51
|
Barbosa Bomfim CC, Pinheiro Amaral E, Santiago-Carvalho I, Almeida Santos G, Machado Salles É, Hastreiter AA, Silva do Nascimento R, Almeida FM, Lopes Biá Ventura Simão T, Linhares Rezende A, Hiroyuki Hirata M, Ambrósio Fock R, Álvarez JM, Lasunskaia EB, D'Império Lima MR. Harmful Effects of Granulocytic Myeloid-Derived Suppressor Cells on Tuberculosis Caused by Hypervirulent Mycobacteria. J Infect Dis 2020; 223:494-507. [PMID: 33206171 DOI: 10.1093/infdis/jiaa708] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The role of myeloid-derived suppressor cells (MDSCs) in patients with severe tuberculosis who suffer from uncontrolled pulmonary inflammation caused by hypervirulent mycobacterial infection remains unclear. METHODS This issue was addressed using C57BL/6 mice infected with highly virulent Mycobacterium bovis strain MP287/03. RESULTS CD11b+GR1int population increased in the bone marrow, blood and lungs during advanced disease. Pulmonary CD11b+GR1int (Ly6GintLy6Cint) cells showed granularity similar to neutrophils and expressed immature myeloid cell markers. These immature neutrophils harbored intracellular bacilli and were preferentially located in the alveoli. T-cell suppression occurred concomitantly with CD11b+GR1int cell accumulation in the lungs. Furthermore, lung and bone marrow GR1+ cells suppressed both T-cell proliferation and interferon γ production in vitro. Anti-GR1 therapy given when MDSCs infiltrated the lungs prevented expansion and fusion of primary pulmonary lesions and the development of intragranulomatous caseous necrosis, along with increased mouse survival and partial recovery of T-cell function. Lung bacterial load was reduced by anti-GR1 treatment, but mycobacteria released from the depleted cells proliferated extracellularly in the alveoli, forming cords and clumps. CONCLUSIONS Granulocytic MDSCs massively infiltrate the lungs during infection with hypervirulent mycobacteria, promoting bacterial growth and the development of inflammatory and necrotic lesions, and are promising targets for host-directed therapies.
Collapse
Affiliation(s)
- Caio César Barbosa Bomfim
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Eduardo Pinheiro Amaral
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Igor Santiago-Carvalho
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Gislane Almeida Santos
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Érika Machado Salles
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Araceli Aparecida Hastreiter
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | | | - Fabrício M Almeida
- Laboratório de Biologia do Reconhecer, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Thatiana Lopes Biá Ventura Simão
- Laboratório de Biologia do Reconhecer, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Andreza Linhares Rezende
- Laboratório de Biologia do Reconhecer, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Mario Hiroyuki Hirata
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Ricardo Ambrósio Fock
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - José Maria Álvarez
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Elena B Lasunskaia
- Laboratório de Biologia do Reconhecer, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | | |
Collapse
|
52
|
Jøntvedt Jørgensen M, Jenum S, Tonby K, Mortensen R, Walzl G, Du Plessis N, Dyrhol-Riise AM. Monocytic myeloid-derived suppressor cells reflect tuberculosis severity and are influenced by cyclooxygenase-2 inhibitors. J Leukoc Biol 2020; 110:177-186. [PMID: 33155730 PMCID: PMC8359170 DOI: 10.1002/jlb.4a0720-409rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 10/02/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022] Open
Abstract
Myeloid‐derived suppressor cells (MDSCs) increase in tuberculosis (TB) and may be targets for host‐directed therapy (HDT). In this study, we use flow cytometry to analyze the effects of cyclooxygenase‐2 inhibitors (COX‐2i) on monocytic (M)‐MDSCs in blood from TB patients attending a clinical trial of COX‐2i. The effects of COX‐2i on M‐MDSCs and mycobacterial uptake were also studied by an in vitro mycobacterial infection model. We found that M‐MDSC frequencies correlated with TB disease severity. Reduced M‐MDSC (P = 0.05) and IDO (P = 0.03) expression was observed in the COX‐2i group. We show that peripheral blood‐derived M‐MDSCs successfully internalized Mycobacterium bovis and that in vitro mycobacterial infection increased COX‐2 (P = 0.002), PD‐L1 (P = 0.01), and Arginase‐1 (P = 0.002) expression in M‐MDSCs. Soluble IL‐1β, IL‐10, and S100A9 were reduced in COX‐2i‐treated M‐MDSCs cultures (P < 0.05). We show novel data that COX‐2i had limited effect in vivo but reduced M‐MDSC cytokine production in vitro. The relevance of COX‐2i in a HDT strategy needs to be further explored.
Collapse
Affiliation(s)
- Marthe Jøntvedt Jørgensen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Synne Jenum
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Kristian Tonby
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Gerhard Walzl
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Nelita Du Plessis
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Anne Ma Dyrhol-Riise
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
53
|
Ji J, Ren L, Liu Y, Zheng M, Fang H, Xu H, He H, Zhang M, Wang X, Shan J, Hou Y. Elevated monocytic myeloid-derived suppressor cells positively correlate with infection frequency in children with RRTIs. Eur J Immunol 2020; 51:2687-2690. [PMID: 33111310 DOI: 10.1002/eji.202048629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 08/28/2020] [Accepted: 10/26/2020] [Indexed: 11/06/2022]
Abstract
In children with recurrent respiratory tract infections (RRTIs), the percentages and numbers of monocytic myeloid-derived suppressor cells (MDSCs) were elevated. The elevated MDSCs positively correlate with respiratory tract infection frequency in RRTIs children. The elevated MDSCs can inhibit CD8+ T-cells proliferation in RRTIs children.
Collapse
Affiliation(s)
- Jianjian Ji
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lishun Ren
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuling Liu
- Institute of Pediatrics, Nanjing Pukou Hospital of TCM, Nanjing, China
| | - Min Zheng
- Department of Pediatric, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Huazhong Fang
- Institute of Pediatrics, Nanjing Pukou Hospital of TCM, Nanjing, China
| | - Hui Xu
- Institute of Pediatrics, Nanjing Pukou Hospital of TCM, Nanjing, China
| | - Huizhen He
- Institute of Pediatrics, Nanjing Pukou Hospital of TCM, Nanjing, China
| | - Min Zhang
- Institute of Pediatrics, Nanjing Pukou Hospital of TCM, Nanjing, China
| | - Xianzheng Wang
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
54
|
Sacchi A, Grassi G, Bordoni V, Lorenzini P, Cimini E, Casetti R, Tartaglia E, Marchioni L, Petrosillo N, Palmieri F, D'Offizi G, Notari S, Tempestilli M, Capobianchi MR, Nicastri E, Maeurer M, Zumla A, Locatelli F, Antinori A, Ippolito G, Agrati C. Early expansion of myeloid-derived suppressor cells inhibits SARS-CoV-2 specific T-cell response and may predict fatal COVID-19 outcome. Cell Death Dis 2020; 11:921. [PMID: 33110074 PMCID: PMC7590570 DOI: 10.1038/s41419-020-03125-1] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 01/08/2023]
Abstract
The immunological mechanisms underlying the clinical presentation of SARS-CoV-2 infection and those influencing the disease outcome remain to be defined. Myeloid-derived suppressor cells (MDSC) have been described to be highly increased during COVID-19, however, their role remains elusive. We performed an in depth analysis of MDSC in 128 SARS-CoV-2 infected patients. Polymorphonuclear (PMN)-MDSC expanded during COVID-19, in particular in patients who required intensive care treatments, and correlated with IL-1β, IL-6, IL-8, and TNF-α plasma levels. PMN-MDSC inhibited T-cells IFN-γ production upon SARS-CoV-2 peptides stimulation, through TGF-β- and iNOS-mediated mechanisms, possibly contrasting virus elimination. Accordingly, a multivariate regression analysis found a strong association between PMN-MDSC percentage and fatal outcome of the disease. The PMN-MDSC frequency was higher in non-survivors than survivors at the admission time, followed by a decreasing trend. Interestingly, this trend was associated with IL-6 increase in non-survivors but not in survivors. In conclusion, this study indicates PMN-MDSC as a novel factor in the pathogenesis of SARS-CoV2 infection, and open up to new therapeutic options.
Collapse
Affiliation(s)
- Alessandra Sacchi
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy.
| | - Germana Grassi
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Veronica Bordoni
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Patrizia Lorenzini
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Eleonora Cimini
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Rita Casetti
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Eleonora Tartaglia
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Luisa Marchioni
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Nicola Petrosillo
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Fabrizio Palmieri
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Gianpiero D'Offizi
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Stefania Notari
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Massimo Tempestilli
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Maria Rosaria Capobianchi
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Emanuele Nicastri
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Markus Maeurer
- Immunotherapy Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
- I Med Clinic, University of Mainz, Mainz, Germany
| | - Alimuddin Zumla
- Division of Infection and Immunity, University College London, London, UK
- National Institute of Health Research Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, UK
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico, Sapienza, University of Rome, Rome, Italy
| | - Andrea Antinori
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Giuseppe Ippolito
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| | - Chiara Agrati
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS- Via Portuense, 292- 00149, Rome, Italy
| |
Collapse
|
55
|
Ault RC, Headley CA, Hare AE, Carruthers BJ, Mejias A, Turner J. Blood RNA signatures predict recent tuberculosis exposure in mice, macaques and humans. Sci Rep 2020; 10:16873. [PMID: 33037303 PMCID: PMC7547102 DOI: 10.1038/s41598-020-73942-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 09/18/2020] [Indexed: 11/18/2022] Open
Abstract
Tuberculosis (TB) is the leading cause of death due to a single infectious disease. Knowing when a person was infected with Mycobacterium tuberculosis (M.tb) is critical as recent infection is the strongest clinical risk factor for progression to TB disease in immunocompetent individuals. However, time since M.tb infection is challenging to determine in routine clinical practice. To define a biomarker for recent TB exposure, we determined whether gene expression patterns in blood RNA correlated with time since M.tb infection or exposure. First, we found RNA signatures that accurately discriminated early and late time periods after experimental infection in mice and cynomolgus macaques. Next, we found a 6-gene blood RNA signature that identified recently exposed individuals in two independent human cohorts, including adult household contacts of TB cases and adolescents who recently acquired M.tb infection. Our work supports the need for future longitudinal studies of recent TB contacts to determine whether biomarkers of recent infection can provide prognostic information of TB disease risk in individuals and help map recent transmission in communities.
Collapse
Affiliation(s)
- Russell C Ault
- Texas Biomedical Research Institute, San Antonio, TX, USA
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
- Biomedical Sciences Graduate Program, Ohio State University, Columbus, OH, USA
- Medical Scientist Training Program, Ohio State University, Columbus, OH, USA
| | - Colwyn A Headley
- Texas Biomedical Research Institute, San Antonio, TX, USA
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
- Biomedical Sciences Graduate Program, Ohio State University, Columbus, OH, USA
| | - Alexander E Hare
- Biomedical Sciences Graduate Program, Ohio State University, Columbus, OH, USA
- Medical Scientist Training Program, Ohio State University, Columbus, OH, USA
| | - Bridget J Carruthers
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
| | - Asuncion Mejias
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Joanne Turner
- Texas Biomedical Research Institute, San Antonio, TX, USA.
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
56
|
Carranza C, Pedraza-Sanchez S, de Oyarzabal-Mendez E, Torres M. Diagnosis for Latent Tuberculosis Infection: New Alternatives. Front Immunol 2020; 11:2006. [PMID: 33013856 PMCID: PMC7511583 DOI: 10.3389/fimmu.2020.02006] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/24/2020] [Indexed: 12/29/2022] Open
Abstract
Latent tuberculosis infection (LTBI) is a subclinical mycobacterial infection defined on the basis of cellular immune response to mycobacterial antigens. The tuberculin skin test (TST) and the interferon gamma release assay (IGRA) are currently used to establish the diagnosis of LTB. However, neither TST nor IGRA is useful to discriminate between active and latent tuberculosis. Moreover, these tests cannot be used to predict whether an individual with LTBI will develop active tuberculosis (TB) or whether therapy for LTBI could be effective to decrease the risk of developing active TB. Therefore, in this article, we review current approaches and some efforts to identify an immunological marker that could be useful in distinguishing LTBI from TB and in evaluating the effectiveness of treatment of LTB on the risk of progression to active TB.
Collapse
Affiliation(s)
- Claudia Carranza
- Departamento de Microbiología, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - Sigifredo Pedraza-Sanchez
- Unidad de Bioquímica Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, Mexico City, Mexico
| | | | - Martha Torres
- Departamento de Microbiología, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico.,Subdirección de Investigación Biomédica, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| |
Collapse
|
57
|
Pai S, Muruganandah V, Kupz A. What lies beneath the airway mucosal barrier? Throwing the spotlight on antigen-presenting cell function in the lower respiratory tract. Clin Transl Immunology 2020; 9:e1158. [PMID: 32714552 PMCID: PMC7376394 DOI: 10.1002/cti2.1158] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/22/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
The global prevalence of respiratory infectious and inflammatory diseases remains a major public health concern. Prevention and management strategies have not kept pace with the increasing incidence of these diseases. The airway mucosa is the most common portal of entry for infectious and inflammatory agents. Therefore, significant benefits would be derived from a detailed understanding of how immune responses regulate the filigree of the airways. Here, the role of different antigen‐presenting cells (APC) in the lower airways and the mechanisms used by pathogens to modulate APC function during infectious disease is reviewed. Features of APC that are unique to the airways and the influence they have on uptake and presentation of antigen to T cells directly in the airways are discussed. Current information on the crucial role that airway APC play in regulating respiratory infection is summarised. We examine the clinical implications of APC dysregulation in the airways on asthma and tuberculosis, two chronic diseases that are the major cause of illness and death in the developed and developing world. A brief overview of emerging therapies that specifically target APC function in the airways is provided.
Collapse
Affiliation(s)
- Saparna Pai
- Centre for Molecular Therapeutics Australian Institute of Tropical Health and Medicine James Cook University Cairns QLD Australia
| | - Visai Muruganandah
- Centre for Molecular Therapeutics Australian Institute of Tropical Health and Medicine James Cook University Cairns QLD Australia
| | - Andreas Kupz
- Centre for Molecular Therapeutics Australian Institute of Tropical Health and Medicine James Cook University Cairns QLD Australia
| |
Collapse
|
58
|
Scott NR, Swanson RV, Al-Hammadi N, Domingo-Gonzalez R, Rangel-Moreno J, Kriel BA, Bucsan AN, Das S, Ahmed M, Mehra S, Treerat P, Cruz-Lagunas A, Jimenez-Alvarez L, Muñoz-Torrico M, Bobadilla-Lozoya K, Vogl T, Walzl G, du Plessis N, Kaushal D, Scriba TJ, Zúñiga J, Khader SA. S100A8/A9 regulates CD11b expression and neutrophil recruitment during chronic tuberculosis. J Clin Invest 2020; 130:3098-3112. [PMID: 32134742 PMCID: PMC7259997 DOI: 10.1172/jci130546] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 02/20/2020] [Indexed: 01/01/2023] Open
Abstract
Neutrophil accumulation is associated with lung pathology during active tuberculosis (ATB). However, the molecular mechanism or mechanisms by which neutrophils accumulate in the lung and contribute to TB immunopathology are not fully delineated. Using the well-established mouse model of TB, our new data provide evidence that the alarmin S100A8/A9 mediates neutrophil accumulation during progression to chronic TB. Depletion of neutrophils or S100A8/A9 deficiency resulted in improved Mycobacterium tuberculosis (Mtb) control during chronic but not acute TB. Mechanistically, we demonstrate that, following Mtb infection, S100A8/A9 expression is required for upregulation of the integrin molecule CD11b specifically on neutrophils, mediating their accumulation during chronic TB disease. These findings are further substantiated by increased expression of S100A8 and S100A9 mRNA in whole blood in human TB progressors when compared with nonprogressors and rapidly decreased S100A8/A9 protein levels in the serum upon TB treatment. Furthermore, we demonstrate that S100A8/A9 serum levels along with chemokines are useful in distinguishing between ATB and asymptomatic Mtb-infected latent individuals. Thus, our results support targeting S100A8/A9 pathways as host-directed therapy for TB.
Collapse
Affiliation(s)
| | | | - Noor Al-Hammadi
- Division of Biostatistics, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Belinda A. Kriel
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, South African Medical Research Council (SAMRC) Centre for Tuberculosis Research, DST-NRF Centre of Excellence for Biomedical TB Research, Stellenbosch University, Stellenbosch, South Africa
| | - Allison N. Bucsan
- Division of Bacteriology and
- Division of Parasitology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | | | | | - Smriti Mehra
- Division of Bacteriology and
- Division of Parasitology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | | | - Alfredo Cruz-Lagunas
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Luis Jimenez-Alvarez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Marcela Muñoz-Torrico
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Karen Bobadilla-Lozoya
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Thomas Vogl
- Institute of Immunology and
- Interdisciplinary Center for Clinical Research, University of Münster, Münster, Germany
| | - Gerhard Walzl
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, South African Medical Research Council (SAMRC) Centre for Tuberculosis Research, DST-NRF Centre of Excellence for Biomedical TB Research, Stellenbosch University, Stellenbosch, South Africa
| | - Nelita du Plessis
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, South African Medical Research Council (SAMRC) Centre for Tuberculosis Research, DST-NRF Centre of Excellence for Biomedical TB Research, Stellenbosch University, Stellenbosch, South Africa
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative and
- Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Joaquín Zúñiga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | | |
Collapse
|
59
|
Kelly-Scumpia KM, Choi A, Shirazi R, Bersabe H, Park E, Scumpia PO, Ochoa MT, Yu J, Ma F, Pellegrini M, Modlin RL. ER Stress Regulates Immunosuppressive Function of Myeloid Derived Suppressor Cells in Leprosy that Can Be Overcome in the Presence of IFN-γ. iScience 2020; 23:101050. [PMID: 32339990 PMCID: PMC7190750 DOI: 10.1016/j.isci.2020.101050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/27/2020] [Accepted: 04/06/2020] [Indexed: 02/06/2023] Open
Abstract
Myeloid derived suppressor cells (MDSCs) are a population of immature myeloid cells that suppress adaptive immune function, yet the factors that regulate their suppressive function in patients with infection remain unclear. We studied MDSCs in patients with leprosy, a disease caused by Mycobacterium leprae, where clinical manifestations present on a spectrum that correlate with immunity to the pathogen. We found that HLA-DR-CD33+CD15+ MDSCs were increased in blood from patients with disseminated/progressive lepromatous leprosy and possessed T cell-suppressive activity as compared with self-limiting tuberculoid leprosy. Mechanistically, we found ER stress played a critical role in regulating the T cell suppressive activity in these MDSCs. Furthermore, ER stress augmented IL-10 production, contributing to MDSC activity, whereas IFN-γ allowed T cells to overcome MDSC suppressive activity. These studies highlight a regulatory mechanism that links ER stress to IL-10 in mediating MDSC suppressive function in human infectious disease. Cells with an MDSC phenotype are increased in blood and skin of patients with leprosy Only MDSCs from patients with leprosy with disseminated infection suppress T cell function MDSC function is dependent on increased ER stress and IL-10 production MDSC function can be reversed in the presence of IFN-γ
Collapse
Affiliation(s)
| | - Aaron Choi
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Roksana Shirazi
- Division of Dermatology, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Hannah Bersabe
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Esther Park
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Philip O Scumpia
- Division of Dermatology, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Maria T Ochoa
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Jing Yu
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Feiyang Ma
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Robert L Modlin
- Division of Dermatology, David Geffen School of Medicine, Los Angeles, CA 90095, USA; Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
60
|
Kelly AM, McLoughlin RM. Target the Host, Kill the Bug; Targeting Host Respiratory Immunosuppressive Responses as a Novel Strategy to Improve Bacterial Clearance During Lung Infection. Front Immunol 2020; 11:767. [PMID: 32425944 PMCID: PMC7203494 DOI: 10.3389/fimmu.2020.00767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022] Open
Abstract
The lung is under constant pressure to protect the body from invading bacteria. An effective inflammatory immune response must be tightly orchestrated to ensure complete clearance of any invading bacteria, while simultaneously ensuring that inflammation is kept under strict control to preserve lung viability. Chronic bacterial lung infections are seen as a major threat to human life with the treatment of these infections becoming more arduous as the prevalence of antibiotic resistance becomes increasingly commonplace. In order to survive within the lung bacteria target the host immune system to prevent eradication. Many bacteria directly target inflammatory cells and cytokines to impair inflammatory responses. However, bacteria also have the capacity to take advantage of and strongly promote anti-inflammatory immune responses in the host lung to inhibit local pro-inflammatory responses that are critical to bacterial elimination. Host cells such as T regulatory cells and myeloid-derived suppressor cells are often enhanced in number and activity during chronic pulmonary infection. By increasing suppressive cell populations and cytokines, bacteria promote a permissive environment suitable for their prolonged survival. This review will explore the anti-inflammatory aspects of the lung immune system that are targeted by bacteria and how bacterial-induced immunosuppression could be inhibited through the use of host-directed therapies to improve treatment options for chronic lung infections.
Collapse
Affiliation(s)
- Alanna M Kelly
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rachel M McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
61
|
Leukes V, Walzl G, du Plessis N. Myeloid-Derived Suppressor Cells as Target of Phosphodiesterase-5 Inhibitors in Host-Directed Therapeutics for Tuberculosis. Front Immunol 2020; 11:451. [PMID: 32269568 PMCID: PMC7109258 DOI: 10.3389/fimmu.2020.00451] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/27/2020] [Indexed: 01/11/2023] Open
Abstract
Resistance toward current and new classes of anti-tuberculosis (anti-TB) antibiotics are rapidly emerging; thus, innovative therapies focused on host processes, termed host-directed therapies (HDTs), are promising novel approaches for shortening therapy regimens without inducing drug resistance. Development of new TB drugs is lengthy and expensive, and success is not guaranteed; thus, alternatives are needed. Repurposed drugs have already passed Food and Drug Administration (FDA) as well as European Medicines Agency (EMA) safety requirements and may only need to prove efficacy against Mycobacterium tuberculosis (M.tb). Phosphodiesterases (PDEs) hydrolyze the catalytic breakdown of both cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) to their inactive mononucleotides. Advances in molecular pharmacology have identified 11 PDE families; and the success of sildenafil, a PDE-5 selective inhibitor (PDE-5i), in treating pulmonary hypertension and erectile dysfunction has invigorated research into the therapeutic potential of selective PDE inhibitors in other conditions. Myeloid-derived suppressor cells (MDSCs) suppress anti-TB T-cell responses, likely contributing to TB disease progression. PDE-5i increases cGMP within MDSC resulting in the downregulation of arginase-1 (ARG1) and nitric oxide synthase 2 (NOS2), reducing MDSC's suppressive potential. The effect of this reduction decreases MDSC-induced T-cell-suppressive mechanisms. This review highlights the possibility of HDT targeting of MDSC, using a PDE-5i in combination with the current TB regimen, resulting in improved TB treatment efficacy.
Collapse
Affiliation(s)
- Vinzeigh Leukes
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Nelita du Plessis
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
62
|
Kotzé LA, Young C, Leukes VN, John V, Fang Z, Walzl G, Lutz MB, du Plessis N. Mycobacterium tuberculosis and myeloid-derived suppressor cells: Insights into caveolin rich lipid rafts. EBioMedicine 2020; 53:102670. [PMID: 32113158 PMCID: PMC7047144 DOI: 10.1016/j.ebiom.2020.102670] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/18/2019] [Accepted: 12/02/2019] [Indexed: 02/06/2023] Open
Abstract
Mycobacterium tuberculosis (M.tb) is likely the most successful human pathogen, capable of evading protective host immune responses and driving metabolic changes to support its own survival and growth. Ineffective innate and adaptive immune responses inhibit effective clearance of the bacteria from the human host, resulting in the progression to active TB disease. Many regulatory mechanisms exist to prevent immunopathology, however, chronic infections result in the overproduction of regulatory myeloid cells, like myeloid-derived suppressor cells (MDSC), which actively suppress protective host T lymphocyte responses among other immunosuppressive mechanisms. The mechanisms of M.tb internalization by MDSC and the involvement of host-derived lipid acquisition, have not been fully elucidated. Targeted research aimed at investigating MDSC impact on phagocytic control of M.tb, would be advantageous to our collective anti-TB arsenal. In this review we propose a mechanism by which M.tb may be internalized by MDSC and survive via the manipulation of host-derived lipid sources.
Collapse
Affiliation(s)
- Leigh A Kotzé
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Carly Young
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Vinzeigh N Leukes
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Vini John
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Zhuo Fang
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Manfred B Lutz
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Nelita du Plessis
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
63
|
Keshavjee S, Amanullah F, Cattamanchi A, Chaisson R, Dobos KM, Fox GJ, Gendelman HE, Gordon R, Hesseling A, Le Van H, Kampmann B, Kana B, Khuller G, Lewinsohn DM, Lewinsohn DA, Lin PL, Lu LL, Maartens G, Owen A, Protopopova M, Rengarajan J, Rubin E, Salgame P, Schurr E, Seddon JA, Swindells S, Tobin DM, Udwadia Z, Walzl G, Srinivasan S, Rustomjee R, Nahid P. Moving toward Tuberculosis Elimination. Critical Issues for Research in Diagnostics and Therapeutics for Tuberculosis Infection. Am J Respir Crit Care Med 2020; 199:564-571. [PMID: 30335466 DOI: 10.1164/rccm.201806-1053pp] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Salmaan Keshavjee
- 1 Harvard Medical School, Boston, Massachusetts.,2 Harvard Medical School Center for Global Health Delivery-Dubai, Dubai, United Arab Emirates
| | | | - Adithya Cattamanchi
- 4 University of California San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, California
| | - Richard Chaisson
- 5 Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Gregory J Fox
- 7 University of Sydney, Sydney, New South Wales, Australia
| | | | - Richard Gordon
- 9 South African Medical Research Council, Cape Town, Western Cape, South Africa
| | | | - Hoi Le Van
- 11 National Lung Hospital, Hanoi, Vietnam.,12 National TB Program in Vietnam, Hanoi, Vietnam
| | - Beate Kampmann
- 13 London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Bavesh Kana
- 14 University of Witwatersrand, National Health Laboratory Service, Johannesburg, South Africa.,15 Centre for the AIDS Programme of Research in South Africa, CAPRISA, Durban, South Africa
| | - Gopal Khuller
- 16 Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - David M Lewinsohn
- 17 Oregon Health & Science University, Portland, Oregon.,18 Portland VA Medical Center, Portland, Oregon
| | | | - Philiana Ling Lin
- 19 University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Lenette Lin Lu
- 20 Massachusetts General Hospital, Boston, Massachusetts
| | - Gary Maartens
- 21 Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Andrew Owen
- 22 University of Liverpool, Liverpool, England
| | - Marina Protopopova
- 23 Division of AIDS, National Institute of Allergy and Infectious Diseases, NIH, Department of Health and Human Services, Rockville, Maryland
| | | | - Eric Rubin
- 25 Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | | | | | - James A Seddon
- 13 London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - David M Tobin
- 28 Duke University School of Medicine, Durham, North Carolina; and
| | - Zarir Udwadia
- 29 Hinduja Hospital & Research Center, Mumbai, India
| | - Gerhard Walzl
- 30 Division of Molecular Biology and Human Genetics, Stellenbosch University, Cape Town, South Africa.,14 University of Witwatersrand, National Health Laboratory Service, Johannesburg, South Africa
| | - Sudha Srinivasan
- 23 Division of AIDS, National Institute of Allergy and Infectious Diseases, NIH, Department of Health and Human Services, Rockville, Maryland
| | - Roxana Rustomjee
- 23 Division of AIDS, National Institute of Allergy and Infectious Diseases, NIH, Department of Health and Human Services, Rockville, Maryland
| | - Payam Nahid
- 4 University of California San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, California
| |
Collapse
|
64
|
Fernández-Ruiz JC, Galindo-De Ávila JC, Martínez-Fierro ML, Garza-Veloz I, Cervantes-Villagrana AR, Valtierra-Alvarado MA, Serrano CJ, García-Hernández MH, Enciso-Moreno JA, Castañeda-Delgado JE. Myeloid-Derived Suppressor Cells Show Different Frequencies in Diabetics and Subjects with Arterial Hypertension. J Diabetes Res 2019; 2019:1568457. [PMID: 31915708 PMCID: PMC6930726 DOI: 10.1155/2019/1568457] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/10/2019] [Accepted: 10/29/2019] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes mellitus (DM2) is strongly associated with other comorbidities such as obesity, atherosclerosis, and hypertension. Obesity is associated with sustained low-grade inflammatory response due to the production of proinflammatory cytokines. This inflammatory process promotes the differentiation of some myeloid cells, including myeloid-derived suppressor cells (MDSCs). In this study, two groups of individuals were included: DM2 patients and non-DM2 individuals with similar characteristics. Immunolabeling of CD15+ CD14- and CD33+ HLA-DR-/low was performed from whole peripheral blood, and samples were analyzed by flow cytometry, and frequencies of MDSCs and the relationship of these with clinical variables, cytokine profile (measured by cytometric bead array), and anthropometric variables were analyzed. The frequency of CD33+ HLA-DR-/low MDSCs (that produce IL-10 and TGF-β, according to an intracellular detection) is higher in patients with DM2 (P < 0.05), and there is a positive correlation between the frequency of CD15+ CD14- and CD33+ HLA-DR-/low MDSC phenotypes. DM2 patients have an increased concentration of serum IL-5 (P < 0.05). Also, a negative correlation between the frequency of CD15+ CD14- MDSCs and LDL cholesterol was found. Our group of DM2 patients have an increased frequency of mononuclear MDSC CD33+ HLA-DR-/low that produce TGF-β and IL-10. These cytokines have been associated with immune modulation and reduced T cell responses. DM2 and non-DM2 subjects show a similar cytokine profile, but the DM2 patients have an increased concentration of IL-5.
Collapse
Affiliation(s)
- Julio C. Fernández-Ruiz
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas, Mexico
- Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Julia C. Galindo-De Ávila
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas, Mexico
- Maestría en Ciencias Biomédicas, Área de Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas, Mexico
| | - Margarita L. Martínez-Fierro
- Laboratorio de Medicina Molecular, Unidad Académica de Medicina Humana y Ciencias de la Salud, Campus UAZ XXI, Universidad Autónoma de Zacatecas, Zacatecas, Mexico
| | - Idalia Garza-Veloz
- Laboratorio de Medicina Molecular, Unidad Académica de Medicina Humana y Ciencias de la Salud, Campus UAZ XXI, Universidad Autónoma de Zacatecas, Zacatecas, Mexico
| | | | - Monica A. Valtierra-Alvarado
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas, Mexico
- Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Carmen J. Serrano
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas, Mexico
| | | | - José A. Enciso-Moreno
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas, Mexico
| | - Julio E. Castañeda-Delgado
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas, Mexico
- Cátedras CONACYT, Consejo Nacional de Ciencia y Tecnología, Mexico
| |
Collapse
|
65
|
Rajamanickam A, Munisankar S, Dolla CK, Babu S. Undernutrition is associated with perturbations in T cell-, B cell-, monocyte- and dendritic cell- subsets in latent Mycobacterium tuberculosis infection. PLoS One 2019; 14:e0225611. [PMID: 31821327 PMCID: PMC6903744 DOI: 10.1371/journal.pone.0225611] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/07/2019] [Indexed: 12/11/2022] Open
Abstract
Undernutrition, as described by low body mass index (BMI), is a foremost risk factor for the progression of active Tuberculosis (TB). Undernutrition is also known to impact the baseline frequencies of innate and adaptive immune cells in animal models. To verify whether undernutrition has any influence on the baseline frequencies of immune cells in latent Mycobacterium tuberculosis infection (LTBI), we examined the frequencies of T cell-, B cell, monocyte- and dendritic cell (DC)- subsets in individuals with LTBI and low BMI (LBMI) and contrasted them with LTBI and normal BMI (NBMI) groups. LBMI was characterized by decreased frequencies and absolute cell counts of T cells, B cells and NK cells in comparison with NBMI. LBMI individuals demonstrated significantly enhanced frequencies of naïve and effector CD4+ and CD8+ T cells and significantly decreased frequencies of central memory, effector memory CD4+ and CD8+ T cells and regulatory T cells. Among B cell subsets, LBMI individuals demonstrated significantly diminished frequencies of naïve, immature, classical memory, activated memory, atypical memory and plasma cells. In addition, LBMI individuals showed significantly decreased frequencies of classical monocytes, myeloid DCs and plasmacytoid DCs and significantly increased frequencies of intermediate and non-classical monocytes and myeloid derived suppressor cells. BMI exhibited a positive correlation with B cell and NK cell counts. Our data, therefore, demonstrates that coexistent undernutrition in LTBI is characterized by the occurrence of a significant modulation in the frequency of innate and adaptive immune cell subsets.
Collapse
Affiliation(s)
- Anuradha Rajamanickam
- National Institute of Health-NIRT-International Center for Excellence in Research, Chennai, India
- * E-mail:
| | - Saravanan Munisankar
- National Institute of Health-NIRT-International Center for Excellence in Research, Chennai, India
| | | | - Subash Babu
- National Institute of Health-NIRT-International Center for Excellence in Research, Chennai, India
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
66
|
John V, Kotze LA, Ribechini E, Walzl G, Du Plessis N, Lutz MB. Caveolin-1 Controls Vesicular TLR2 Expression, p38 Signaling and T Cell Suppression in BCG Infected Murine Monocytic Myeloid-Derived Suppressor Cells. Front Immunol 2019; 10:2826. [PMID: 31849990 PMCID: PMC6901706 DOI: 10.3389/fimmu.2019.02826] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022] Open
Abstract
Monocytic myeloid-derived suppressor cells (M-MDSCs) and granulocytic MDSCs (G-MDSCs) have been found to be massively induced in TB patients as well in murine Mtb infection models. However, the interaction of mycobacteria with MDSCs and its role in TB infection is not well studied. Here, we investigated the role of Cav-1 for MDSCs infected with Mycobacterium bovis Bacille-Calmette-Guerín (BCG). MDSCs that were generated from murine bone marrow (MDSCs) of wild-type (WT) or Cav1−/− mice upregulated Cav-1, TLR4 and TLR2 expression after BCG infection on the cell surface. However, Cav-1 deficiency resulted in a selective defect of intracellular TLR2 levels predominantly in the M-MDSC subset. Further analysis indicated no difference in the phagocytosis of BCG by M-MDSCs from WT and Cav1−/− mice or caveosome formation, but a reduced capacity to up-regulate surface markers, to secrete various cytokines, to induce iNOS and NO production required for suppression of T cell proliferation, whereas Arg-1 was not affected. Among the signaling pathways affected by Cav-1 deficiency, we found lower phosphorylation of the p38 mitogen-activated protein kinase (MAPK). Together, our findings implicate that (i) Cav-1 is dispensable for the internalization of BCG, (ii) vesicular TLR2 signaling in M-MDSCs is a major signaling pathway induced by BCG, (iii) vesicular TLR2 signals are controlled by Cav-1, (iv) vesicular TLR2/Cav-1 signaling is required for T cell suppressor functions.
Collapse
Affiliation(s)
- Vini John
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Leigh A Kotze
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Eliana Ribechini
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Gerhard Walzl
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Nelita Du Plessis
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Manfred B Lutz
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
67
|
Agallou M, Athanasiou E, Kammona O, Tastsoglou S, Hatzigeorgiou AG, Kiparissides C, Karagouni E. Transcriptome Analysis Identifies Immune Markers Related to Visceral Leishmaniasis Establishment in the Experimental Model of BALB/c Mice. Front Immunol 2019; 10:2749. [PMID: 31849951 PMCID: PMC6902045 DOI: 10.3389/fimmu.2019.02749] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/11/2019] [Indexed: 12/29/2022] Open
Abstract
Visceral leishmaniasis (VL) caused by Leishmania donovani and L. infantum is a potentially fatal disease. To date there are no registered vaccines for disease prevention despite the fact that several vaccines are in preclinical development. Thus, new strategies are needed to improve vaccine efficacy based on a better understanding of the mechanisms mediating protective immunity and mechanisms of host immune responses subversion by immunopathogenic components of Leishmania. We found that mice vaccinated with CPA162−189-loaded p8-PLGA nanoparticles, an experimental nanovaccine, induced the differentiation of antigen-specific CD8+ T cells in spleen compared to control mice, characterized by increased dynamics of proliferation and high amounts of IFN-γ production after ex vivo re-stimulation with CPA162−189 antigen. Vaccination with CPA162−189-loaded p8-PLGA nanoparticles resulted in about 80% lower parasite load in spleen and liver at 4 weeks after challenge with L. infantum promastigotes as compared to control mice. However, 16 weeks after infection the parasite load in spleen was comparable in both mouse groups. Decreased protection levels in vaccinated mice were followed by up-regulation of the anti-inflammatory IL-10 production although at lower levels in comparison to control mice. Microarray analysis in spleen tissue at 4 weeks post challenge revealed different immune-related profiles among the two groups. Specifically, vaccinated mice were characterized by similar profile to naïve mice. On the other hand, the transcriptome of the non-vaccinated mice was dominated by increased expression of genes related to interferon type I, granulocyte chemotaxis, and immune cells suppression. This profile was significantly enriched at 16 weeks post challenge, a time-point which is relative to disease establishment, and was common for both groups, further suggesting that type I signaling and granulocyte influx has a significant role in disease establishment, pathogenesis and eventually in decreased vaccine efficacy for stimulating long-term protection. Overall, we put a spotlight on host immune networks during active VL as potential targets to improve and design more effective vaccines against disease.
Collapse
Affiliation(s)
- Maria Agallou
- Parasite Immunology Group, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Evita Athanasiou
- Parasite Immunology Group, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Olga Kammona
- Chemical Process & Energy Resources Institute, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Spyros Tastsoglou
- DIANA-Lab, Department of Electrical & Computer Engineering, University of Thessaly, Volos, Greece
| | - Artemis G Hatzigeorgiou
- DIANA-Lab, Department of Electrical & Computer Engineering, University of Thessaly, Volos, Greece.,DIANA-Lab, Hellenic Pasteur Institute, Athens, Greece
| | - Costas Kiparissides
- Chemical Process & Energy Resources Institute, Centre for Research and Technology Hellas, Thessaloniki, Greece.,Department of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evdokia Karagouni
- Parasite Immunology Group, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| |
Collapse
|
68
|
Zhan Y, Lew AM, Chopin M. The Pleiotropic Effects of the GM-CSF Rheostat on Myeloid Cell Differentiation and Function: More Than a Numbers Game. Front Immunol 2019; 10:2679. [PMID: 31803190 PMCID: PMC6873328 DOI: 10.3389/fimmu.2019.02679] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 10/30/2019] [Indexed: 12/27/2022] Open
Abstract
Granulocyte Macrophage-Colony Stimulating Factor (GM-CSF) is a myelopoietic growth factor that has pleiotropic effects not only in promoting the differentiation of immature precursors into polymorphonuclear neutrophils (PMNs), monocytes/macrophages (MØs) and dendritic cells (DCs), but also in controlling the function of fully mature myeloid cells. This broad spectrum of GM-CSF action may elicit paradoxical outcomes-both immunostimulation and immunosuppression-in infection, inflammation, and cancer. The complexity of GM-CSF action remains to be fully unraveled. Several aspects of GM-CSF action could contribute to its diverse biological consequences. Firstly, GM-CSF as a single cytokine affects development of most myeloid cells from progenitors to mature immune cells. Secondly, GM-CSF activates JAK2/STAT5 and also activate multiple signaling modules and transcriptional factors that direct different biological processes. Thirdly, GM-CSF can be produced by different cell types including tumor cells in response to different environmental cues; thus, GM-CSF quantity can vary greatly under different pathophysiological settings. Finally, GM-CSF signaling is also fine-tuned by other less defined feedback mechanisms. In this review, we will discuss the role of GM-CSF in orchestrating the differentiation, survival, and proliferation during the generation of multiple lineages of myeloid cells (PMNs, MØs, and DCs). We will also discuss the role of GM-CSF in regulating the function of DCs and the functional polarization of MØs. We highlight how the dose of GM-CSF and corresponding signal strength acts as a rheostat to fine-tune cell fate, and thus the way GM-CSF may best be targeted for immuno-intervention in infection, inflammation and cancer.
Collapse
Affiliation(s)
- Yifan Zhan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Andrew M Lew
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Department of Immunology and Microbiology, University of Melbourne, Parkville, VIC, Australia
| | - Michael Chopin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
69
|
Gideon HP, Phuah J, Junecko BA, Mattila JT. Neutrophils express pro- and anti-inflammatory cytokines in granulomas from Mycobacterium tuberculosis-infected cynomolgus macaques. Mucosal Immunol 2019; 12:1370-1381. [PMID: 31434990 PMCID: PMC6824993 DOI: 10.1038/s41385-019-0195-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 02/04/2023]
Abstract
Neutrophils are implicated in the pathogenesis of tuberculosis (TB), a disease caused by Mycobacterium tuberculosis infection, but the mechanisms by which they promote disease are not fully understood. Neutrophils can express cytokines that influence TB progression, and so we compared neutrophil and T-cell expression of the Th1 cytokines IFNγ and TNF, the Th2 cytokine IL-4, and regulatory cytokine IL-10 in M. tuberculosis-infected macaques to determine if neutrophil cytokine expression contributes to dysregulated immunity in TB. We found that peripheral blood neutrophils produced cytokines after stimulation by mycobacterial antigens and inactive and viable M. tuberculosis. M. tuberculosis antigen-stimulated neutrophils inhibited antigen-specific T-cell IFNγ production. In lung granulomas, neutrophil cytokine expression resembled T-cell cytokine expression, and although there was histologic evidence for neutrophil interaction with T cells, neutrophil cytokine expression was not correlated with T-cell cytokine expression or bacteria load. There was substantial overlap in the spatial arrangement of cytokine-expressing neutrophils and T cells, but IL-10-expressing neutrophils were also abundant in bacteria-rich areas between caseum and epithelioid macrophages. These results suggest that neutrophils contribute to the cytokine milieu in granulomas and may be important immunoregulatory cells in TB granulomas.
Collapse
Affiliation(s)
- Hannah P Gideon
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Jiayao Phuah
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Beth A Junecko
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Joshua T Mattila
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
70
|
Ribechini E, Eckert I, Beilhack A, Du Plessis N, Walzl G, Schleicher U, Ritter U, Lutz MB. Heat-killed Mycobacterium tuberculosis prime-boost vaccination induces myeloid-derived suppressor cells with spleen dendritic cell-killing capability. JCI Insight 2019; 5:128664. [PMID: 31162143 DOI: 10.1172/jci.insight.128664] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tuberculosis patients and mice infected with live Mycobacterium tuberculosis (Mtb) accumulate high numbers of myeloid-derived suppressor cells (MDSCs). Here, we hypothesized that also dead Mtb vaccines may induce MDSCs that could impair the efficacy of vaccination. We found that repeated injections of Mtb vaccines (heat-killed Mtb in Incomplete Freund's Adjuvant, like Montanide) but not single or control vaccines without Mtb strongly expanded CD11b+ myeloid cells in the spleen, that suppressed T cell proliferation and killing ex vivo. Dead Mtb vaccination induced the generation of CD11b+ Ly-6Chigh CD115+ iNOS/Nos2+ monocytic MDSCs (M-MDSCs) upon application of inflammatory or microbial activation signals. In vivo these M-MDSCs positioned strategically in the spleen by infiltrating the splenic bridging channels and white pulp areas. Notably, within 6 to 24 hours in a Nos2-dependent fashion they produced NO to rapidly kill conventional and plasmacytoid dendritic cells (cDCs, pDCs) while, surprisingly, sparing T cells in vivo. Thus, we demonstrate that Mtb vaccine induced M-MDSCs to not directly suppress T cell in vivo but, instead, M-MDSCs directly target DC subpopulations thereby indirectly suppressing effector T cell responses. Collectively, we demonstrate that Mtb booster vaccines induce M-MDSCs in the spleen that can be activated to kill DCs cautioning to thoroughly investigate MDSC formation in individuals after Mtb vaccination in clinical trials.
Collapse
Affiliation(s)
- Eliana Ribechini
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Ina Eckert
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Andreas Beilhack
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Nelita Du Plessis
- South African Medical Research Council, Centre for Tuberculosis Research, Department of Science and Technology - National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- South African Medical Research Council, Centre for Tuberculosis Research, Department of Science and Technology - National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Ulrike Schleicher
- Microbiology Institute, Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Uwe Ritter
- RCI Regensburg Center for Interventional Immunology, Institute of Immunology, University Medical Center Regensburg, University of Regensburg, Regensburg, Germany
| | - Manfred B Lutz
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
71
|
Pawelec G, Verschoor CP, Ostrand-Rosenberg S. Myeloid-Derived Suppressor Cells: Not Only in Tumor Immunity. Front Immunol 2019; 10:1099. [PMID: 31156644 PMCID: PMC6529572 DOI: 10.3389/fimmu.2019.01099] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/30/2019] [Indexed: 12/15/2022] Open
Abstract
Since the realization that immature myeloid cells are powerful modulators of the immune response, many studies on “myeloid-derived suppressor cells” (MDSCs) have documented their ability to promote tumor progression in melanoma and other cancers. Whether MDSCs are induced solely pathologically in tumorigenesis, or whether they also represent physiological immune control mechanisms, is not well-understood, but is particularly important in the light of ongoing attempts to block their activities in order to enhance anti-tumor immunity. Here, we briefly review studies which explore (1) how best to identify MDSCs in the context of cancer and how this compares to other conditions in humans; (2) what the suppressive mechanisms of MDSCs are and how to target them pharmacologically; (3) whether levels of MDSCs with various phenotypes are informative for clinical outcome not only in cancer but also other diseases, and (4) whether MDSCs are only found under pathological conditions or whether they also represent a physiological regulatory mechanism for the feedback control of immunity. Studies unequivocally document that MDSCs strongly influence cancer outcomes, but are less informative regarding their relevance to infection, autoimmunity, transplantation and aging, especially in humans. So far, the results of clinical interventions to reverse their negative effects in cancer have been disappointing; thus, developing differential approaches to modulate MSDCs in cancer and other diseases without unduly comprising any normal physiological function requires further exploration.
Collapse
Affiliation(s)
- Graham Pawelec
- Department of Immunology, University of Tübingen, Tübingen, Germany.,Health Sciences North Research Institute, Sudbury, ON, Canada
| | - Chris P Verschoor
- Health Sciences North Research Institute, Sudbury, ON, Canada.,Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - Suzanne Ostrand-Rosenberg
- Department of Pathology and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
72
|
Esher SK, Fidel PL, Noverr MC. Candida/Staphylococcal Polymicrobial Intra-Abdominal Infection: Pathogenesis and Perspectives for a Novel Form of Trained Innate Immunity. J Fungi (Basel) 2019; 5:E37. [PMID: 31075836 PMCID: PMC6617080 DOI: 10.3390/jof5020037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 01/10/2023] Open
Abstract
Polymicrobial sepsis is difficult to diagnose and treat and causes significant morbidity and mortality, especially when fungi are involved. In vitro, synergism between Candida albicans and various bacterial species has been described for many years. Our laboratory has developed a murine model of polymicrobial intra-abdominal infection with Candida albicans and Staphylococcus aureus, demonstrating that polymicrobial infections cause high levels of mortality, while monoinfections do not. By contrast, closely related Candida dubliniensis does not cause synergistic lethality and rather provides protection against lethal polymicrobial infection. This protection is thought to be driven by a novel form of trained innate immunity mediated by myeloid-derived suppressor cells (MDSCs), which we are proposing to call "trained tolerogenic immunity". MDSC accumulation has been described in patients with sepsis, as well as in in vivo sepsis models. However, clinically, MDSCs are considered detrimental in sepsis, while their role in in vivo models differs depending on the sepsis model and timing. In this review, we will discuss the role of MDSCs in sepsis and infection and summarize our perspectives on their development and function in the spectrum of trained innate immune protection against fungal-bacterial sepsis.
Collapse
Affiliation(s)
- Shannon K Esher
- Center of Excellence in Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA 70119, USA.
- Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA 70112, USA.
| | - Paul L Fidel
- Center of Excellence in Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA 70119, USA.
| | - Mairi C Noverr
- Center of Excellence in Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA 70119, USA.
- Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA 70112, USA.
| |
Collapse
|
73
|
Lalvani A, Berrocal-Almanza LC, Halliday A. Predicting progression to active tuberculosis: A rate-limiting step on the path to elimination. PLoS Med 2019; 16:e1002814. [PMID: 31125334 PMCID: PMC6534286 DOI: 10.1371/journal.pmed.1002814] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In a Perspective, Ajit Lalvani and colleagues discuss new approaches to predicting progression to active tuberculosis.
Collapse
Affiliation(s)
- Ajit Lalvani
- National Institute for Health Research Health Protection Research Unit in Respiratory Infections, Imperial College London, London, United Kingdom
- Tuberculosis Research Centre, National Heart and Lung Institute, St Mary's Campus, Imperial College London, London, United Kingdom
- * E-mail:
| | - Luis C. Berrocal-Almanza
- National Institute for Health Research Health Protection Research Unit in Respiratory Infections, Imperial College London, London, United Kingdom
| | - Alice Halliday
- Tuberculosis Research Centre, National Heart and Lung Institute, St Mary's Campus, Imperial College London, London, United Kingdom
- Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
74
|
Magcwebeba T, Dorhoi A, du Plessis N. The Emerging Role of Myeloid-Derived Suppressor Cells in Tuberculosis. Front Immunol 2019; 10:917. [PMID: 31114578 PMCID: PMC6502992 DOI: 10.3389/fimmu.2019.00917] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/09/2019] [Indexed: 12/22/2022] Open
Abstract
Myeloid cells are crucial for the host control of a Mycobacterium tuberculosis (M.tb) infection, however the adverse role of specific myeloid subsets has increasingly been appreciated. The relevance of such cells in therapeutic strategies and predictive/prognostic algorithms is to promote interest in regulatory myeloid cells in tuberculosis (TB). Myeloid-derived suppressor cells (MDSC) are a heterogeneous collection of phagocytes comprised of monocytic- and polymorphonuclear cells that exhibit a potent suppression of innate- and adaptive immune responses. Accumulation of MDSC under pathological conditions associated with chronic inflammation, most notably cancer, has been well-described. Evidence supporting the involvement of MDSC in TB is increasing, yet their significance in this infection continues to be viewed with skepticism, primarily due to their complex nature and the lack of genetic evidence unequivocally discriminating these cells from other terminally differentiated myeloid populations. Here we highlight recent advances in MDSC characterization and summarize findings on the TB-induced hematopoietic shift associated with MDSC expansion. Lastly, the mechanisms of MDSC-mediated disease progression and future research avenues in the context of TB therapy and prophylaxis are discussed.
Collapse
Affiliation(s)
- Tandeka Magcwebeba
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, South African MRC Centre for Tuberculosis Research, DST and NRF Centre of Excellence for Biomedical TB Research, Stellenbosch University, Stellenbosch, South Africa
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Greifswald, Germany.,Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany.,Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Nelita du Plessis
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, South African MRC Centre for Tuberculosis Research, DST and NRF Centre of Excellence for Biomedical TB Research, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
75
|
Khan A, Singh VK, Hunter RL, Jagannath C. Macrophage heterogeneity and plasticity in tuberculosis. J Leukoc Biol 2019; 106:275-282. [PMID: 30938876 DOI: 10.1002/jlb.mr0318-095rr] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 12/14/2022] Open
Abstract
Macrophages are the primary host cells for Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), during its intracellular survival in humans. The pathogen has a remarkable capacity to survive within the hostile environment of macrophages. However, primary infection does not result in active TB disease in most individuals. The majority of individuals remain latently infected, wherein the bacteria are held in check by the host immune response. Nevertheless, such individuals can develop active TB later upon the decline in their immune status. In contrast, in a small fraction of infected individuals, the host immune response fails to control the growth of M. tuberculosis bacilli, and granulomatous TB develops progressively. Elucidating the molecular and phenotypic events that govern the outcome of the infection within macrophages is fundamental to understanding the key features of these cells that could be equally critical in infection control. The molecular details of the M. tuberculosis-macrophage interaction continue to be discerned, and emerging evidence suggests that macrophage population that participate in infection is heterogeneous. While the local environment and developmental origin could influence the phenotypic heterogeneity and functional plasticity of macrophages, M. tuberculosis has also been demonstrated to modulate the polarization of macrophages. In this review, we draw on work investigating specialized macrophage populations and their interactions with M. tuberculosis with respect to pathogenesis and specific immune responses. Understanding the mechanisms that control the repertoire of macrophage phenotypes and behaviors during infection may provide prospects for novel TB control strategies through modulation of immunobiological functions of macrophages.
Collapse
Affiliation(s)
- Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Vipul Kumar Singh
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Robert L Hunter
- Department of Pathology and Laboratory Medicine, McGovern Medical School, University of Texas Health Sciences Center-Houston, Houston, Texas, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
76
|
Dorhoi A, Glaría E, Garcia-Tellez T, Nieuwenhuizen NE, Zelinskyy G, Favier B, Singh A, Ehrchen J, Gujer C, Münz C, Saraiva M, Sohrabi Y, Sousa AE, Delputte P, Müller-Trutwin M, Valledor AF. MDSCs in infectious diseases: regulation, roles, and readjustment. Cancer Immunol Immunother 2019; 68:673-685. [PMID: 30569204 PMCID: PMC11028159 DOI: 10.1007/s00262-018-2277-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 10/29/2018] [Indexed: 12/24/2022]
Abstract
Many pathogens, ranging from viruses to multicellular parasites, promote expansion of MDSCs, which are myeloid cells that exhibit immunosuppressive features. The roles of MDSCs in infection depend on the class and virulence mechanisms of the pathogen, the stage of the disease, and the pathology associated with the infection. This work compiles evidence supported by functional assays on the roles of different subsets of MDSCs in acute and chronic infections, including pathogen-associated malignancies, and discusses strategies to modulate MDSC dynamics to benefit the host.
Collapse
Affiliation(s)
- Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493, Greifswald, Insel Riems, Germany.
- Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany.
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany.
| | - Estibaliz Glaría
- Nuclear Receptor Group, Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Av. Diagonal, 643, 3rd floor, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | | | | | - Gennadiy Zelinskyy
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Benoit Favier
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, CEA, Université Paris Sud 11, INSERM U1184, IBJF, Fontenay-aux-Roses, France
| | - Anurag Singh
- University Children's Hospital and Interdisciplinary Center for Infectious Diseases, University of Tübingen, Tübingen, Germany
| | - Jan Ehrchen
- Department of Dermatology, University Hospital Münster, Münster, Germany
| | - Cornelia Gujer
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zurich, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zurich, Switzerland
| | - Margarida Saraiva
- i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Yahya Sohrabi
- Molecular and Translational Cardiology, Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Ana E Sousa
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Annabel F Valledor
- Nuclear Receptor Group, Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Av. Diagonal, 643, 3rd floor, 08028, Barcelona, Spain.
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain.
| |
Collapse
|
77
|
Remot A, Doz E, Winter N. Neutrophils and Close Relatives in the Hypoxic Environment of the Tuberculous Granuloma: New Avenues for Host-Directed Therapies? Front Immunol 2019; 10:417. [PMID: 30915076 PMCID: PMC6423059 DOI: 10.3389/fimmu.2019.00417] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) is one of the most prevalent lung infections of humans and kills ~1.7 million people each year. TB pathophysiology is complex with a central role played by granuloma where a delicate balance takes place to both constrain bacilli and prevent excessive inflammation that may destroy lung functions. Neutrophils reach the lung in waves following first encounter with bacilli and contribute both to early Mtb elimination and late deleterious inflammation. The hypoxic milieu where cells and bacilli cohabit inside the granuloma favors metabolism changes and the impact on TB infection needs to be more thoroughly understood. At the cellular level while the key role of the alveolar macrophage has long been established, behavior of neutrophils in the hypoxic granuloma remains poorly explored. This review will bring to the front new questions that are now emerging regarding neutrophils activity in TB. Are different neutrophil subsets involved in Mtb infection and how? How do neutrophils and close relatives contribute to shaping the granuloma immune environment? What is the role of hypoxia and hypoxia induced factors inside granuloma on neutrophil fate and functions and TB pathophysiology? Addressing these questions is key to the development of innovative host-directed therapies to fight TB.
Collapse
Affiliation(s)
- Aude Remot
- INRA, Universite de Tours, UMR Infectiologie et Sante Publique, Nouzilly, France
| | - Emilie Doz
- INRA, Universite de Tours, UMR Infectiologie et Sante Publique, Nouzilly, France
| | - Nathalie Winter
- INRA, Universite de Tours, UMR Infectiologie et Sante Publique, Nouzilly, France
| |
Collapse
|
78
|
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
|
79
|
Chinta KC, Rahman MA, Saini V, Glasgow JN, Reddy VP, Lever JM, Nhamoyebonde S, Leslie A, Wells RM, Traylor A, Madansein R, Siegal GP, Antony VB, Deshane J, Wells G, Nargan K, George JF, Ramdial PK, Agarwal A, Steyn AJC. Microanatomic Distribution of Myeloid Heme Oxygenase-1 Protects against Free Radical-Mediated Immunopathology in Human Tuberculosis. Cell Rep 2018; 25:1938-1952.e5. [PMID: 30428359 PMCID: PMC6250977 DOI: 10.1016/j.celrep.2018.10.073] [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: 10/30/2017] [Revised: 07/18/2018] [Accepted: 10/19/2018] [Indexed: 11/26/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is a cytoprotective enzyme that controls inflammatory responses and redox homeostasis; however, its role during pulmonary tuberculosis (TB) remains unclear. Using freshly resected human TB lung tissue, we examined the role of HO-1 within the cellular and pathological spectrum of TB. Flow cytometry and histopathological analysis of human TB lung tissues showed that HO-1 is expressed primarily in myeloid cells and that HO-1 levels in these cells were directly proportional to cytoprotection. HO-1 mitigates TB pathophysiology by diminishing myeloid cell-mediated oxidative damage caused by reactive oxygen and/or nitrogen intermediates, which control granulocytic karyorrhexis to generate a zonal HO-1 response. Using whole-body or myeloid-specific HO-1-deficient mice, we demonstrate that HO-1 is required to control myeloid cell infiltration and inflammation to protect against TB progression. Overall, this study reveals that zonation of HO-1 in myeloid cells modulates free-radical-mediated stress, which regulates human TB immunopathology.
Collapse
Affiliation(s)
- Krishna C Chinta
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Vikram Saini
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Joel N Glasgow
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Vineel P Reddy
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremie M Lever
- Nephrology Research and Training Center, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | - Ryan M Wells
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Amie Traylor
- Nephrology Research and Training Center, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Gene P Siegal
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Veena B Antony
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jessy Deshane
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gordon Wells
- Africa Health Research Institute, Durban 4001, South Africa
| | | | - James F George
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Pratistadevi K Ramdial
- Department of Anatomical Pathology, NHLS, Inkosi Albert Luthuli Central Hospital, University of KwaZulu-Natal, Durban 4091, South Africa
| | - Anupam Agarwal
- Nephrology Research and Training Center, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Veterans Affairs, Birmingham, AL 35294, USA
| | - Adrie J C Steyn
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Africa Health Research Institute, Durban 4001, South Africa; UAB Center for AIDS Research, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Center for Free Radical Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
80
|
Goletti D, Lindestam Arlehamn CS, Scriba TJ, Anthony R, Cirillo DM, Alonzi T, Denkinger CM, Cobelens F. Can we predict tuberculosis cure? What tools are available? Eur Respir J 2018; 52:13993003.01089-2018. [PMID: 30361242 DOI: 10.1183/13993003.01089-2018] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/24/2018] [Indexed: 01/08/2023]
Abstract
Antibiotic treatment of tuberculosis takes ≥6 months, putting a major burden on patients and health systems in large parts of the world. Treatment beyond 2 months is needed to prevent tuberculosis relapse by clearing remaining, drug-tolerant Mycobacterium tuberculosis bacilli. However, the majority of patients treated for only 2-3 months will cure without relapse and do not need prolonged treatment. Assays that can identify these patients at an early stage of treatment may significantly help reduce the treatment burden, while a test to identify those patients who will fail treatment may help target host-directed therapies.In this review we summarise the state of the art with regard to discovery of biomarkers that predict relapse-free cure for pulmonary tuberculosis. Positron emission tomography/computed tomography scanning to measure pulmonary inflammation enhances our understanding of "cure". Several microbiological and immunological markers seem promising; however, they still need a formal validation. In parallel, new research strategies are needed to generate reliable tests.
Collapse
Affiliation(s)
- Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases "L. Spallanzani" IRCCS, Dept of Epidemiology and Preclinical Research, Rome, Italy
| | | | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Dept of Pathology, University of Cape Town, Cape Town, South Africa
| | - Richard Anthony
- National Institute for Public Health and the Environment (RIVM), Utrecht, The Netherlands
| | - Daniela Maria Cirillo
- Emerging Bacterial Pathogens Unit, San Raffaele Scientific Institute, HSR, Division of Immunology and Infectious Diseases Milan, Milan, Italy
| | - Tonino Alonzi
- Translational Research Unit, National Institute for Infectious Diseases "L. Spallanzani" IRCCS, Dept of Epidemiology and Preclinical Research, Rome, Italy
| | | | - Frank Cobelens
- Dept of Global Health and Amsterdam Institute for Global Health and Development, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
81
|
Agrawal N, Streata I, Pei G, Weiner J, Kotze L, Bandermann S, Lozza L, Walzl G, du Plessis N, Ioana M, Kaufmann SHE, Dorhoi A. Human Monocytic Suppressive Cells Promote Replication of Mycobacterium tuberculosis and Alter Stability of in vitro Generated Granulomas. Front Immunol 2018; 9:2417. [PMID: 30405617 PMCID: PMC6205994 DOI: 10.3389/fimmu.2018.02417] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 10/01/2018] [Indexed: 12/20/2022] Open
Abstract
Tuberculosis (TB) has tremendous public health relevance. It most frequently affects the lung and is characterized by the development of unique tissue lesions, termed granulomas. These lesions encompass various immune populations, with macrophages being most extensively investigated. Myeloid derived suppressor cells (MDSCs) have been recently identified in TB patients, both in the circulation and at the site of infection, however their interactions with Mycobacterium tuberculosis (Mtb) and their impact on granulomas remain undefined. We generated human monocytic MDSCs and observed that their suppressive capacities are retained upon Mtb infection. We employed an in vitro granuloma model, which mimics human TB lesions to some extent, with the aim of analyzing the roles of MDSCs within granulomas. MDSCs altered the structure of and affected bacterial containment within granuloma-like structures. These effects were partly controlled through highly abundant secreted IL-10. Compared to macrophages, MDSCs activated primarily the NF-κB and MAPK pathways and the latter largely contributed to the release of IL-10 and replication of bacteria within in vitro generated granulomas. Moreover, MDSCs upregulated PD-L1 and suppressed proliferation of lymphocytes, albeit with negligible effects on Mtb replication. Further comprehensive characterization of MDSCs in TB will contribute to a better understanding of disease pathogenesis and facilitate the design of novel immune-based interventions for this deadly infection.
Collapse
Affiliation(s)
- Neha Agrawal
- Max Planck Institute for Infection Biology, Department of Immunology, Berlin, Germany
| | - Ioana Streata
- University of Medicine and Pharmacy Craiova, Human Genomics Laboratory, Craiova, Romania
| | - Gang Pei
- Max Planck Institute for Infection Biology, Department of Immunology, Berlin, Germany
| | - January Weiner
- Max Planck Institute for Infection Biology, Department of Immunology, Berlin, Germany
| | - Leigh Kotze
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, SAMRC Centre for Tuberculosis Research, DST and NRF Centre of Excellence for Biomedical TB Research, Stellenbosch University, Tygerberg, South Africa
| | - Silke Bandermann
- Max Planck Institute for Infection Biology, Department of Immunology, Berlin, Germany
| | - Laura Lozza
- Max Planck Institute for Infection Biology, Department of Immunology, Berlin, Germany
| | - Gerhard Walzl
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, SAMRC Centre for Tuberculosis Research, DST and NRF Centre of Excellence for Biomedical TB Research, Stellenbosch University, Tygerberg, South Africa
| | - Nelita du Plessis
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, SAMRC Centre for Tuberculosis Research, DST and NRF Centre of Excellence for Biomedical TB Research, Stellenbosch University, Tygerberg, South Africa
| | - Mihai Ioana
- University of Medicine and Pharmacy Craiova, Human Genomics Laboratory, Craiova, Romania
| | - Stefan H E Kaufmann
- Max Planck Institute for Infection Biology, Department of Immunology, Berlin, Germany
| | - Anca Dorhoi
- Max Planck Institute for Infection Biology, Department of Immunology, Berlin, Germany.,Institute of Immunology, Federal Research Institute for Animal Health, Friedrich-Loeffler-Institut, Insel Riems, Germany.,Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| |
Collapse
|
82
|
du Plessis N, Kotze LA, Leukes V, Walzl G. Translational Potential of Therapeutics Targeting Regulatory Myeloid Cells in Tuberculosis. Front Cell Infect Microbiol 2018; 8:332. [PMID: 30298121 PMCID: PMC6160538 DOI: 10.3389/fcimb.2018.00332] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 08/28/2018] [Indexed: 12/11/2022] Open
Abstract
Despite recent advances in tuberculosis (TB) drug development and availability, successful antibiotic treatment is challenged by the parallel development of antimicrobial resistance. As a result, new approaches toward improving TB treatment have been proposed in an attempt to reduce the high TB morbidity and mortality rates. Host-directed therapies (HDTs), designed to modulate host immune components, provide an alternative approach for improving treatment outcome in both non-communicable and infectious diseases. Many candidate immunotherapeutics, designed to target regulatory myeloid immune components in cancer, have so far proven to be of value as repurposed HDT in TB. Several of these studies do however lack detailed description of the mechanism or host pathway affected by TB HDT treatment. In this review, we present an argument for greater appreciation of the role of regulatory myeloid cells, such as myeloid-derived suppressor cells (MDSC), as potential targets for the development of candidate TB HDT compounds. We discuss the role of MDSC in the context of Mycobacterium tuberculosis infection and disease, focussing primarily on their specific cellular functions and highlight the impact of HDTs on MDSC frequency and function.
Collapse
Affiliation(s)
- Nelita du Plessis
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Leigh A Kotze
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Vinzeigh Leukes
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
83
|
Stoll H, Ost M, Singh A, Mehling R, Neri D, Schäfer I, Velic A, Macek B, Kretschmer D, Weidenmaier C, Hector A, Handgretinger R, Götz F, Peschel A, Hartl D, Rieber N. Staphylococcal Enterotoxins Dose-Dependently Modulate the Generation of Myeloid-Derived Suppressor Cells. Front Cell Infect Microbiol 2018; 8:321. [PMID: 30271756 PMCID: PMC6146041 DOI: 10.3389/fcimb.2018.00321] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/22/2018] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus is one of the major human bacterial pathogens causing a broad spectrum of serious infections. Myeloid-derived suppressor cells (MDSC) represent an innate immune cell subset capable of regulating host-pathogen interactions, yet their role in the pathogenesis of S. aureus infections remains incompletely defined. The aim of this study was to determine the influence of different S. aureus strains and associated virulence factors on human MDSC generation. Using an in vitro MDSC generation assay we demonstrate that low concentrations of supernatants of different S. aureus strains led to an induction of functional MDSC, whereas increased concentrations, conversely, reduced MDSC numbers. The concentration-dependent reduction of MDSC correlated with T cell proliferation and cytotoxicity. Several findings supported a role for staphylococcal enterotoxins in modulating MDSC generation. Staphylococcal enterotoxins recapitulated concentration-dependent MDSC induction and inhibition, T cell proliferation and cytotoxicity, while an enterotoxin-deficient S. aureus strain largely failed to alter MDSC. Taken together, we identified staphylococcal enterotoxins as main modulators of MDSC generation. The inhibition of MDSC generation by staphylococcal enterotoxins might represent a novel therapeutic target in S. aureus infections and beyond in non-infectious conditions, such as cancer.
Collapse
Affiliation(s)
- Hartmut Stoll
- Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Michael Ost
- Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Anurag Singh
- Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Roman Mehling
- Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Davide Neri
- Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Iris Schäfer
- Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Ana Velic
- Proteome Center Tuebingen, Interfaculty Institute for Cell Biology, University of Tuebingen, Tuebingen, Germany
| | - Boris Macek
- Proteome Center Tuebingen, Interfaculty Institute for Cell Biology, University of Tuebingen, Tuebingen, Germany
| | - Dorothee Kretschmer
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Germany
| | - Christopher Weidenmaier
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Germany
| | - Andreas Hector
- Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | | | - Friedrich Götz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Germany
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Germany.,German Centre for Infection Research (DZIF), Partner Site Tuebingen, Tuebingen, Germany
| | - Dominik Hartl
- Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany.,German Centre for Infection Research (DZIF), Partner Site Tuebingen, Tuebingen, Germany
| | - Nikolaus Rieber
- Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany.,German Centre for Infection Research (DZIF), Partner Site Tuebingen, Tuebingen, Germany.,Department of Pediatrics, Kinderklinik Muenchen Schwabing, Klinikum Schwabing, StKM GmbH und Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
84
|
Sacchi A, Tumino N, Grassi G, Casetti R, Cimini E, Bordoni V, Ammassari A, Antinori A, Agrati C. A new procedure to analyze polymorphonuclear myeloid derived suppressor cells in cryopreserved samples cells by flow cytometry. PLoS One 2018; 13:e0202920. [PMID: 30161175 PMCID: PMC6117014 DOI: 10.1371/journal.pone.0202920] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 08/10/2018] [Indexed: 12/20/2022] Open
Abstract
Myeloid derived suppressor cells (MDSC) is a heterogeneous subset of immature and mature cells of the myeloid lineage, undergoing expansion during pathologic conditions, and able to perform strong immune suppressive functions. It has been shown that cryopreservation selectively impacts the polimorphonuclear (PMN) MDSC viability and recovery, and alters the correct analysis of MDSC subsets. In laboratory practice, cryopreservation is often inevitable, in particular in multicenter studies where samples have to be shipped to a centralized laboratory. Aim of the present work was to set out a new protocol to evaluate the frequency of PMN-MDSC in thawed cells by flow-cytometry. PBMC were isolated from HIV+ patients and healthy donors, and were cryopreserved for at least ten days. After thawing, two different protocols were used: 1. standard protocol (SP) consisting of staining with the antibodies mix and then fixing with formalin 1%; 2. thawed protocol (TP) in which fixation foregoes the staining with the antibodies mix. Results showed that PMN-MDSC frequency in ex vivo PBMC evaluated by means TP was comparable to that analysed by SP, indicating that the protocol did not alter PMN-MDSC quantification in ex vivo cells. We then demonstrated that PMN-MDSC frequency in thawed PBMC tested by TP was almost identical to the frequency obtained in ex vivo cells evaluated by using SP. However, we observed that after three hours of culture post-thawing, PMN-MDSC were not assessable anymore with both SP and TP. In conclusion, we herein demonstrated that fixing PBMC soon after thawing and before antibody staining allows preservation of PMN-MDSC integrity and a reliable cells quantification. Thus, it is possible to phenotipically identify PMN-MDSC in cryopreserved PBMC, consenting adequate test precision and accuracy as well as making multicentre research more feasible.
Collapse
Affiliation(s)
- Alessandra Sacchi
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
- * E-mail:
| | - Nicola Tumino
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Germana Grassi
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Rita Casetti
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Eleonora Cimini
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Veronica Bordoni
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Adriana Ammassari
- Clinical Department, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Andrea Antinori
- Clinical Department, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Chiara Agrati
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| |
Collapse
|
85
|
Abstract
Tuberculosis has a much shorter incubation period than is widely thought, say Marcel A Behr and colleagues, and this has implications for prioritising research and public health strategies
Collapse
Affiliation(s)
- Marcel A Behr
- McGill International TB Centre, Infectious Diseases and Immunity in Global Health Program, McGill University Health Centre Research Institute, Montreal H4A 3J1, Canada
| | - Paul H Edelstein
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Lalita Ramakrishnan
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| |
Collapse
|
86
|
Myeloid-Derived Suppressor Cells and Pulmonary Hypertension. Int J Mol Sci 2018; 19:ijms19082277. [PMID: 30081463 PMCID: PMC6121540 DOI: 10.3390/ijms19082277] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 01/04/2023] Open
Abstract
Myeloid–derived suppressor cells (MDSCs) comprised a heterogeneous subset of bone marrow–derived myeloid cells, best studied in cancer research, that are increasingly implicated in the pathogenesis of pulmonary vascular remodeling and the development of pulmonary hypertension. Stem cell transplantation represents one extreme interventional strategy for ablating the myeloid compartment but poses a number of translational challenges. There remains an outstanding need for additional therapeutic targets to impact MDSC function, including the potential to alter interactions with innate and adaptive immune subsets, or alternatively, alter trafficking receptors, metabolic pathways, and transcription factor signaling with readily available and safe drugs. In this review, we summarize the current literature on the role of myeloid cells in the development of pulmonary hypertension, first in pulmonary circulation changes associated with myelodysplastic syndromes, and then by examining intrinsic myeloid cell changes that contribute to disease progression in pulmonary hypertension. We then outline several tractable targets and pathways relevant to pulmonary hypertension via MDSC regulation. Identifying these MDSC-regulated effectors is part of an ongoing effort to impact the field of pulmonary hypertension research through identification of myeloid compartment-specific therapeutic applications in the treatment of pulmonary vasculopathies.
Collapse
|
87
|
Leisching GR. Susceptibility to Tuberculosis Is Associated With PI3K-Dependent Increased Mobilization of Neutrophils. Front Immunol 2018; 9:1669. [PMID: 30065729 PMCID: PMC6056613 DOI: 10.3389/fimmu.2018.01669] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/04/2018] [Indexed: 12/19/2022] Open
Abstract
Neutrophilia is a condition commonly observed in patients with late-stage tuberculosis, but evidence suggests that increased neutrophil influx begins early after infection in susceptible hosts and functions to promote a nutrient-replete niche that promotes Mycobacterium tuberculosis survival and persistence. As the disease progresses, an increase in the number of neutrophil-like cells is observed, all of which exhibit characteristics associated with (i) phenotypic and biochemical features of immaturity, (ii) the inability to activate T-cells, (iii) hyper-inflammation, and (iv) prolonged survival. Transcriptomics reveal a common set of molecules associated with the PI3–Kinase pathway that are dysregulated in patients with active tuberculosis. Closer inspection of their individual biological roles reveal their ability to modulate the IL-17/G–CSF axis, induce leukocyte receptor activation, and regulate apoptosis and motility. This review draws attention to neutrophil hyper-reactivity as a driving force for both the establishment and progression of tuberculosis disease in susceptible individuals.
Collapse
Affiliation(s)
- Gina R Leisching
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
88
|
Payne KK, Aqbi HF, Butler SE, Graham L, Keim RC, Wan W, Idowu MO, Bear HD, Wang XY, Manjili MH. Gr1 -/low CD11b -/low MHCII + myeloid cells boost T cell anti-tumor efficacy. J Leukoc Biol 2018; 104:1215-1228. [PMID: 29985529 PMCID: PMC6258302 DOI: 10.1002/jlb.5a0717-276rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 01/04/2023] Open
Abstract
Conventional APCs that express MHC class II (MHCII) and co-stimulatory molecules include dendritic cells (DCs) and macrophages. Beyond these conventional APCs, immune stimulatory cells have been more recently shown to extend to a class of atypical APCs, composed of mast cells, basophils, and eosinophils. Here, we describe a unique type of APC, Gr1-/low CD11b-/low cells with a granularity and size characteristic of myeloid cells and with the ability to present Ag for crosspresentation. These cells constitutively express MHCII and the costimulatory molecules, CD80, CD86, and CD40. They do not express pan markers of myeloid DCs (CD11c), plasmacytoid DCs (Ly6C), or macrophages (F4/80), and their frequency is inversely correlated with myeloid-derived suppressor cells (MDSCs) in tumor-bearing mice. Among splenocytes, they are more abundant than DCs and macrophages, and they exhibit antitumor immune stimulatory function at a steady state without further activation, ex vivo. They are also found within the tumor bed where they retain their immune stimulatory function. Our findings suggest the use of these novel APCs in additional preclinical studies to further investigate their utility in APC-based cancer immunotherapies.
Collapse
Affiliation(s)
- Kyle K Payne
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Department of Immunology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Hussein F Aqbi
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Savannah E Butler
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Laura Graham
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Rebecca C Keim
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Wen Wan
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Department of Biostatistics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Michael O Idowu
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Harry D Bear
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Xiang-Yang Wang
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
89
|
Medina E, Hartl D. Myeloid-Derived Suppressor Cells in Infection: A General Overview. J Innate Immun 2018; 10:407-413. [PMID: 29945134 DOI: 10.1159/000489830] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 04/23/2018] [Indexed: 01/09/2023] Open
Abstract
After initial infection, the immune response that serves to restrict the invading pathogen needs to be tightly calibrated in order to avoid collateral immunopathological damage. This calibration is performed by specialized suppressor mechanisms, which are capable of dampening overwhelming or unremitting inflammation in order to prevent tissue damage. Myeloid-derived suppressor cells (MDSC) are emerging as key players in counter-balancing inflammatory responses and pathogenesis during infection. However, some pathogens are able to exploit the suppressive activities of MDSC to favor pathogen persistence and chronic infections. In this article, we review the current knowledge about the importance of MDSC in the context of bacterial, virus, parasites, and fungal infections.
Collapse
Affiliation(s)
- Eva Medina
- Helmholtz Centre for Infection Research, Infection Immunology Research Group, Braunschweig,
| | - Dominik Hartl
- Children's Hospital, University of Tübingen, Tübingen, Germany
| |
Collapse
|
90
|
Lange C, Alghamdi WA, Al-Shaer MH, Brighenti S, Diacon AH, DiNardo AR, Grobbel HP, Gröschel MI, von Groote-Bidlingmaier F, Hauptmann M, Heyckendorf J, Köhler N, Kohl TA, Merker M, Niemann S, Peloquin CA, Reimann M, Schaible UE, Schaub D, Schleusener V, Thye T, Schön T. Perspectives for personalized therapy for patients with multidrug-resistant tuberculosis. J Intern Med 2018; 284:163-188. [PMID: 29806961 DOI: 10.1111/joim.12780] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
According to the World Health Organization (WHO), tuberculosis is the leading cause of death attributed to a single microbial pathogen worldwide. In addition to the large number of patients affected by tuberculosis, the emergence of Mycobacterium tuberculosis drug-resistance is complicating tuberculosis control in many high-burden countries. During the past 5 years, the global number of patients identified with multidrug-resistant tuberculosis (MDR-TB), defined as bacillary resistance at least against rifampicin and isoniazid, the two most active drugs in a treatment regimen, has increased by more than 20% annually. Today we experience a historical peak in the number of patients affected by MDR-TB. The management of MDR-TB is characterized by delayed diagnosis, uncertainty of the extent of bacillary drug-resistance, imprecise standardized drug regimens and dosages, very long duration of therapy and high frequency of adverse events which all translate into a poor prognosis for many of the affected patients. Major scientific and technological advances in recent years provide new perspectives through treatment regimens tailor-made to individual needs. Where available, such personalized treatment has major implications on the treatment outcomes of patients with MDR-TB. The challenge now is to bring these adances to those patients that need them most.
Collapse
Affiliation(s)
- C Lange
- Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- Tuberculosis Unit, German Center for Infection Research (DZIF), Borstel, Germany
- International Health/Infectious Diseases, University of Lübeck, Lübeck, Germany
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - W A Alghamdi
- Department of Pharmacotherapy and Translational Research, Infectious Disease Pharmacokinetics Laboratory, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - M H Al-Shaer
- Department of Pharmacotherapy and Translational Research, Infectious Disease Pharmacokinetics Laboratory, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - S Brighenti
- Department of Medicine, Center for Infectious Medicine (CIM), Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - A H Diacon
- Task Applied Science, Bellville, South Africa
- Division of Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - A R DiNardo
- Section of Global and Immigrant Health, Baylor College of Medicine, Houston, TX, USA
| | - H P Grobbel
- Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- Tuberculosis Unit, German Center for Infection Research (DZIF), Borstel, Germany
- International Health/Infectious Diseases, University of Lübeck, Lübeck, Germany
| | - M I Gröschel
- Department of Pumonary Diseases & Tuberculosis, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Molecular and Experimental Mycobacteriology, National Reference Center for Mycobacteria, Research Center Borstel, Borstel, Germany
| | | | - M Hauptmann
- Tuberculosis Unit, German Center for Infection Research (DZIF), Borstel, Germany
- Cellular Microbiology, Research Center Borstel, Borstel, Germany
| | - J Heyckendorf
- Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- Tuberculosis Unit, German Center for Infection Research (DZIF), Borstel, Germany
- International Health/Infectious Diseases, University of Lübeck, Lübeck, Germany
| | - N Köhler
- Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- Tuberculosis Unit, German Center for Infection Research (DZIF), Borstel, Germany
- International Health/Infectious Diseases, University of Lübeck, Lübeck, Germany
| | - T A Kohl
- Molecular and Experimental Mycobacteriology, National Reference Center for Mycobacteria, Research Center Borstel, Borstel, Germany
| | - M Merker
- Molecular and Experimental Mycobacteriology, National Reference Center for Mycobacteria, Research Center Borstel, Borstel, Germany
| | - S Niemann
- Tuberculosis Unit, German Center for Infection Research (DZIF), Borstel, Germany
- Molecular and Experimental Mycobacteriology, National Reference Center for Mycobacteria, Research Center Borstel, Borstel, Germany
| | - C A Peloquin
- Department of Pharmacotherapy and Translational Research, Infectious Disease Pharmacokinetics Laboratory, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - M Reimann
- Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- Tuberculosis Unit, German Center for Infection Research (DZIF), Borstel, Germany
- International Health/Infectious Diseases, University of Lübeck, Lübeck, Germany
| | - U E Schaible
- Tuberculosis Unit, German Center for Infection Research (DZIF), Borstel, Germany
- Cellular Microbiology, Research Center Borstel, Borstel, Germany
- Biochemical Microbiology & Immunochemistry, University of Lübeck, Lübeck, Germany
- LRA INFECTIONS'21, Borstel, Germany
| | - D Schaub
- Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- Tuberculosis Unit, German Center for Infection Research (DZIF), Borstel, Germany
- International Health/Infectious Diseases, University of Lübeck, Lübeck, Germany
| | - V Schleusener
- Molecular and Experimental Mycobacteriology, National Reference Center for Mycobacteria, Research Center Borstel, Borstel, Germany
| | - T Thye
- Department of Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - T Schön
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- Department of Clinical Microbiology and Infectious Diseases, Kalmar County Hospital, Linköping University, Linköping, Sweden
| |
Collapse
|
91
|
Abstract
Protective immunity in tuberculosis (TB) is subject of debate in the TB research community, as this is key to fully understand TB pathogenesis and to develop new promising tools for TB diagnosis and prognosis as well as a more efficient TB vaccine. IFN-γ producing CD4+ T cells are key in TB control, but may not be sufficient to provide protection. Additional subsets have been identified that contribute to protection such as multifunctional and cytolytic T-cell subsets, including classical and nonclassical T cells as well as novel innate immune cell subsets resulting from trained immunity. However, to define protective immune responses against TB, the complexity of balancing TB immunity also has to be considered. In this review, insights into effector cell immunity and how this is modulated by regulatory cells, associated comorbidities and the host microbiome, is discussed. We systematically map how different suppressive immune cell subsets may affect effector cell responses at the local site of infection. We also dissect how common comorbidities such as HIV, helminths and diabetes may bias protective TB immunity towards pathogenic and regulatory responses. Finally, also the composition and diversity of the microbiome in the lung and gut could affect host TB immunity. Understanding these various aspects of the immunological balance in the human host is fundamental to prevent TB infection and disease.
Collapse
Affiliation(s)
- Susanna Brighenti
- Karolinska Institutet, Department of Medicine, Center for Infectious Medicine (CIM), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Simone A. Joosten
- Leiden University Medical Center, Department of Infectious Diseases, Leiden, The Netherlands
| |
Collapse
|
92
|
Ben-Meir K, Twaik N, Baniyash M. Plasticity and biological diversity of myeloid derived suppressor cells. Curr Opin Immunol 2018; 51:154-161. [PMID: 29614426 DOI: 10.1016/j.coi.2018.03.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 01/27/2018] [Accepted: 03/13/2018] [Indexed: 10/17/2022]
Abstract
Myeloid derived suppressor cells (MDSCs) are immature myeloid cells characterized by diverse phenotypes and functions. They impair effector functions of immune cells and promote tumor growth, angiogenesis, and tissue damage. In pathologies characterized by chronic inflammation, MDSCs are arrested in their immature state and migrate from the bone marrow to the periphery and to the site of inflammation, where they mediate immunosuppression. When reaching new environments, which exhibit a different array of cytokines, chemokines, and pro-inflammatory mediators, MDSCs sense and adapt to the altered micro-environment by virtue of acquiring different suppressive features/functions that involve changing their cell fate, surface receptors, metabolism and intracellular as well as secreted molecules. This review summarizes some of the latest publications highlighting various layers of MDSC plasticity in relation to different pathologies. We discuss treatments capitalizing on MDSC plasticity aimed at combating MDSCs or manipulating their suppressive activity for improved therapy.
Collapse
Affiliation(s)
- Kerem Ben-Meir
- The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Nira Twaik
- The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Michal Baniyash
- The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel.
| |
Collapse
|
93
|
Tiberi S, du Plessis N, Walzl G, Vjecha MJ, Rao M, Ntoumi F, Mfinanga S, Kapata N, Mwaba P, McHugh TD, Ippolito G, Migliori GB, Maeurer MJ, Zumla A. Tuberculosis: progress and advances in development of new drugs, treatment regimens, and host-directed therapies. THE LANCET. INFECTIOUS DISEASES 2018; 18:e183-e198. [PMID: 29580819 DOI: 10.1016/s1473-3099(18)30110-5] [Citation(s) in RCA: 242] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/19/2017] [Accepted: 01/02/2018] [Indexed: 12/16/2022]
Abstract
Tuberculosis remains the world's leading cause of death from an infectious disease, responsible for an estimated 1 674 000 deaths annually. WHO estimated 600 000 cases of rifampicin-resistant tuberculosis in 2016-of which 490 000 were multidrug resistant (MDR), with less than 50% survival after receiving recommended treatment regimens. Concerted efforts of stakeholders, advocates, and researchers are advancing further development of shorter course, more effective, safer, and better tolerated treatment regimens. We review the developmental pipeline and landscape of new and repurposed tuberculosis drugs, treatment regimens, and host-directed therapies (HDTs) for drug-sensitive and drug-resistant tuberculosis. 14 candidate drugs for drug-susceptible, drug-resistant, and latent tuberculosis are in clinical stages of drug development; nine are novel in phase 1 and 2 trials, and three new drugs are in advanced stages of development for MDR tuberculosis. Specific updates are provided on clinical trials of bedaquiline, delamanid, pretomanid, and other licensed or repurposed drugs that are undergoing investigation, including trials aimed at shortening duration of tuberculosis treatment, improving treatment outcomes and patient adherence, and reducing toxic effects. Ongoing clinical trials for shortening tuberculosis treatment duration, improving treatment outcomes in MDR tuberculosis, and preventing disease in people with latent tuberculosis infection are reviewed. A range of HDTs and immune-based treatments are under investigation as adjunctive therapy for shortening duration of therapy, preventing permanent lung injury, and improving treatment outcomes of MDR tuberculosis. We discuss the HDT development pipeline, ongoing clinical trials, and translational research efforts for adjunct tuberculosis treatment.
Collapse
Affiliation(s)
- Simon Tiberi
- Division of Infection, Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Nelita du Plessis
- South African Department of Science and Technology, and National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- South African Department of Science and Technology, and National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | | | - Martin Rao
- Champalimaud Foundation, Lisbon, Portugal; Krankenhaus Nordwest, Frankfurt, Germany
| | - Francine Ntoumi
- Fondation Congolaise pour la Recherche Medicale, and Faculte des Sciences et Techniques, Universite M Ngouabi, Brazzaville, Republic of the Congo
| | - Sayoki Mfinanga
- National Institute for Medical Research, Muhimbili Medical Research Centre, Dar es Salaam, Tanzania
| | - Nathan Kapata
- Institute of Public Health, Ministry of Health, Lusaka, Zambia
| | - Peter Mwaba
- UNZA-UCLMS Research and Training Programme, and Apex University, Lusaka, Zambia
| | - Timothy D McHugh
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, UK
| | - Giuseppe Ippolito
- National Institute for Infectious Disease, L Spallanzani, Rome, Italy
| | - Giovanni Battista Migliori
- World Health Organization Collaborating Centre for Tuberculosis and Lung Diseases, Maugeri Care and Research Institute, Istituto di Ricovero e Cura a Carattere Sceintifico, Tradate, Italy
| | - Markus J Maeurer
- Champalimaud Foundation, Lisbon, Portugal; Krankenhaus Nordwest, Frankfurt, Germany
| | - Alimuddin Zumla
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, UK; National Institute of Health and Research Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, UK.
| |
Collapse
|
94
|
IFN-γ decreased the suppressive function of CD33+HLA-DRlow myeloid cells through down-regulation of PD-1/PD-L2 signaling pathway. Mol Immunol 2018; 94:107-120. [DOI: 10.1016/j.molimm.2017.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/11/2017] [Accepted: 10/11/2017] [Indexed: 11/21/2022]
|
95
|
Dorhoi A, Du Plessis N. Monocytic Myeloid-Derived Suppressor Cells in Chronic Infections. Front Immunol 2018; 8:1895. [PMID: 29354120 PMCID: PMC5758551 DOI: 10.3389/fimmu.2017.01895] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/11/2017] [Indexed: 01/04/2023] Open
Abstract
Heterogeneous populations of myeloid regulatory cells (MRC), including monocytes, macrophages, dendritic cells, and neutrophils, are found in cancer and infectious diseases. The inflammatory environment in solid tumors as well as infectious foci with persistent pathogens promotes the development and recruitment of MRC. These cells help to resolve inflammation and establish host immune homeostasis by restricting T lymphocyte function, inducing regulatory T cells and releasing immune suppressive cytokines and enzyme products. Monocytic MRC, also termed monocytic myeloid-derived suppressor cells (M-MDSC), are bona fide phagocytes, capable of pathogen internalization and persistence, while exerting localized suppressive activity. Here, we summarize molecular pathways controlling M-MDSC genesis and functions in microbial-induced non-resolved inflammation and immunopathology. We focus on the roles of M-MDSC in infections, including opportunistic extracellular bacteria and fungi as well as persistent intracellular pathogens, such as mycobacteria and certain viruses. Better understanding of M-MDSC biology in chronic infections and their role in antimicrobial immunity, will advance development of novel, more effective and broad-range anti-infective therapies.
Collapse
Affiliation(s)
- Anca Dorhoi
- Institute of Immunology, Bundesforschungsinstitut für Tiergesundheit, Friedrich-Loeffler-Institut (FLI), Insel Riems, Germany.,Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany.,Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Nelita Du Plessis
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, SAMRC Centre for Tuberculosis Research, DST and NRF Centre of Excellence for Biomedical TB Research, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
96
|
Neutrophils and PMN-MDSC: Their biological role and interaction with stromal cells. Semin Immunol 2017; 35:19-28. [PMID: 29254756 DOI: 10.1016/j.smim.2017.12.004] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/08/2017] [Indexed: 02/06/2023]
Abstract
Neutrophils and polymorphonucler myeloid-derived suppressor cells (PMN-MDSC) share origin and many morphological and phenotypic features. However, they have different biological role. Neutrophils are one of the major mechanisms of protection against invading pathogens, whereas PMN-MDSC have immune suppressive activity and restrict immune responses in cancer, chronic infectious disease, trauma, sepsis, and many other pathological conditions. Although in healthy adult individuals, PMN-MDSC are not or barely detectable, in patients with cancer and many other diseases they accumulate at various degree and co-exist with neutrophils. Recent advances allow for better distinction of these cells and better understanding of their biological role. Accumulating evidence indicates PMN-MDSC as pathologically activated neutrophils, with important role in regulation of immune responses. In this review, we provide an overview on the definition and characterization of PMN-MDSC and neutrophils, their pathological significance in a variety of diseases, and their interaction with other stromal components.
Collapse
|
97
|
Schaible UE, Linnemann L, Redinger N, Patin EC, Dallenga T. Strategies to Improve Vaccine Efficacy against Tuberculosis by Targeting Innate Immunity. Front Immunol 2017; 8:1755. [PMID: 29312298 PMCID: PMC5732265 DOI: 10.3389/fimmu.2017.01755] [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: 08/21/2017] [Accepted: 11/27/2017] [Indexed: 01/08/2023] Open
Abstract
The global tuberculosis epidemic is the most common cause of death after infectious disease worldwide. Increasing numbers of infections with multi- and extensively drug-resistant variants of the Mycobacterium tuberculosis complex, resistant even to newly discovered and last resort antibiotics, highlight the urgent need for an efficient vaccine. The protective efficacy to pulmonary tuberculosis in adults of the only currently available vaccine, M. bovis BCG, is unsatisfactory and geographically diverse. More importantly, recent clinical studies on new vaccine candidates did not prove to be better than BCG, yet. Here, we propose and discuss novel strategies to improve efficacy of existing anti-tuberculosis vaccines. Modulation of innate immune responses upon vaccination already provided promising results in animal models of tuberculosis. For instance, neutrophils have been shown to influence vaccine efficacy, both, positively and negatively, and stimulate specific antibody secretion. Modulating immune regulatory properties after vaccination such as induction of different types of innate immune cell death, myeloid-derived suppressor or regulatory T cells, production of anti-inflammatory cytokines such as IL-10 may have beneficial effects on protection efficacy. Incorporation of lipid antigens presented via CD1 molecules to T cells have been discussed as a way to enhance vaccine efficacy. Finally, concepts of dendritic cell-based immunotherapies or training the innate immune memory may be exploitable for future vaccination strategies against tuberculosis. In this review, we put a spotlight on host immune networks as potential targets to boost protection by old and new tuberculosis vaccines.
Collapse
Affiliation(s)
- Ulrich E Schaible
- Cellular Microbiology, Priority Program Infections, Research Center Borstel, Borstel, Germany.,Thematic Translation Unit Tuberculosis, German Center for Infection Research, Research Center Borstel, Borstel, Germany
| | - Lara Linnemann
- Cellular Microbiology, Priority Program Infections, Research Center Borstel, Borstel, Germany
| | - Natalja Redinger
- Cellular Microbiology, Priority Program Infections, Research Center Borstel, Borstel, Germany
| | - Emmanuel C Patin
- Cellular Microbiology, Priority Program Infections, Research Center Borstel, Borstel, Germany.,Retroviral Immunology, The Francis Crick Institute, London, United Kingdom
| | - Tobias Dallenga
- Cellular Microbiology, Priority Program Infections, Research Center Borstel, Borstel, Germany.,Thematic Translation Unit Tuberculosis, German Center for Infection Research, Research Center Borstel, Borstel, Germany
| |
Collapse
|
98
|
Herzmann C, Ernst M, Lange C, Stenger S, Kaufmann SHE, Reiling N, Schaberg T, van der Merwe L, Maertzdorf J, for the Tb or not Tb consortium. Pulmonary immune responses to Mycobacterium tuberculosis in exposed individuals. PLoS One 2017; 12:e0187882. [PMID: 29125874 PMCID: PMC5695274 DOI: 10.1371/journal.pone.0187882] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/28/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Blood based Interferon-(IFN)-γ release assays (IGRAs) have a poor predictive value for the development of tuberculosis. This study aimed to investigate the correlation between IGRAs and pulmonary immune responses in tuberculosis contacts in Germany. METHODS IGRAs were performed on bronchoalveolar lavage (BAL) cells and peripheral blood from close healthy contacts of patients with culturally confirmed tuberculosis. Cellular BAL composition was determined by flow cytometry. BAL cells were co-cultured with three strains of Mycobacterium tuberculosis (Mtb) and Mtb derived antigens including Purified Protein Derivative (PPD), 6 kD Early Secretory Antigenic Target (ESAT-6) and 10 kD Culture Filtrate Protein (CFP-10). Levels of 29 cytokines and chemokines were analyzed in the supernatants by multiplex assay. Associations and effects were examined using linear mixed-effects models. RESULTS There were wide variations of inter-individual cytokine levels in BAL cell culture supernatants. Mycobacterial infection and stimulation with PPD showed a clear induction of several macrophage and lymphocyte associated cytokines, reflecting activation of these cell types. No robust correlation between cytokine patterns and blood IGRA status of the donor was observed, except for slightly higher Interleukin-2 (IL-2) responses in BAL cells from IGRA-positive donors upon mycobacterial infection compared to cells from IGRA-negative donors. Stronger correlations were observed when cytokine patterns were stratified according to BAL IGRA status. BAL cells from donors with BAL IGRA-positive responses produced significantly more IFN-γ and IL-2 upon PPD stimulation and mycobacterial infection than cells from BAL IGRA-negative individuals. Correlations between BAL composition and basal cytokine release from unstimulated cells were suggestive of pre-activated lymphocytes but impaired macrophage activity in BAL IGRA-positive donors, in contrast to BAL IGRA-negative donors. CONCLUSIONS In vitro BAL cell cytokine responses to M. tuberculosis antigens or infection do not reflect blood IGRA status but do correlate with stronger cellular responses in BAL IGRA-positive donors. The cytokine patterns observed suggest a pre-activated state of lymphocytes and suppressed macrophage responsiveness in BAL cells from BAL IGRA-positive individuals.
Collapse
Affiliation(s)
| | - Martin Ernst
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
| | - Christoph Lange
- German Center for Infection Research (DZIF), Clinical Tuberculosis Unit, Borstel, Germany
- International Health / Infectious Diseases, University of Lübeck, Lübeck, Germany
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Steffen Stenger
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Stefan H. E. Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Norbert Reiling
- Division of Microbial Interface Biology, Research Center Borstel, Borstel, Germany
| | - Tom Schaberg
- Center of Pneumology, Agaplesion Deaconess Hospital Rotenburg, Rotenburg, Germany
| | - Lize van der Merwe
- Center for Clinical Studies, Research Center Borstel, Borstel, Germany
- LizeStats Consulting, Frankraal, Overstrand, Western Cape, South Africa
| | - Jeroen Maertzdorf
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | | |
Collapse
|
99
|
Domingo-Gonzalez R, Das S, Griffiths KL, Ahmed M, Bambouskova M, Gopal R, Gondi S, Muñoz-Torrico M, Salazar-Lezama MA, Cruz-Lagunas A, Jiménez-Álvarez L, Ramirez-Martinez G, Espinosa-Soto R, Sultana T, Lyons-Weiler J, Reinhart TA, Arcos J, de la Luz Garcia-Hernandez M, Mastrangelo MA, Al-Hammadi N, Townsend R, Balada-Llasat JM, Torrelles JB, Kaplan G, Horne W, Kolls JK, Artyomov MN, Rangel-Moreno J, Zúñiga J, Khader SA. Interleukin-17 limits hypoxia-inducible factor 1α and development of hypoxic granulomas during tuberculosis. JCI Insight 2017; 2:92973. [PMID: 28978810 PMCID: PMC5841875 DOI: 10.1172/jci.insight.92973] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 08/31/2017] [Indexed: 12/24/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) is a global health threat, compounded by the emergence of drug-resistant strains. A hallmark of pulmonary tuberculosis (TB) is the formation of hypoxic necrotic granulomas, which upon disintegration, release infectious Mtb. Furthermore, hypoxic necrotic granulomas are associated with increased disease severity and provide a niche for drug-resistant Mtb. However, the host immune responses that promote the development of hypoxic TB granulomas are not well described. Using a necrotic Mtb mouse model, we show that loss of Mtb virulence factors, such as phenolic glycolipids, decreases the production of the proinflammatory cytokine IL-17 (also referred to as IL-17A). IL-17 production negatively regulates the development of hypoxic TB granulomas by limiting the expression of the transcription factor hypoxia-inducible factor 1α (HIF1α). In human TB patients, HIF1α mRNA expression is increased. Through genotyping and association analyses in human samples, we identified a link between the single nucleotide polymorphism rs2275913 in the IL-17 promoter (-197G/G), which is associated with decreased IL-17 production upon stimulation with Mtb cell wall. Together, our data highlight a potentially novel role for IL-17 in limiting the development of hypoxic necrotic granulomas and reducing disease severity in TB.
Collapse
Affiliation(s)
| | | | | | | | - Monika Bambouskova
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Radha Gopal
- Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | | | - Marcela Muñoz-Torrico
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | | | - Alfredo Cruz-Lagunas
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Luis Jiménez-Álvarez
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | | | - Ramón Espinosa-Soto
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Tamanna Sultana
- Bioinformatics Analysis Core, Genomics and Proteomics Core Laboratories, and
| | - James Lyons-Weiler
- Bioinformatics Analysis Core, Genomics and Proteomics Core Laboratories, and
| | - Todd A. Reinhart
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jesus Arcos
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | | | - Michael A. Mastrangelo
- Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Reid Townsend
- Proteomics Shared Resource, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Jordi B. Torrelles
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Gilla Kaplan
- Public Health Research Institute Center, New Jersey Medical School-Rutgers, State University of New Jersey, Newark, New Jersey, USA
| | - William Horne
- Richard King Mellon Institute for Pediatric Research, Department of Pediatrics and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jay K. Kolls
- Richard King Mellon Institute for Pediatric Research, Department of Pediatrics and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Maxim N. Artyomov
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Joaquín Zúñiga
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | | |
Collapse
|
100
|
Kaufmann SHE, Dockrell HM, Drager N, Ho MM, McShane H, Neyrolles O, Ottenhoff THM, Patel B, Roordink D, Spertini F, Stenger S, Thole J, Verreck FAW, Williams A. TBVAC2020: Advancing Tuberculosis Vaccines from Discovery to Clinical Development. Front Immunol 2017; 8:1203. [PMID: 29046674 PMCID: PMC5632681 DOI: 10.3389/fimmu.2017.01203] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/11/2017] [Indexed: 01/24/2023] Open
Abstract
TBVAC2020 is a research project supported by the Horizon 2020 program of the European Commission (EC). It aims at the discovery and development of novel tuberculosis (TB) vaccines from preclinical research projects to early clinical assessment. The project builds on previous collaborations from 1998 onwards funded through the EC framework programs FP5, FP6, and FP7. It has succeeded in attracting new partners from outstanding laboratories from all over the world, now totaling 40 institutions. Next to the development of novel vaccines, TB biomarker development is also considered an important asset to facilitate rational vaccine selection and development. In addition, TBVAC2020 offers portfolio management that provides selection criteria for entry, gating, and priority settings of novel vaccines at an early developmental stage. The TBVAC2020 consortium coordinated by TBVI facilitates collaboration and early data sharing between partners with the common aim of working toward the development of an effective TB vaccine. Close links with funders and other consortia with shared interests further contribute to this goal.
Collapse
Affiliation(s)
- Stefan H E Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hazel M Dockrell
- Immunology and Infection Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Nick Drager
- Tuberculosis Vaccine Initiative (TBVI), Lelystad, Netherlands
| | - Mei Mei Ho
- Bacteriology Division, MHRA-NIBSC, Potters Bar, United Kingdom
| | | | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Brij Patel
- RegExcel Consulting Ltd, Surrey, United Kingdom
| | | | | | | | - Jelle Thole
- Tuberculosis Vaccine Initiative (TBVI), Lelystad, Netherlands
| | | | | | | |
Collapse
|