junfang Zhao
- Division of Pathology and Laboratory Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Rebecca L Bricker
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; and
| | - Stephanie M Schmidt
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; and
| | - Kenneth D R Setchell
- Division of Pathology and Laboratory Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Joseph E Qualls
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; and
| |
Collapse
|
90
|
Lack of the Transcription Factor Hypoxia-Inducible Factor 1α (HIF-1α) in Macrophages Accelerates the Necrosis of Mycobacterium avium-Induced Granulomas. Infect Immun 2015; 83:3534-44. [PMID: 26099585 DOI: 10.1128/iai.00144-15] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 06/17/2015] [Indexed: 02/02/2023] Open
Abstract
The establishment of mycobacterial infection is characterized by the formation of granulomas, which are well-organized aggregates of immune cells, namely, infected macrophages. The granuloma's main function is to constrain and prevent dissemination of the mycobacteria while focusing the immune response to a limited area. In some cases these lesions can grow progressively into large granulomas which can undergo central necrosis, thereby leading to their caseation. Macrophages are the most abundant cells present in the granuloma and are known to adapt under hypoxic conditions in order to avoid cell death. Our laboratory has developed a granuloma necrosis model that mimics the human pathology of Mycobacterium tuberculosis, using C57BL/6 mice infected intravenously with a low dose of a highly virulent strain of Mycobacterium avium. In this work, a mouse strain deleted of the hypoxia inducible factor 1α (HIF-1α) under the Cre-lox system regulated by the lysozyme M gene promoter was used to determine the relevance of HIF-1α in the caseation of granulomas. The genetic ablation of HIF-1α in the myeloid lineage causes the earlier emergence of granuloma necrosis and clearly induces an impairment of the resistance against M. avium infection coincident with the emergence of necrosis. The data provide evidence that granulomas become hypoxic before undergoing necrosis through the analysis of vascularization and quantification of HIF-1α in a necrotizing mouse model. Our results show that interfering with macrophage adaptation to hypoxia, such as through HIF-1α inactivation, accelerates granuloma necrosis.
Collapse
|
91
|
Keynan Y, Rueda ZV, Aguilar Y, Trajtman A, Vélez LA. Unique cytokine and chemokine patterns in bronchoalveolar lavage are associated with specific causative pathogen among HIV infected patients with pneumonia, in Medellin, Colombia. Cytokine 2015; 73:295-301. [PMID: 25837522 DOI: 10.1016/j.cyto.2015.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/03/2015] [Accepted: 03/05/2015] [Indexed: 12/23/2022]
Abstract
We wanted to investigate the pro-inflammatory cytokine/chemokine profile associated with the etiological agents identified in HIV patients. Immunosuppressed patients admitted to two hospitals in Medellin, Colombia, with clinical and radiographic diagnosis of pneumonia were enrolled in the study. After consent, bronchoalveolar lavage (BAL) was collected for bacterial, mycobacterial and fungal diagnosis. All patients were followed for a year. A stored BAL sample was used for cytokine/chemokine detection and measurement using commercial, magnetic human cytokine bead-based 19-plex assays. Statistical analysis was performed by assigning cytokine/chemokine concentrations levels into <25 percentile (lower), 25-75 percentile (normal) and >75 percentile (higher). Principal component analysis (PCA) and Kruskal-Wallis analysis were conducted to identify the clustering of cytokines with the various infectious etiologies (fungi, Mycobacterium tuberculosis - MTB, and bacteria). Average age of patients was 35, of whom 77% were male, and the median CD4 count of 33cells/μl. Of the 57 HIV infected patients, in-hospital mortality was 12.3% and 33% died within a year of follow up. The PCA revealed increased IL-10, IL-12, IL-13, IL-17, Eotaxin, GCSF, MIP-1α, and MIP-1β concentrations to be associated with MTB infection. In patients with proven fungal infection, low concentrations of IL-1RA, IL-8, TNF-α and VEGF were identified. Bacterial infections displayed a distinct cytokine pattern and were not misclassified using the MTB or fungi cytokine patterns (p-value<0.0001). Our results indicate a unique pattern of pro-inflammatory cytokine/chemokine, allowing differentiation between bacterial and non-bacterial pathogens. Moreover, we found distinct, if imperfectly discriminatory, cytokine/chemokine patterns associated with MTB and fungal infections.
Collapse
Affiliation(s)
- Yoav Keynan
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada.
| | - Zulma V Rueda
- Facultad de Medicina, Universidad Pontificia Bolivariana, Medellin, Colombia
| | - Yudy Aguilar
- Grupo Investigador de Problemas en Enfermedades Infecciosas, Universidad de Antioquia UdeA, Medellin, Colombia
| | - Adriana Trajtman
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Lázaro A Vélez
- Grupo Investigador de Problemas en Enfermedades Infecciosas, Universidad de Antioquia UdeA, Medellin, Colombia; Hospital Universitario San Vicente Fundación, Medellin, Colombia
| |
Collapse
|
92
|
van Meijgaarden KE, Haks MC, Caccamo N, Dieli F, Ottenhoff THM, Joosten SA. Human CD8+ T-cells recognizing peptides from Mycobacterium tuberculosis (Mtb) presented by HLA-E have an unorthodox Th2-like, multifunctional, Mtb inhibitory phenotype and represent a novel human T-cell subset. PLoS Pathog 2015; 11:e1004671. [PMID: 25803478 PMCID: PMC4372528 DOI: 10.1371/journal.ppat.1004671] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/08/2015] [Indexed: 02/04/2023] Open
Abstract
Mycobacterial antigens are not exclusively presented to T-cells by classical HLA-class Ia and HLA-class II molecules, but also through alternative antigen presentation molecules such as CD1a/b/c, MR1 and HLA-E. We recently described mycobacterial peptides that are presented in HLA-E and recognized by CD8+ T-cells. Using T-cell cloning, phenotyping, microbiological, functional and RNA-expression analyses, we report here that these T-cells can exert cytolytic or suppressive functions, inhibit mycobacterial growth, yet express GATA3, produce Th2 cytokines (IL-4,-5,-10,-13) and activate B-cells via IL-4. In TB patients, Mtb specific cells were detectable by peptide-HLA-E tetramers, and IL-4 and IL-13 were produced following peptide stimulation. These results identify a novel human T-cell subset with an unorthodox, multifunctional Th2 like phenotype and cytolytic or regulatory capacities, which is involved in the human immune response to mycobacteria and demonstrable in active TB patients' blood. The results challenge the current dogma that only Th1 cells are able to inhibit Mtb growth and clearly show that Th2 like cells can strongly inhibit outgrowth of Mtb from human macrophages. These insights significantly expand our understanding of the immune response in infectious disease.
Collapse
Affiliation(s)
| | - Mariëlle C. Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Nadia Caccamo
- Central Laboratory for Advanced Diagnostic and Biomedical Research (CLADIBIOR), Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di Palermo, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory for Advanced Diagnostic and Biomedical Research (CLADIBIOR), Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di Palermo, Palermo, Italy
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
93
|
Sloot R, Schim van der Loeff MF, van Zwet EW, Haks MC, Keizer ST, Scholing M, Ottenhoff TH, Borgdorff MW, Joosten SA. Biomarkers Can Identify Pulmonary Tuberculosis in HIV-infected Drug Users Months Prior to Clinical Diagnosis. EBioMedicine 2014; 2:172-9. [PMID: 26137541 PMCID: PMC4484511 DOI: 10.1016/j.ebiom.2014.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 11/28/2014] [Accepted: 12/01/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Current diagnostic tests cannot identify which infected individuals are at risk for progression to tuberculosis (TB). Our aim was to identify biomarkers which can predict the development of TB prior to clinical diagnosis. METHOD In a retrospective case-control study, RNA of 14 HIV-infected drug users obtained before TB diagnosis (cases) and of 15 who did not develop TB (controls) was analyzed for the expression of 141 genes by dcRT-MLPA followed by Lasso regression analysis. FINDINGS A combined analysis of IL13 and AIRE had the highest discriminatory power to identify cases up to 8 months prior to clinical diagnosis. Cases expressing IL13 had a gene expression pattern strongly enriched for type I IFN related signaling genes, suggesting that these genes represent processes that contribute to TB pathogenesis. INTERPRETATION We here demonstrated that biomarkers, such as IL13-AIRE, can identify individuals that progress to TB within a high risk population, months prior to clinical diagnosis.
Collapse
Affiliation(s)
- Rosa Sloot
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Center for Infections and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Maarten F. Schim van der Loeff
- Center for Infections and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Infectious Diseases, Public Health Service, Amsterdam, The Netherlands
| | - Erik W. van Zwet
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Mariëlle C. Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Sytze T. Keizer
- Department of Infectious Diseases, Public Health Service, Amsterdam, The Netherlands
| | - Maarten Scholing
- Department of Infectious Diseases, Public Health Service, Amsterdam, The Netherlands
- Department of Medical Microbiology, Onze Lieve Vrouwe Gasthuis, Amsterdam, The Netherlands
| | - Tom H.M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Martien W. Borgdorff
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Center for Infections and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Infectious Diseases, Public Health Service, Amsterdam, The Netherlands
| | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
94
|
Rath M, Müller I, Kropf P, Closs EI, Munder M. Metabolism via Arginase or Nitric Oxide Synthase: Two Competing Arginine Pathways in Macrophages. Front Immunol 2014; 5:532. [PMID: 25386178 PMCID: PMC4209874 DOI: 10.3389/fimmu.2014.00532] [Citation(s) in RCA: 854] [Impact Index Per Article: 77.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 10/09/2014] [Indexed: 12/23/2022] Open
Abstract
Macrophages play a major role in the immune system, both as antimicrobial effector cells and as immunoregulatory cells, which induce, suppress or modulate adaptive immune responses. These key aspects of macrophage biology are fundamentally driven by the phenotype of macrophage arginine metabolism that is prevalent in an evolving or ongoing immune response. M1 macrophages express the enzyme nitric oxide synthase, which metabolizes arginine to nitric oxide (NO) and citrulline. NO can be metabolized to further downstream reactive nitrogen species, while citrulline might be reused for efficient NO synthesis via the citrulline–NO cycle. M2 macrophages are characterized by expression of the enzyme arginase, which hydrolyzes arginine to ornithine and urea. The arginase pathway limits arginine availability for NO synthesis and ornithine itself can further feed into the important downstream pathways of polyamine and proline syntheses, which are important for cellular proliferation and tissue repair. M1 versus M2 polarization leads to opposing outcomes of inflammatory reactions, but depending on the context, M1 and M2 macrophages can be both pro- and anti-inflammatory. Notably, M1/M2 macrophage polarization can be driven by microbial infection or innate danger signals without any influence of adaptive immune cells, secondarily driving the T helper (Th)1/Th2 polarization of the evolving adaptive immune response. Since both arginine metabolic pathways cross-inhibit each other on the level of the respective arginine break-down products and Th1 and Th2 lymphocytes can drive or amplify macrophage M1/M2 dichotomy via cytokine activation, this forms the basis of a self-sustaining M1/M2 polarization of the whole immune response. Understanding the arginine metabolism of M1/M2 macrophage phenotypes is therefore central to find new possibilities to manipulate immune responses in infection, autoimmune diseases, chronic inflammatory conditions, and cancer.
Collapse
Affiliation(s)
- Meera Rath
- Department of Pharmacology, Institute of Medical Sciences, Faculty of Medical Sciences, Siksha 'O' Anusandhan University , Bhubaneshwar , India
| | - Ingrid Müller
- Section of Immunology, Department of Medicine, Imperial College London , London , UK
| | - Pascale Kropf
- Section of Immunology, Department of Medicine, Imperial College London , London , UK
| | - Ellen I Closs
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University , Mainz , Germany
| | - Markus Munder
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center, Johannes Gutenberg University , Mainz , Germany ; Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University , Mainz , Germany
| |
Collapse
|
95
|
Macrophage arginase-1 controls bacterial growth and pathology in hypoxic tuberculosis granulomas. Proc Natl Acad Sci U S A 2014; 111:E4024-32. [PMID: 25201986 DOI: 10.1073/pnas.1408839111] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Lung granulomas develop upon Mycobacterium tuberculosis (Mtb) infection as a hallmark of human tuberculosis (TB). They are structured aggregates consisting mainly of Mtb-infected and -uninfected macrophages and Mtb-specific T cells. The production of NO by granuloma macrophages expressing nitric oxide synthase-2 (NOS2) via l-arginine and oxygen is a key protective mechanism against mycobacteria. Despite this protection, TB granulomas are often hypoxic, and bacterial killing via NOS2 in these conditions is likely suboptimal. Arginase-1 (Arg1) also metabolizes l-arginine but does not require oxygen as a substrate and has been shown to regulate NOS2 via substrate competition. However, in other infectious diseases in which granulomas occur, such as leishmaniasis and schistosomiasis, Arg1 plays additional roles such as T-cell regulation and tissue repair that are independent of NOS2 suppression. To address whether Arg1 could perform similar functions in hypoxic regions of TB granulomas, we used a TB murine granuloma model in which NOS2 is absent. Abrogation of Arg1 expression in macrophages in this setting resulted in exacerbated lung granuloma pathology and bacterial burden. Arg1 expression in hypoxic granuloma regions correlated with decreased T-cell proliferation, suggesting that Arg1 regulation of T-cell immunity is involved in disease control. Our data argue that Arg1 plays a central role in the control of TB when NOS2 is rendered ineffective by hypoxia.
Collapse
|