1
|
Kim TS, Ikeuchi T, Theofilou VI, Williams DW, Greenwell-Wild T, June A, Adade EE, Li L, Abusleme L, Dutzan N, Yuan Y, Brenchley L, Bouladoux N, Sakamachi Y, Palmer RJ, Iglesias-Bartolome R, Trinchieri G, Garantziotis S, Belkaid Y, Valm AM, Diaz PI, Holland SM, Moutsopoulos NM. Epithelial-derived interleukin-23 promotes oral mucosal immunopathology. Immunity 2024; 57:859-875.e11. [PMID: 38513665 PMCID: PMC11058479 DOI: 10.1016/j.immuni.2024.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/05/2024] [Accepted: 02/29/2024] [Indexed: 03/23/2024]
Abstract
At mucosal surfaces, epithelial cells provide a structural barrier and an immune defense system. However, dysregulated epithelial responses can contribute to disease states. Here, we demonstrated that epithelial cell-intrinsic production of interleukin-23 (IL-23) triggers an inflammatory loop in the prevalent oral disease periodontitis. Epithelial IL-23 expression localized to areas proximal to the disease-associated microbiome and was evident in experimental models and patients with common and genetic forms of disease. Mechanistically, flagellated microbial species of the periodontitis microbiome triggered epithelial IL-23 induction in a TLR5 receptor-dependent manner. Therefore, unlike other Th17-driven diseases, non-hematopoietic-cell-derived IL-23 served as an initiator of pathogenic inflammation in periodontitis. Beyond periodontitis, analysis of publicly available datasets revealed the expression of epithelial IL-23 in settings of infection, malignancy, and autoimmunity, suggesting a broader role for epithelial-intrinsic IL-23 in human disease. Collectively, this work highlights an important role for the barrier epithelium in the induction of IL-23-mediated inflammation.
Collapse
Affiliation(s)
- Tae Sung Kim
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tomoko Ikeuchi
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vasileios Ionas Theofilou
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA; Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| | - Drake Winslow Williams
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Teresa Greenwell-Wild
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Armond June
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, University at Buffalo, Buffalo, NY 14214, USA
| | - Emmanuel E Adade
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12210, USA
| | - Lu Li
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, University at Buffalo, Buffalo, NY 14214, USA
| | - Loreto Abusleme
- Department of Pathology and Oral Medicine, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Nicolas Dutzan
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Yao Yuan
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laurie Brenchley
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicolas Bouladoux
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yosuke Sakamachi
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Robert J Palmer
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ramiro Iglesias-Bartolome
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Giorgio Trinchieri
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stavros Garantziotis
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alex M Valm
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12210, USA
| | - Patricia I Diaz
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, University at Buffalo, Buffalo, NY 14214, USA
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Niki M Moutsopoulos
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
2
|
McGeachy MJ. Gums make IL-23, no professionals needed. Immunity 2024; 57:832-834. [PMID: 38599173 DOI: 10.1016/j.immuni.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 04/12/2024]
Abstract
IL-23 activates pathogenic Th17 cells to drive inflammatory disease at barrier surfaces. Kim et al. now identify oral epithelial cells as the critical producers of IL-23 in human and mouse periodontitis, linking microbial dysbiosis to non-hematopoietic regulation of IL-17-associated inflammation.
Collapse
Affiliation(s)
- Mandy J McGeachy
- Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca NY 14853, USA.
| |
Collapse
|
3
|
Marzhoseyni Z, Mousavi MJ, Saffari M, Ghotloo S. Immune escape strategies of Pseudomonas aeruginosa to establish chronic infection. Cytokine 2023; 163:156135. [PMID: 36724716 DOI: 10.1016/j.cyto.2023.156135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/08/2023] [Accepted: 01/12/2023] [Indexed: 02/02/2023]
Abstract
The infection caused by P. aeruginosa still is dangerous throughout the world. This is partly due to its immune escape mechanisms considerably increasing the bacterial survival in the host. By escape from recognition by TLRs, interference with complement system activation, phagocytosis inhibition, production of ROS, inhibition of NET production, interference with the generation of cytokines, inflammasome inhibition, reduced antigen presentation, interference with cellular and humoral immunity, and induction of apoptotic cell death and MDSc, P. aeruginosa breaks down the barriers of the immune system and causes lethal infections in the host. Recognition of other immune escape mechanisms of P. aeruginosa may provide a basis for the future treatment of the infection. This manuscript may provide new insights and information for the development of new strategies to combat P. aeruginosa infection. In the present manuscript, the escape mechanisms of P. aeruginosa against immune response would be reviewed.
Collapse
Affiliation(s)
- Zeynab Marzhoseyni
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Javad Mousavi
- Department of Hematology, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mahmood Saffari
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Somayeh Ghotloo
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
4
|
Zhou Y, Xiang C, Wang N, Zhang X, Xie Y, Yang H, Guo G, Liu K, Li Y, Shi Y. Acinetobacter baumannii reinforces the pathogenesis by promoting IL-17 production in a mouse pneumonia model. Med Microbiol Immunol 2023; 212:65-73. [PMID: 36463365 DOI: 10.1007/s00430-022-00757-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022]
Abstract
Interleukin-17 (IL-17) is involved in host defense against bacterial infection. Little is known about the role of IL-17 in A. baumannii-infected pneumonia. Our objective was to investigate the role of IL-17 in pulmonary A. baumannii infection in a mouse model. We infected C57BL/6 mice intra-tracheally (i.t.) with A. baumannii to establish pneumonia model and found A. baumannii infection elevated IL-17 expression in lungs. IL-17-deficient (Il17-/-) mice were resistant to pulmonary A. baumannii infection, showing improved mice survival, reduced bacteria burdens, and alleviated lung inflammation. Further, treatment of A. baumannii-infected Il17-/- mice with IL-17 exacerbated the severity of pneumonia. These data suggest a pathogenic role of IL-17 in pulmonary A. baumannii infection. Further, the infiltration and phagocytic function of neutrophils in broncho-alveolar lavage fluid were detected by flow cytometry. The results showed that Il17-/- mice had increased neutrophil infiltration and enhanced phagocytosis in neutrophils at the early time of infection. Treatment of mice with IL-17 suppressed phagocytic function of neutrophils. All data suggest that IL-17 promotes susceptibility of mice to pulmonary A. baumannii infection by suppressing neutrophil phagocytosis at early time of infection. Targeting IL-17 might be a potential therapeutic strategy in controlling the outcome of A. baumannii pneumonia.
Collapse
Affiliation(s)
- Yangyang Zhou
- West China Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chuanying Xiang
- West China Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ning Wang
- West China Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiaomin Zhang
- West China Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yu Xie
- West China Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hong Yang
- West China Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Gang Guo
- West China Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Kaiyun Liu
- West China Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan Li
- West China Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yun Shi
- West China Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
5
|
Pseudomonas Aeruginosa Lung Infection Subverts Lymphocytic Responses through IL-23 and IL-22 Post-Transcriptional Regulation. Int J Mol Sci 2022; 23:ijms23158427. [PMID: 35955566 PMCID: PMC9369422 DOI: 10.3390/ijms23158427] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 02/01/2023] Open
Abstract
Pseudomonas aeruginosa (P.a) is a pathogen causing significant morbidity and mortality, particularly in hospital patients undergoing ventilation and in individuals with cystic fibrosis. Although we and others have investigated mechanisms used by P.a to subvert innate immunity, relatively less is known about the potential strategies used by this bacterium to fight the adaptive immune system and, in particular, T cells. Here, using RAG KO (devoid of ‘classical’ αβ and γδ TCR T lymphocytes) and double RAG γC KO mice (devoid of T, NK and ILC cells), we demonstrate that the lymphocytic compartment is important to combat P.a (PAO1 strain). Indeed, we show that PAO1 load was increased in double RAG γC KO mice. In addition, we show that PAO1 down-regulates IL-23 and IL-22 protein accumulation in the lungs of infected mice while up-regulating their RNA production, thereby pointing towards a specific post-transcriptional regulatory mechanism not affecting other inflammatory mediators. Finally, we demonstrate that an adenovirus-mediated over-expression of IL-1, IL-23 and IL-7 induced lung neutrophil and lymphocytic influx and rescued mice against P.a-induced lethality in all WT, RAG γC KO and RAG γC KO RAG-deficient mice, suggesting that this regimen might be of value in ‘locally immunosuppressed’ individuals such as cystic fibrosis patients.
Collapse
|
6
|
Albrecht M, Halle O, Gaedcke S, Pallenberg ST, Camargo Neumann J, Witt M, Roediger J, Schumacher M, Jirmo AC, Warnecke G, Jonigk D, Braubach P, DeLuca D, Hansen G, Dittrich AM. Interleukin-17A and interleukin-22 production by conventional and non-conventional lymphocytes in three different end-stage lung diseases. Clin Transl Immunology 2022; 11:e1398. [PMID: 35757569 PMCID: PMC9202301 DOI: 10.1002/cti2.1398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 05/11/2022] [Accepted: 06/01/2022] [Indexed: 11/10/2022] Open
Abstract
Objectives The contribution of adaptive vs. innate lymphocytes to IL-17A and IL-22 secretion at the end stage of chronic lung diseases remains largely unexplored. In order to uncover tissue- and disease-specific secretion patterns, we compared production patterns of IL-17A and IL-22 in three different human end-stage lung disease entities. Methods Production of IL-17A, IL-22 and associated cytokines was assessed in supernatants of re-stimulated lymphocytes by multiplex assays and multicolour flow cytometry of conventional T cells, iNKT cells, γδ T cells and innate lymphoid cells in bronchial lymph node and lung tissue from patients with emphysema (n = 19), idiopathic pulmonary fibrosis (n = 14) and cystic fibrosis (n = 23), as well as lung donors (n = 17). Results We detected secretion of IL-17A and IL-22 by CD4+ T cells, CD8+ T cells, innate lymphoid cells, γδ T cells and iNKT cells in all end-stage lung disease entities. Our analyses revealed disease-specific contributions of individual lymphocyte subpopulations to cytokine secretion patterns. We furthermore found the high levels of microbial detection in CF samples to associate with a more pronounced IL-17A signature upon antigen-specific and unspecific re-stimulation compared to other disease entities and lung donors. Conclusion Our results show that both adaptive and innate lymphocyte populations contribute to IL-17A-dependent pathologies in different end-stage lung disease entities, where they establish an IL-17A-rich microenvironment. Microbial colonisation patterns and cytokine secretion upon microbial re-stimulation suggest that pathogens drive IL-17A secretion patterns in end-stage lung disease.
Collapse
Affiliation(s)
- Melanie Albrecht
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany.,Molecular Allergology Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines Langen Germany.,Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| | - Olga Halle
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany.,Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| | - Svenja Gaedcke
- Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| | - Sophia T Pallenberg
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany
| | - Julia Camargo Neumann
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany
| | - Marius Witt
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany
| | - Johanna Roediger
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany
| | - Marina Schumacher
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany
| | - Adan Chari Jirmo
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany.,Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| | - Gregor Warnecke
- Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany.,Department of Cardiac Surgery Heidelberg Medical School Heidelberg Germany
| | - Danny Jonigk
- Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany.,Institute of Pathology Hannover Medical School Hannover Germany
| | - Peter Braubach
- Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany.,Institute of Pathology Hannover Medical School Hannover Germany
| | - David DeLuca
- Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| | - Gesine Hansen
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany.,Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| | - Anna-Maria Dittrich
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany.,Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| |
Collapse
|
7
|
Allergic airway inflammation induces upregulation of the expression of IL-23R by macrophages and not in CD3 + T cells and CD11c +F4/80 - dendritic cells of the lung. Cell Tissue Res 2022; 389:85-98. [PMID: 35475923 PMCID: PMC9200692 DOI: 10.1007/s00441-021-03538-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 10/06/2021] [Indexed: 11/05/2022]
Abstract
Interleukin 23 and the interleukin 23 receptor (IL-23-IL23R) are described as the major enhancing factors for Interleukin 17 (IL-17) in allergic airway inflammation. IL-17 is considered to induce neutrophilic inflammation in the lung, which is often observed in severe, steroid-resistant asthma-phenotypes. For that reason, understanding of IL-23 and IL-17 axis is very important for future therapy strategies, targeting neutrophil pathway of bronchial asthma. This study aimed to investigate the distribution and expression of IL-23R under physiological and inflammatory conditions. Therefore, a house dust mite (HDM) model of allergic airway inflammation was performed by treating mice with HDM intranasally. Immunofluorescence staining with panel of antibodies was performed in lung tissues to examine the macrophage, dendritic cell, and T cell subpopulations. The allergic airway inflammation was quantified by histopathological analysis, ELISA measurements, and airway function. HDM-treated mice exhibited a significant allergic airway inflammation including higher amounts of NE+ cells in lung parenchyma. We found only a small amount of IL-23R positives, out of total CD3+T cells, and no upregulation in HDM-treated animals. In contrast, the populations of F4/80+ macrophages and CD11c+F4/80− dendritic cells (DCs) with IL-23R expression were found to be higher. But HDM treatment leads to a significant increase of IL-23R+ macrophages, only. IL-23R was expressed by every examined macrophage subpopulation, whereas only Mϕ1 and hybrids between Mϕ1 and Mϕ2 phenotype and not Mϕ2 were found to upregulate IL-23R. Co-localization of IL-23R and IL-17 was only observed in F4/80+ macrophages, suggesting F4/80+ macrophages express IL-23R along with IL-17 in lung tissue. The study revealed that macrophages involving the IL-23 and IL-17 pathway may provide a potential interesting therapeutic target in neutrophilic bronchial asthma.
Collapse
|
8
|
Anti-virulence Bispecific Monoclonal Antibody Mediated Protection Against Pseudomonas aeruginosa Ventilator-Associated Pneumonia in a Rabbit Model. Antimicrob Agents Chemother 2021; 66:e0202221. [PMID: 34902264 PMCID: PMC8846318 DOI: 10.1128/aac.02022-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Ventilator-associated pneumonia is an important clinical manifestation of the nosocomial pathogen Pseudomonas aeruginosa. We characterized the correlates of protection of MEDI3902, a bispecific human IgG1 mAb that targets the P. aeruginosa type-3-secretion PcrV protein and the Psl exopolysaccharide, in a rabbit model of ventilator-associated pneumonia using lung-protective, low-tidal volume mechanical ventilation. Rabbits infused with MEDI3902 prophylactically were protected, whereas those pretreated with irrelevant isotype-control IgG (c-IgG) succumbed between 12 and 44 hours post infection [100% (8/8) vs. 0% (8/8) survival, P<0.01 by log-rank test]. Lungs from rabbits pretreated with c-IgG, but not those with MEDI3902, had bilateral, multifocal areas of marked necrosis, hemorrhage, neutrophilic inflammatory infiltrate, diffuse fibrinous edema in alveolar spaces. All rabbits pretreated with c-IgG developed worsening bacteremia that peaked at the time of death, whereas only 38% (3/8) rabbits pretreated with MEDI3902 developed such high-grade bacteremia (two-sided Fisher's exact test, P=0.026). Biomarkers associated with acute respiratory distress syndrome were evaluated longitudinally in blood samples collected every 2-4 hours to assess systemic pathophysiological changes in rabbits pretreated with MEDI3902 or c-IgG. Biomarkers were sharply increased or decreased in rabbits pretreated with c-IgG, but not those pretreated with MEDI3902, including ratio of arterial oxygen partial pressure to fractional inspired oxygen PaO2/FiO2 <300, hypercapnia or hypocapnia, severe lactic acidosis, leukopenia and neutropenia. Cytokines and chemokines associated with ARDS were significantly downregulated in lungs from rabbits pretreated with MEDI3902 compared with c-IgG. These results suggest that MEDI3902 prophylaxis could have potential clinical utility for decreasing severity of P. aeruginosa ventilator-associated pneumonia.
Collapse
|
9
|
Sun J, LaRock DL, Skowronski EA, Kimmey JM, Olson J, Jiang Z, O'Donoghue AJ, Nizet V, LaRock CN. The Pseudomonas aeruginosa protease LasB directly activates IL-1β. EBioMedicine 2020; 60:102984. [PMID: 32979835 PMCID: PMC7511813 DOI: 10.1016/j.ebiom.2020.102984] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pulmonary damage by Pseudomonas aeruginosa during cystic fibrosis lung infection and ventilator-associated pneumonia is mediated both by pathogen virulence factors and host inflammation. Impaired immune function due to tissue damage and inflammation, coupled with pathogen multidrug resistance, complicates the management of these deep-seated infections. Pathological inflammation during infection is driven by interleukin-1β (IL-1β), but the molecular processes involved are not fully understood. METHODS We examined IL-1β activation in a pulmonary model infection of Pseudomonas aeruginosa and in vitro using genetics, specific inhibitors, recombinant proteins, and targeted reporters of protease activity and IL-1β bioactivity. FINDINGS Caspase-family inflammasome proteases canonically regulate maturation of this proinflammatory cytokine, but we report that plasticity in IL-1β proteolytic activation allows for its direct maturation by the pseudomonal protease LasB. LasB promotes IL-1β activation, neutrophilic inflammation, and destruction of lung architecture characteristic of severe P. aeruginosa pulmonary infection. INTERPRETATION Preservation of lung function and effective immune clearance may be enhanced by selectively controlling inflammation. Discovery of this IL-1β regulatory mechanism provides a distinct target for anti-inflammatory therapeutics, such as matrix metalloprotease inhibitors that inhibit LasB and limit inflammation and pathology during P. aeruginosa pulmonary infections. FUNDING Full details are provided in the Acknowledgements section.
Collapse
Affiliation(s)
- Josh Sun
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA, United States
| | - Doris L LaRock
- Department of Microbiology and Immunology, Emory School of Medicine, Atlanta GA, United States
| | - Elaine A Skowronski
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA, United States
| | | | - Joshua Olson
- Department of Pediatrics, UC San Diego, La Jolla, CA, United States
| | - Zhenze Jiang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA, United States
| | - Anthony J O'Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA, United States
| | - Victor Nizet
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA, United States; Department of Pediatrics, UC San Diego, La Jolla, CA, United States
| | - Christopher N LaRock
- Department of Microbiology and Immunology, Emory School of Medicine, Atlanta GA, United States; Division of Infectious Diseases, Emory School of Medicine, Atlanta GA, United States; Antimicrobial Resistance Center, Emory University, Atlanta GA, United States.
| |
Collapse
|
10
|
Caffarelli C, Santamaria F, Mirra V, Bacchini E, Santoro A, Bernasconi S, Corsello G. Advances in paediatrics in 2019: current practices and challenges in allergy, endocrinology, gastroenterology, public health, neonatology, nutrition, nephrology, neurology, respiratory diseases and rheumatic diseases. Ital J Pediatr 2020; 46:89. [PMID: 32600434 PMCID: PMC7325159 DOI: 10.1186/s13052-020-00853-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 06/18/2020] [Indexed: 11/10/2022] Open
Abstract
We highlight the main developments that have been published during the first semester of the last year in the Italian Journal of Pediatrics. We have carefully chosen information from numerous exciting progresses issued in the Journal in the field of allergy, endocrinology, gastroenterology, neonatology, nutrition, nephrology, neurology, public health, respiratory diseases and rheumatic diseases. The impact on the care of patients has been placed in the broader context of studies that appeared in other journals. We think that many observations can be used directly to upgrade management of patients.
Collapse
Affiliation(s)
- Carlo Caffarelli
- Clinica Pediatrica, Department of Medicine and Surgery, Azienda Ospedaliera-Universitaria, University of Parma, Parma, Italy
| | - Francesca Santamaria
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Virginia Mirra
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Ermanno Bacchini
- Unità Polispecialistica Pediatrica Centro Medi Saluser, Parma, Italy
| | - Angelica Santoro
- Clinica Pediatrica, Department of Medicine and Surgery, Azienda Ospedaliera-Universitaria, University of Parma, Parma, Italy
| | | | - Giovanni Corsello
- Department of Sciences for Health Promotion and Mother and Child Care “G. D’Alessandro”, University of Palermo, Palermo, Italy
| |
Collapse
|
11
|
Alam MS, Cavanaugh C, Pereira M, Babu U, Williams K. Susceptibility of aging mice to listeriosis: Role of anti-inflammatory responses with enhanced Treg-cell expression of CD39/CD73 and Th-17 cells. Int J Med Microbiol 2020; 310:151397. [PMID: 31974050 DOI: 10.1016/j.ijmm.2020.151397] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 11/14/2019] [Accepted: 12/18/2019] [Indexed: 12/22/2022] Open
Abstract
Foodborne Listeria monocytogenes (Lm) causes serious illness and death in immunosuppressed hosts, including the elderly population. We investigated Lm susceptibility and inflammatory cytokines in geriatric mice. Young-adult and old mice were gavaged with a Lm strain Lmo-InlAm. Tissues were assayed for Lm burden and splenocytes were analyzed for Th1/Th2/Th17/Treg responses and expression of CD39 and CD73. Old Lm-infected mice lost body-weight dose-dependently, had higher Lm colonization, and showed higher inflammatory responses than Lm-infected young-adult mice. After infection, IL-17 levels increased significantly in old mice whereas IFN-γ levels were unchanged. Levels of IL-10 and Treg cells were increased in infected old mice as compared to infected young-adult mice. Age-dependent enhanced expression of CD39/CD73 was observed in purified Treg prior to infection, suggesting increased baseline adenosine production in old mice. Lm lysate-treated splenocytes from older mice produced significantly higher levels of IL-10, IL17, and IL-1β, produced less IFN-γ and IL-2, and proliferated less than splenocytes from young-adult mice. Data suggests that older mice maybe more susceptible to Lm infection due to an imbalance of Th cell responses with disproportionate and persistent anti-inflammatory responses. Lm infection enhanced differentiation of proinflammatory Th17 cells, which may also exacerbate pathological responses during listeriosis.
Collapse
Affiliation(s)
- M Samiul Alam
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA.
| | - Christopher Cavanaugh
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA
| | - Marion Pereira
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA
| | - Uma Babu
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA
| | - Kristina Williams
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA
| |
Collapse
|
12
|
Olszowiec-Chlebna M, Koniarek-Maniecka A, Brzozowska A, Błauż A, Rychlik B, Stelmach I. Vitamin D inhibits pro-inflammatory cytokines in the airways of cystic fibrosis patients infected by Pseudomonas aeruginosa- pilot study. Ital J Pediatr 2019; 45:41. [PMID: 30922377 PMCID: PMC6440129 DOI: 10.1186/s13052-019-0634-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/15/2019] [Indexed: 12/19/2022] Open
Abstract
Background Vitamin D plays an important role in inflammatory responses after antigen exposure. Interleukin-23 (Il-23) promotes Il-17-dependent inflammation during Pseudomonas aeruginosa (P. aeruginosa) pulmonary infection. We aimed to compare the ability of calcitriol and cholecalciferol to modulate the inflammatory response of the CF airways infected with P. aeruginosa. Methods This was a randomized, placebo-controlled, double-blind, cross-over trial. Twenty-three patients with CF (aged 6–19), chronically infected by P. aeruginosa were randomly assigned to: calcitriol group receiving 1,25(OH)2D 0,5 mcg daily or cholecalciferol group receiving cholecalciferol 1000 IU daily for three months. The levels of Il-23 and Il-17A in the exhaled breath concentrate (EBC) were measured. Calcium-phosphorus balance was also evaluated (serum concentration of calcium, phosphorus, 25OHD, parathormone (PTH) and calcium/creatinine ratio in urine). Data were analyzed using means of Stata/Special Edition, release 14.2. A level of P < 0.05 was considered statistically significant. Results The level of Il-17A in EBC significantly decreased in calcitriol group from 0,475 pg/mL (± SD 0,515 pg/mL) to 0,384 pg/mL (± SD 0,429 pg/mL) (p = 0,008); there was no change in cholecalciferol group (p = 0,074). The level of Il-23 in EBC did not significantly change in calcitriol group (p = 0,086); there was significant decrease in cholecalciferol group from 8,90 pg/mL (± SD 4,07 pg/mL) to 7,33 pg/mL (± SD 3,88 pg/mL) (p = 0,001). In calcitriol group serum phosphorus and PTH significantly decreased (p = 0,021 and p = 0,019 respectively), the concentration of calcium significantly increased (p = 0,001); there were no changes in cholecalciferol group. Conclusions Both analogs of vitamin D revealed their anti-inflammatory effect and reduced the level of Il-17A and Il-23 in the airway of CF patients with chronic P. aeruginosa infection. We observed improvement in calcium-phosphorus metabolism after supplementation with calcitriol, without adverse effects. It is recommended to use vitamin D in CF patients.
Collapse
Affiliation(s)
- M Olszowiec-Chlebna
- Department of Pediatrics and Allergy, Medical University of Lodz, Copernicus Memorial Hospital, Korczak Paediatric Center, Piłsudskiego 71 Str, 90-329, Lodz, Poland
| | - A Koniarek-Maniecka
- Department of Pediatrics and Allergy, Medical University of Lodz, Copernicus Memorial Hospital, Korczak Paediatric Center, Piłsudskiego 71 Str, 90-329, Lodz, Poland
| | - A Brzozowska
- Department of Pediatrics and Allergy, Medical University of Lodz, Copernicus Memorial Hospital, Korczak Paediatric Center, Piłsudskiego 71 Str, 90-329, Lodz, Poland
| | - A Błauż
- Laboratory of Cytometry, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - B Rychlik
- Laboratory of Cytometry, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - I Stelmach
- Department of Pediatrics and Allergy, Medical University of Lodz, Copernicus Memorial Hospital, Korczak Paediatric Center, Piłsudskiego 71 Str, 90-329, Lodz, Poland.
| |
Collapse
|
13
|
Epigenetic Regulation of IL-17-Induced Chemokines in Lung Epithelial Cells. Mediators Inflamm 2019; 2019:9050965. [PMID: 31080358 PMCID: PMC6441531 DOI: 10.1155/2019/9050965] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 12/18/2018] [Indexed: 01/25/2023] Open
Abstract
Epithelial cells are known to have barrier functions in multiple organs and regulate innate immune responses. Airway epithelial cells respond to IL-17 by altering their transcriptional profiles and producing antimicrobial proteins and neutrophil chemoattractants. Although IL-17 has been shown to promote inflammation through stabilizing mRNA of CXCR2 ligands, how IL-17 exerts its downstream effects on its target cells through epigenetic mechanisms is largely unknown. Using primary human bronchial epithelial cells and immortalized epithelial cell line from both human and mouse, we demonstrated that IL-17-induced CXCR2 ligand production is dependent on histone acetylation specifically through repressing HDAC5. Furthermore, the chemokine production induced by IL-17 is strictly dependent on the bromodomain and extraterminal domain (BET) family as BET inhibition abolished the IL-17A-induced proinflammatory chemokine production, indicating a pivotal role of the recognition of acetylated histones. In combination with single-cell RNA-seq analysis, we revealed that the cell lines we employed represent specific lineages and their IL-17 responses were regulated differently by the DNA methylation mechanisms. Taken together, our data strongly support that IL-17 sustains epithelial CXCR2 ligand production through epigenetic regulation and the therapeutic potential of interrupting histone modification as well as the recognition of modified histones could be evaluated in neutrophilic lung diseases.
Collapse
|
14
|
Iwanaga N, Kolls JK. Updates on T helper type 17 immunity in respiratory disease. Immunology 2018; 156:3-8. [PMID: 30260473 DOI: 10.1111/imm.13006] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/12/2022] Open
Abstract
Interleukin-17 (IL-17)-producing cells play a critical role in mucosal immunity including the respiratory tract. This review will highlight recent advances in our understanding of these cells in mucosal immunity in the lung as well as their potential pathogenic roles in respiratory diseases. The IL-17-producing cells include γδ T cells, natural killer cells, group 3 innate lymphoid cells, and T helper type 17 (Th17) cells. There have been recent advances in our understanding of these cell populations in the lung as well as emerging data on how these cells are regulated in the lung. Moreover, Th17 cells may be a key component of tissue-resident memory cells that may be acquired over time or elicited by mucosal immunization that provides the host with enhanced immunity against certain pathogens.
Collapse
Affiliation(s)
- Naoki Iwanaga
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
15
|
MEDI3902 Correlates of Protection against Severe Pseudomonas aeruginosa Pneumonia in a Rabbit Acute Pneumonia Model. Antimicrob Agents Chemother 2018; 62:AAC.02565-17. [PMID: 29483116 PMCID: PMC5923159 DOI: 10.1128/aac.02565-17] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/20/2018] [Indexed: 12/24/2022] Open
Abstract
Pseudomonas aeruginosa is among the most formidable antibiotic-resistant pathogens and is a leading cause of hospital-associated infections. With dwindling options for antibiotic-resistant infections, a new paradigm for treatment and disease resolution is required. MEDI3902, a bispecific antibody targeting the P. aeruginosa type III secretion (T3S) protein PcrV and Psl exopolysaccharide, was previously shown to mediate potent protective activity in murine infection models. With the current challenges associated with the clinical development of narrow-spectrum agents, robust preclinical efficacy data in multiple animal species are desirable. Here, we sought to develop a rabbit P. aeruginosa acute pneumonia model to further evaluate the activity of MEDI3902 intervention. In the rabbit model of acute pneumonia, prophylaxis with MEDI3902 exhibited potent dose-dependent protection, whereas those receiving control IgG developed fatal hemorrhagic necrotizing pneumonia between 12 and 54 h after infection. Blood biomarkers (e.g., partial pressure of oxygen [pO2], partial pressure of carbon dioxide [pCO2], base excess, lactate, and creatinine) were grossly deranged for the vast majority of control IgG-treated animals but remained within normal limits for MEDI3902-treated animals. In addition, MEDI3902-treated animals exhibited a profound reduction in P. aeruginosa organ burden and a marked reduction in the expression of proinflammatory mediators from lung tissue, which correlated with reduced lung histopathology. These results confirm that targeting PcrV and Psl via MEDI3902 is a promising candidate for immunotherapy against P. aeruginosa pneumonia.
Collapse
|
16
|
Monin L, Gaffen SL. Interleukin 17 Family Cytokines: Signaling Mechanisms, Biological Activities, and Therapeutic Implications. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028522. [PMID: 28620097 DOI: 10.1101/cshperspect.a028522] [Citation(s) in RCA: 224] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The cytokines of the interleukin 17 (IL-17) family play a central role in the control of infections, especially extracellular fungi. Conversely, if unrestrained, these inflammatory cytokines contribute to the pathology of numerous autoimmune and chronic inflammatory conditions. Recent advances have led to the approval of IL-17A-blocking biologics for the treatment of moderate to severe plaque psoriasis, but much remains to be understood about the biological functions, regulation, and signaling pathways downstream of these factors. In this review, we outline the current knowledge of signal transduction and known physiological activities of IL-17 family cytokines. We will highlight in particular the current understanding of these cytokines in the context of skin manifestations of disease.
Collapse
Affiliation(s)
- Leticia Monin
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Sarah L Gaffen
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
17
|
Fernandez CP, Afrin F, Flores RA, Kim WH, Jeong J, Kim S, Lillehoj HS, Min W. Identification of duck IL-4 and its inhibitory effect on IL-17A expression in R. anatipestifer- stimulated splenic lymphocytes. Mol Immunol 2018; 95:20-29. [DOI: 10.1016/j.molimm.2018.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 12/29/2017] [Accepted: 01/19/2018] [Indexed: 12/13/2022]
|
18
|
Garcia M, Morello E, Garnier J, Barrault C, Garnier M, Burucoa C, Lecron JC, Si-Tahar M, Bernard FX, Bodet C. Pseudomonas aeruginosa flagellum is critical for invasion, cutaneous persistence and induction of inflammatory response of skin epidermis. Virulence 2018; 9:1163-1175. [PMID: 30070169 PMCID: PMC6086312 DOI: 10.1080/21505594.2018.1480830] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 05/21/2018] [Indexed: 12/22/2022] Open
Abstract
Pseudomonas aeruginosa, an opportunistic pathogen involved in skin and lung diseases, possesses numerous virulence factors, including type 2 and 3 secretion systems (T2SS and T3SS) and its flagellum, whose functions remain poorly known during cutaneous infection. Using isogenic mutants deleted from genes encoding each or all of these three virulence factors, we investigated their role in induction of inflammatory response and in tissue invasiveness in human primary keratinocytes and reconstructed epidermis. Our results showed that flagellum, but not T2SS and T3SS, is involved in induction of a large panel of cytokine, chemokine, and antimicrobial peptide (AMP) mRNA in the infected keratinocytes. Chemokine secretion and AMP tissular production were also dependent on the presence of the bacterial flagellum. This pro-inflammatory effect was significantly reduced in keratinocytes infected in presence of anti-toll-like receptor 5 (TLR5) neutralizing antibody. Bacterial invasion of human epidermis and persistence in a mouse model of sub-cutaneous infection were dependent on the P. aeruginosa flagellum. We demonstrated that flagellum constitutes the main virulence factor of P. aeruginosa involved not only in early induction of the epidermis inflammatory response but also in bacterial invasion and cutaneous persistence. P. aeruginosa is mainly sensed by TLR5 during the early innate immune response of human primary keratinocytes.
Collapse
Affiliation(s)
- Magali Garcia
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
-
Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers
, Poitiers, France
| | - Eric Morello
-
Centre d'Etude des Pathologies Respiratoires, INSERM UMR 1100, Université de Tours
, Tours, France
| | | | | | - Martine Garnier
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
| | - Christophe Burucoa
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
-
Laboratoire de Bactériologie et Hygiène, CHU de Poitiers
, Poitiers, France
| | - Jean-Claude Lecron
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
-
Laboratoire d’Immunologie et Inflammation, CHU de Poitiers
, Poitiers, France
| | - Mustapha Si-Tahar
-
Centre d'Etude des Pathologies Respiratoires, INSERM UMR 1100, Université de Tours
, Tours, France
| | | | - Charles Bodet
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
| |
Collapse
|
19
|
Ray A, Kolls JK. Neutrophilic Inflammation in Asthma and Association with Disease Severity. Trends Immunol 2017; 38:942-954. [PMID: 28784414 PMCID: PMC5711587 DOI: 10.1016/j.it.2017.07.003] [Citation(s) in RCA: 284] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 06/28/2017] [Accepted: 07/11/2017] [Indexed: 01/22/2023]
Abstract
Asthma is a chronic inflammatory disorder of the airways. While the local infiltration of eosinophils and mast cells, and their role in the disease have long been recognized, neutrophil infiltration has also been assessed in many clinical studies. In these studies, airway neutrophilia was associated with asthma severity. Importantly, neutrophilia also correlates with asthma that is refractory to corticosteroids, the mainstay of asthma treatment. However, it is now increasingly recognized that neutrophils are a heterogeneous population, and a more precise phenotyping of these cells may help delineate different subtypes of asthma. Here, we review current knowledge of the role of neutrophils in asthma and highlight future avenues of research in this field.
Collapse
Affiliation(s)
- Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; University of Pittsburgh Asthma Institute@UPMC/UPSOM, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Jay K Kolls
- Richard King Mellon Institute for Pediatric Research, Children's Hospital of Pittsburgh at University of Pittsburgh Medical Center/University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
20
|
Sheikh Z, Ong HX, Pozzoli M, Young PM, Traini D. Is there a role for inhaled anti-inflammatory drugs in cystic fibrosis treatment? Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2018.1409110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Zara Sheikh
- Respiratory Technology, The Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Hui Xin Ong
- Respiratory Technology, The Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Michele Pozzoli
- Respiratory Technology, The Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Paul M Young
- Respiratory Technology, The Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Daniela Traini
- Respiratory Technology, The Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
21
|
McDermott AJ, Falkowski NR, McDonald RA, Frank CR, Pandit CR, Young VB, Huffnagle GB. Role of interferon-γ and inflammatory monocytes in driving colonic inflammation during acute Clostridium difficile infection in mice. Immunology 2017; 150:468-477. [PMID: 27995603 DOI: 10.1111/imm.12700] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/30/2016] [Accepted: 12/05/2016] [Indexed: 02/06/2023] Open
Abstract
The inflammatory response to the colonic pathogen Clostridium difficile is characterized by the induction of inflammatory cytokines including Interleukin-23 (IL-23) and interferon-γ (IFN-γ) and the recruitment of myeloid cells including Ly6CHigh monocytes. IL-23 knockout mice showed reduced expression of the monocyte chemokines Ccl4 and Ccl7, but not Ccl2, as well as reduced Ly6CHigh Ly6GMid monocyte recruitment to the colon in response to C. difficile colitis. Clostridium difficile-infected CCR2-/- (CCR2 KO) mice showed a significant defect in Ly6CHigh Ly6GMid monocyte recruitment to the colon in response to C. difficile. Although there was no decrease in expression of the inflammatory cytokines Il1b, Il6 or Tnf or reduction in the severity of colonic histopathology associated with ablation of monocyte recruitment, Slpi and Inos expression was significantly reduced in the colons of these animals. Additionally, neutralization of IFN-γ through the administration of anti-IFN-γ monoclonal antibody resulted in a significant reduction in the expression of the IFN-γ-inducible chemokines Cxcl9 and Cxcl10, but not a reduction in the neutrophil chemokines Cxcl1, Cxcl2 and Ccl3 or the monocyte chemokine Ccl2. Consistently, monocyte and neutrophil recruitment were unchanged following anti-IFN-γ treatment. Additionally, Inos and Slpi expression were unchanged following anti-IFN-γ treatment, suggesting that Inos and Slpi regulation is independent of IFN-γ during C. difficile colitis. Taken together, these data strongly suggest that IL-23 and CCR2 signalling are required for monocyte recruitment during C. difficile colitis. Additionally, these studies also suggest that monocytes, but not IFN-γ, are necessary for full expression of Inos and Slpi in the colon.
Collapse
Affiliation(s)
- Andrew J McDermott
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Nicole R Falkowski
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Roderick A McDonald
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Charles R Frank
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Chinmay R Pandit
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Vincent B Young
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.,Division of Infectious Diseases, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gary B Huffnagle
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
22
|
The host genetic background defines diverse immune-reactivity and susceptibility to chronic Pseudomonas aeruginosa respiratory infection. Sci Rep 2016; 6:36924. [PMID: 27848994 PMCID: PMC5111113 DOI: 10.1038/srep36924] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 10/21/2016] [Indexed: 01/31/2023] Open
Abstract
Patients with P. aeruginosa airways infection show markedly variable clinical phenotypes likely influenced by genetic backgrounds. Here, we investigated the cellular events involved in resistance and susceptibility to P. aeruginosa chronic infection using genetically distinct inbred mouse strains. As for patients, different murine genotypes revealed variable susceptibility to infection. When directly compared, resistant C3H/HeOuJ and susceptible A/J strains revealed distinct immune responsiveness to the pathogen. In C3H/HeOuJ resistant mice, IL17-producing cells rapidly and transiently infiltrated the infected lung, and this was paralleled by the acute accumulation of alveolar macrophages, bacterial clearance and resolution of infection. In contrast, A/J susceptible mice revealed a more delayed and prolonged lung infiltration by IL17+ and IFNγ+ cells, persistence of innate inflammatory cells and establishment of chronic infection. We conclude that the host genetic background confers diverse immunoreactivity to P. aeruginosa and IL17-producing cells might contribute to the progress of chronic lung infection.
Collapse
|
23
|
Chen K, Eddens T, Trevejo-Nunez G, Way EE, Elsegeiny W, Ricks DM, Garg AV, Erb CJ, Bo M, Wang T, Chen W, Lee JS, Gaffen SL, Kolls JK. IL-17 Receptor Signaling in the Lung Epithelium Is Required for Mucosal Chemokine Gradients and Pulmonary Host Defense against K. pneumoniae. Cell Host Microbe 2016; 20:596-605. [PMID: 27923703 DOI: 10.1016/j.chom.2016.10.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 08/17/2016] [Accepted: 09/21/2016] [Indexed: 12/16/2022]
Abstract
The cytokine IL-17, and signaling via its heterodimeric IL-17RA/IL-17RC receptor, is critical for host defense against extracellular bacterial and fungal pathogens. Polarized lung epithelial cells express IL-17RA and IL-17RC basolaterally. However, their contribution to IL-17-dependent pulmonary defenses in vivo remains to be determined. To address this, we generated mice with conditional deletion of Il17ra or Il17rc in Scgb1a1-expressing club cells, a major component of the murine bronchiolar epithelium. These mice displayed an impaired ability to recruit neutrophils into the airway lumen in response to IL-17, a defect in bacterial clearance upon mucosal challenge with the pulmonary pathogen Klebsiella pneumoniae, and substantially reduced epithelial expression of the chemokine Cxcl5. Neutrophil recruitment and bacterial clearance were restored by intranasal administration of recombinant CXCL5. Our data show that IL-17R signaling in the lung epithelium plays a critical role in establishing chemokine gradients that are essential for mucosal immunity against pulmonary bacterial pathogens.
Collapse
Affiliation(s)
- Kong Chen
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Taylor Eddens
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Giraldina Trevejo-Nunez
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Emily E Way
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Waleed Elsegeiny
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - David M Ricks
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Abhishek V Garg
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Carla J Erb
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Meihua Bo
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Ting Wang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Wei Chen
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Janet S Lee
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sarah L Gaffen
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jay K Kolls
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA.
| |
Collapse
|
24
|
Wolf L, Sapich S, Honecker A, Jungnickel C, Seiler F, Bischoff M, Wonnenberg B, Herr C, Schneider-Daum N, Lehr CM, Bals R, Beisswenger C. IL-17A-mediated expression of epithelial IL-17C promotes inflammation during acute Pseudomonas aeruginosa pneumonia. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1015-L1022. [PMID: 27694471 DOI: 10.1152/ajplung.00158.2016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 09/26/2016] [Indexed: 12/31/2022] Open
Abstract
Lung epithelial cells are suggested to promote pathogen-induced pulmonary inflammation by the release of chemokines, resulting in enhanced recruitment of circulating leukocytes. Recent studies have shown that the interleukin-17C (IL-17C) regulates innate immune functions of epithelial cells in an autocrine manner. The aim of this study was to investigate the contribution of IL-17C to pulmonary inflammation in a mouse model of acute Pseudomonas aeruginosa pneumonia. Infection with P. aeruginosa resulted in an increased expression of IL-17C in lung tissue of wild-type mice. Numbers of neutrophils and the expression of the neutrophil-recruiting chemokines keratinocyte-derived chemokine and macrophage inflammatory protein 2 were significantly decreased in lungs of IL-17C-deficient (IL-17C-/-) mice infected with P. aeruginosa at 24 h. Systemic concentrations of interleukin-6 (IL-6) were significantly decreased in infected IL-17C-/- mice at 24 h and the survival of IL-17C-/- mice was significantly increased at 48 h. The expression of IL-17C was reduced in infected mice deficient for interleukin-17A (IL-17A), whereas pulmonary concentrations of IL-17A were not affected by the deficiency for IL-17C. Stimulation of primary alveolar epithelial cells with IL-17A resulted in a significantly increased expression of IL-17C in vitro. Our data suggest that IL-17A-mediated expression of epithelial IL-17C amplifies the release of chemokines by epithelial cells and thereby contributes to the recruitment of neutrophils and systemic inflammation during acute P. aeruginosa pneumonia.
Collapse
Affiliation(s)
- Lisa Wolf
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Sandra Sapich
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany; and
| | - Anja Honecker
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Christopher Jungnickel
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Frederik Seiler
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Markus Bischoff
- Institute of Medical Microbiology and Hygiene, Saarland University, Homburg, Germany
| | - Bodo Wonnenberg
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Christian Herr
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Nicole Schneider-Daum
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany; and
| | - Claus-Michael Lehr
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany; and.,Biopharmaceutics and Pharmaceutical Technology, Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Robert Bals
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany;
| |
Collapse
|
25
|
Wonnenberg B, Jungnickel C, Honecker A, Wolf L, Voss M, Bischoff M, Tschernig T, Herr C, Bals R, Beisswenger C. IL-17A attracts inflammatory cells in murine lung infection with P. aeruginosa. Innate Immun 2016; 22:620-625. [DOI: 10.1177/1753425916668244] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
IL-17A-dependent immunity is of importance in the protection against extracellular bacterial pathogens. However, IL-17A is also suggested to mediate the pathogenesis of lung diseases, such as acute respiratory distress syndrome. Here, we studied the role of IL-17A in a mouse model of acute pneumonia. IL-17A mediated the expression of keratinocyte-derived chemokine (KC) and the recruitment of inflammatory cells in mice infected with a sub-lethal dose of Pseudomonas aeruginosa. IL-17A deficiency protected mice from lethal P. aeruginosa lung infection. A sub-lethal infection with Streptococcus pneumoniae resulted in increased bacterial burden associated with increased pulmonary inflammation. Thus, the type of infectious bacteria seemed to influence the way in which IL-17A functions during pulmonary infection. Reducing pulmonary inflammation by targeting IL-17A may be a therapeutic option in acute P. aeruginosa pneumonia.
Collapse
Affiliation(s)
| | - Christopher Jungnickel
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Germany
| | - Anja Honecker
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Germany
| | - Lisa Wolf
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Germany
| | - Meike Voss
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Germany
| | - Markus Bischoff
- Institute of Medical Microbiology and Hygiene Saarland University, Germany
| | | | - Christian Herr
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Germany
| | - Robert Bals
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Germany
| |
Collapse
|
26
|
Way EE, Trevejo-Nunez G, Kane LP, Steiner BH, Puri KD, Kolls JK, Chen K. Dose-Dependent Suppression of Cytokine production from T cells by a Novel Phosphoinositide 3-Kinase Delta Inhibitor. Sci Rep 2016; 6:30384. [PMID: 27461849 PMCID: PMC4961957 DOI: 10.1038/srep30384] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/04/2016] [Indexed: 12/20/2022] Open
Abstract
There remains a significant need for development of effective small molecules that can inhibit cytokine-mediated inflammation. Phosphoinositide 3 kinase (PI3K) is a direct upstream activator of AKT, and plays a critical role in multiple cell signaling pathways, cell cycle progression, and cell growth, and PI3K inhibitors have been approved or are in clinical development. We examined novel PI3Kdelta inhibitors, which are highly selective for the p110delta isoform of in CD3/CD28 stimulated T-cell cytokine production. In vitro generated CD4+ T effector cells stimulated in the presence of a PI3Kdelta inhibitor demonstrated a dose-dependent suppression of cytokines produced by Th1, Th2, and Th17 cells. This effect was T-cell intrinsic, and we observed similar effects on human PBMCs. Th17 cells expressing a constitutively activated form of AKT were resistant to PI3Kdelta inhibition, suggesting that the inhibitor is acting through AKT signaling pathways. Additionally, PI3Kdelta inhibition decreased IL-17 production in vivo and decreased neutrophil recruitment to the lung in a murine model of acute pulmonary inflammation. These experiments show that targeting PI3Kdelta activity can modulate T-cell cytokine production and reduce inflammation in vivo, suggesting that PI3Kdelta inhibition could have therapeutic potential in treating inflammatory diseases.
Collapse
Affiliation(s)
- Emily E Way
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Giraldina Trevejo-Nunez
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Jay K Kolls
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Kong Chen
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
27
|
Chen K, Campfield BT, Wenzel SE, McAleer JP, Kreindler JL, Kurland G, Gopal R, Wang T, Chen W, Eddens T, Quinn KM, Myerburg MM, Horne WT, Lora JM, Albrecht BK, Pilewski JM, Kolls JK. Antiinflammatory effects of bromodomain and extraterminal domain inhibition in cystic fibrosis lung inflammation. JCI Insight 2016; 1. [PMID: 27517095 DOI: 10.1172/jci.insight.87168] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Significant morbidity in cystic fibrosis (CF) results from chronic lung inflammation, most commonly due to Pseudomonas aeruginosa infection. Recent data suggest that IL-17 contributes to pathological inflammation in the setting of abnormal mucosal immunity, and type 17 immunity-driven inflammatory responses may represent a target to block aberrant inflammation in CF. Indeed, transcriptomic analysis of the airway epithelium from CF patients undergoing clinical bronchoscopy revealed upregulation of IL-17 downstream signature genes, implicating a substantial contribution of IL-17-mediated immunity in CF lungs. Bromodomain and extraterminal domain (BET) chromatin modulators can regulate T cell responses, specifically Th17-mediated inflammation, by mechanisms that include bromodomain-dependent inhibition of acetylated histones at the IL17 locus. Here, we show that, in vitro, BET inhibition potently suppressed Th17 cell responses in explanted CF tissue and inhibited IL-17-driven chemokine production in human bronchial epithelial cells. In an acute P. aeruginosa lung infection murine model, BET inhibition decreased inflammation, without exacerbating infection, suggesting that BET inhibition may be a potential therapeutic target in patients with CF.
Collapse
Affiliation(s)
- Kong Chen
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
| | - Brian T Campfield
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA; University of Pittsburgh, Division of Pediatric Infectious Diseases, Department of Pediatrics, Pittsburgh, Pennsylvania, USA
| | - Sally E Wenzel
- University of Pittsburgh Asthma Institute at UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jeremy P McAleer
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
| | - James L Kreindler
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Geoffrey Kurland
- Division of Pediatric Pulmonology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Radha Gopal
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
| | - Ting Wang
- Division of Pulmonary Medicine, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Wei Chen
- Division of Pulmonary Medicine, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Taylor Eddens
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
| | - Kathleen M Quinn
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
| | - Mike M Myerburg
- Division of Pulmonary, Allergy, and Critical Care Medicine, UPMC, Pittsburgh, Pennsylvania, USA
| | - William T Horne
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
| | - Jose M Lora
- Constellation Pharmaceuticals Inc., Cambridge, Massachusetts, USA
| | - Brian K Albrecht
- Constellation Pharmaceuticals Inc., Cambridge, Massachusetts, USA
| | - Joseph M Pilewski
- Division of Pulmonary, Allergy, and Critical Care Medicine, UPMC, Pittsburgh, Pennsylvania, USA
| | - Jay K Kolls
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
| |
Collapse
|
28
|
Lorè NI, Cigana C, Riva C, De Fino I, Nonis A, Spagnuolo L, Sipione B, Cariani L, Girelli D, Rossi G, Basso V, Colombo C, Mondino A, Bragonzi A. IL-17A impairs host tolerance during airway chronic infection by Pseudomonas aeruginosa. Sci Rep 2016; 6:25937. [PMID: 27189736 PMCID: PMC4870500 DOI: 10.1038/srep25937] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/22/2016] [Indexed: 11/09/2022] Open
Abstract
Resistance and tolerance mechanisms participate to the interplay between host and pathogens. IL-17-mediated response has been shown to be crucial for host resistance to respiratory infections, whereas its role in host tolerance during chronic airway colonization is still unclear. Here, we investigated whether IL-17-mediated response modulates mechanisms of host tolerance during airways chronic infection by P. aeruginosa. First, we found that IL-17A levels were sustained in mice at both early and advanced stages of P. aeruginosa chronic infection and confirmed these observations in human respiratory samples from cystic fibrosis patients infected by P. aeruginosa. Using IL-17a−/− or IL-17ra−/− mice, we found that the deficiency of IL-17A/IL-17RA axis was associated with: i) increased incidence of chronic infection and bacterial burden, indicating its role in the host resistance to P. aeruginosa; ii) reduced cytokine levels (KC), tissue innate immune cells and markers of tissue damage (pro-MMP-9, elastin degradation, TGF-β1), proving alteration of host tolerance. Blockade of IL-17A activity by a monoclonal antibody, started when chronic infection is established, did not alter host resistance but increased tolerance. In conclusion, this study identifies IL-17-mediated response as a negative regulator of host tolerance during P. aeruginosa chronic airway infection.
Collapse
Affiliation(s)
- Nicola Ivan Lorè
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Cristina Cigana
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Camilla Riva
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Ida De Fino
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Alessandro Nonis
- University Center for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milano, Italy
| | - Lorenza Spagnuolo
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Barbara Sipione
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Lisa Cariani
- Cystic Fibrosis Microbiology Laboratory, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy
| | - Daniela Girelli
- Cystic Fibrosis Microbiology Laboratory, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy
| | - Giacomo Rossi
- School of Biosciences and Veterinary Medicine, University of Camerino, Italy
| | - Veronica Basso
- Lymphocytes Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Carla Colombo
- Cystic Fibrosis Center, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy
| | - Anna Mondino
- Lymphocytes Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Alessandra Bragonzi
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
29
|
Brand JD, Mathews JA, Kasahara DI, Wurmbrand AP, Shore SA. Regulation of IL-17A expression in mice following subacute ozone exposure. J Immunotoxicol 2016; 13:428-38. [PMID: 27043160 DOI: 10.3109/1547691x.2015.1120829] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Exposure to subacute ozone (O3) causes pulmonary neutrophil recruitment. In mice, this recruitment requires IL-17A. Ozone also causes expression of IL-23 and IL-1, which can induce IL-17A. The purpose of this study was to examine the hypothesis that IL-23 and IL-1 contribute to IL-17A expression and subsequent neutrophil recruitment after subacute O3 exposure. Wild-type, IL-23(-/-), and Flt3l(-/-) mice were exposed to air or 0.3 ppm O3 for 72 h. Flt3l(-/-) mice lack conventional dendritic cells (cDC) that can express IL-23 and IL-1. Other wild-type mice were pre-treated with saline or the IL-1R1 antagonist anakinra prior to O3 exposure. After exposure, bronchoalveolar lavage (BAL) was performed and lung tissue harvested. The results indicated that pulmonary Il17a mRNA abundance and IL-17A(+) F4/80(+) cells were significantly reduced in O3-exposed IL-23(-/-) vs in wild-type mice. In contrast, anakinra had no effect on Il23a or Il17a pulmonary mRNA abundance or on BAL concentrations of the neutrophil survival factor G-CSF, but anakinra did reduce BAL neutrophil numbers, likely because anakinra also reduced BAL IL-6. Compared to air, O3 caused a significant increase in DC numbers in wild-type, but not in Flt3(-/-) mice. However, there was no significant difference in Il23a or Il17a mRNA abundance or in BAL neutrophil count in O3-exposed Flt3(-/-) vs in wild-type mice. From these results, it was concluded that IL-23 but not IL-1 contributes to the IL-17A expression induced by subacute O3 exposure. Induction of IL-23 by O3 does not appear to require cDC.
Collapse
Affiliation(s)
- Jeffrey D Brand
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Joel A Mathews
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - David I Kasahara
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Alison P Wurmbrand
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Stephanie A Shore
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| |
Collapse
|
30
|
Lorè NI, Bragonzi A, Cigana C. The IL-17A/IL-17RA axis in pulmonary defence and immunopathology. Cytokine Growth Factor Rev 2016; 30:19-27. [PMID: 27033174 DOI: 10.1016/j.cytogfr.2016.03.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/17/2016] [Accepted: 03/17/2016] [Indexed: 01/11/2023]
Abstract
The interleukin (IL)-17A/IL-17 receptor A (IL-17RA) axis is emerging as a key player in host defence. Several studies have demonstrated that IL-17A-mediated responses play a critical role in both acute and chronic inflammation induced by infectious agents, environmental stimuli and genetic diseases in the airways. In this regard, it is becoming evident that IL-17A/IL-17RA signalling may have a protective and beneficial impact on health, but that it can also result in detrimental outcomes. On one hand, the IL-17A/IL-17RA axis can contribute to the elimination of noxious stimuli and to the resolution of acute inflammatory processes; on the other hand, it can exacerbate immunopathological responses, contributing to the development and progression of chronic respiratory illnesses. In addition, cellular and molecular signatures underlying IL-17A/IL-17RA signalling have been increasingly identified, although further studies are needed to clarify such complex responses. Here, we discuss the latest discoveries on the role of the IL-17A/IL-17RA axis in driving host pulmonary defence and immunopathology.
Collapse
Affiliation(s)
- Nicola Ivan Lorè
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy.
| | - Alessandra Bragonzi
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Cristina Cigana
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
31
|
McDermott AJ, Falkowski NR, McDonald RA, Pandit CR, Young VB, Huffnagle GB. Interleukin-23 (IL-23), independent of IL-17 and IL-22, drives neutrophil recruitment and innate inflammation during Clostridium difficile colitis in mice. Immunology 2016; 147:114-24. [PMID: 26455347 PMCID: PMC4693884 DOI: 10.1111/imm.12545] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/17/2015] [Accepted: 10/05/2015] [Indexed: 12/11/2022] Open
Abstract
Our objective was to determine the role of the inflammatory cytokine interleukin-23 (IL-23) in promoting neutrophil recruitment, inflammatory cytokine expression and intestinal histopathology in response to Clostridium difficile infection. Wild-type (WT) and p19(-/-) (IL-23KO) mice were pre-treated with cefoperazone in their drinking water for 5 days, and after a 2-day recovery period were challenged with spores from C. difficile strain VPI 10463. Interleukin-23 deficiency was associated with significant defects in both the recruitment of CD11b(High) Ly6G(H) (igh) neutrophils to the colon and the expression of neutrophil chemoattractants and stabilization factors including Cxcl1, Cxcl2, Ccl3 and Csf3 within the colonic mucosa as compared with WT animals. Furthermore, the expression of inflammatory cytokines including Il33, Tnf and Il6 was significantly reduced in IL-23-deficient animals. There was also a trend towards less severe colonic histopathology in the absence of IL-23. The induction of Il17a and Il22 was also significantly abrogated in IL-23KO mice. Inflammatory cytokine expression and neutrophilic inflammation were not reduced in IL-17a-deficient mice or in mice treated with anti-IL-22 depleting monoclonal antibody. However, induction of RegIIIg was significantly reduced in animals treated with anti-IL-22 antibody. Taken together, these data indicate that IL-23, but not IL-17a or IL-22, promotes neutrophil recruitment and inflammatory cytokine and chemokine expression in the colon in response to C. difficile infection.
Collapse
Affiliation(s)
- Andrew J. McDermott
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Nicole R. Falkowski
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Roderick A. McDonald
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Chinmay R. Pandit
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Vincent B. Young
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
- Division of Infectious DiseasesDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Gary B. Huffnagle
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
| |
Collapse
|
32
|
Gonçalves-de-Albuquerque CF, Silva AR, Burth P, Rocco PRM, Castro-Faria MV, Castro-Faria-Neto HC. Possible mechanisms of Pseudomonas aeruginosa-associated lung disease. Int J Med Microbiol 2015; 306:20-8. [PMID: 26652129 DOI: 10.1016/j.ijmm.2015.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 11/11/2015] [Accepted: 11/14/2015] [Indexed: 12/22/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic bacterium causing lung injury in immunocompromised patients correlated with high morbidity and mortality. Many bacteria, including P. aeruginosa, use extracellular signals to synchronize group behaviors, a process known as quorum sensing (QS). In the P. aeruginosa complex QS system controls expression of over 300 genes, including many involved in host colonization and disease. P. aeruginosa infection elicits a complex immune response due to a large number of immunogenic factors present in the bacteria or released during infection. Here, we focused on the mechanisms by which P. aeruginosa triggers lung injury and inflammation, debating the possible ways that P. aeruginosa evades the host immune system, which leads to immune suppression and resistance.
Collapse
Affiliation(s)
| | - Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Patrícia Burth
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Patricia Rieken Macêdo Rocco
- Laboratório de Investigação Pulmonar, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mauro Velho Castro-Faria
- Laboratório Integrado de Nefrologia, Departamento de Medicina Interna, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
33
|
Ethanol impairs mucosal immunity against Streptococcus pneumoniae infection by disrupting interleukin 17 gene expression. Infect Immun 2015; 83:2082-8. [PMID: 25754201 DOI: 10.1128/iai.02869-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 02/21/2015] [Indexed: 12/31/2022] Open
Abstract
Acute ethanol intoxication suppresses the host immune responses against Streptococcus pneumoniae. As interleukin 17 (IL-17) is a critical cytokine in host defense against extracellular pathogens, including S. pneumoniae, we hypothesized that ethanol impairs mucosal immunity against this pathogen by disrupting IL-17 production or IL-17 receptor (IL-17R) signaling. A chronic ethanol feeding model in simian immunodeficiency virus (SIV)-infected rhesus macaques and acute ethanol intoxication in a murine model were used. Transcriptome analysis of bronchial brushes in the nonhuman primate model showed downregulation of the expression of IL-17-regulated chemokines in ethanol-fed animals, a finding also replicated in the murine model. Surprisingly, recombinant CXCL1 and CXCL5 but not IL-17 or IL-23 plus IL-1β rescued bacterial burden in the ethanol group to control levels. Taken together, the results of this study suggest that ethanol impairs IL-17-mediated chemokine production in the lung. Thus, exogenous luminal restoration of IL-17-related chemokines, CXCL1 and CXCL5, improves host defenses against S. pneumoniae.
Collapse
|
34
|
Beisswenger C, Honecker A, Kamyschnikow A, Bischoff M, Tschernig T, Bals R. Moxifloxacin modulates inflammation during murine pneumonia. Respir Res 2014; 15:82. [PMID: 25034539 PMCID: PMC4118268 DOI: 10.1186/1465-9921-15-82] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 07/10/2014] [Indexed: 12/14/2022] Open
Abstract
Background Moxifloxacin is a synthetic antibacterial agent belonging to the fluoroquinolone family. The antimicrobial activity of quinolones against Gram-positive and Gram-negative bacteria is based on their ability to inhibit topoisomerases. Quinolones are described to have immunomodulatory features in addition to their antimicrobial activities. It was the goal of this study to examine whether a short term treatment with moxifloxacin modulates the inflammation during a subsequently induced bacterial infection in an animal model. Methods Mice were treated with moxifloxacin or saline for two consecutive days and were subsequently intranasally infected with viable or heat-inactivated bacterial pathogens (Streptococcus pneumoniae, Pseudomonas aeruginosa) for 6 and 24 hours. Measurements of cytokines in the lungs and plasma were performed. Alveolar cells were determined in bronchoalveolar lavage fluits. Results The inflammation was increased after the inoculation of viable bacteria compared to inactivated bacteria. Numbers of total immune cells and neutrophils and concentrations of inflammatory mediators (e.g. KC, IL-1β, IL-17A) were significantly reduced in lungs of moxifloxacin-treated mice infected with inactivated and viable bacterial pathogens as compared to infected control mice. Plasma concentrations of inflammatory mediators were significantly reduced in moxifloxacin-treated mice. Immunohistochemistry showed a stronger infiltrate of TNF-α-expressing cells into lungs of saline-treated mice infected with viable P. aeruginosa as compared to moxifloxacin-treated mice. Conclusions These data show that in this pneumonia model moxifloxacin has anti-inflammatory properties beyond its antibacterial activity.
Collapse
Affiliation(s)
- Christoph Beisswenger
- Department of Internal Medicine V - Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany.
| | | | | | | | | | | |
Collapse
|
35
|
Faure E, Mear JB, Faure K, Normand S, Couturier-Maillard A, Grandjean T, Balloy V, Ryffel B, Dessein R, Chignard M, Uyttenhove C, Guery B, Gosset P, Chamaillard M, Kipnis E. Pseudomonas aeruginosa type-3 secretion system dampens host defense by exploiting the NLRC4-coupled inflammasome. Am J Respir Crit Care Med 2014; 189:799-811. [PMID: 24555512 DOI: 10.1164/rccm.201307-1358oc] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
RATIONALE Pseudomonas aeruginosa, a major problem pathogen responsible for severe infections in critically ill patients, triggers, through a functional type-3 secretion system (T3SS), the activation of an intracellular cytosolic sensor of innate immunity, NLRC4. Although the NLRC4-inflammasome-dependent response contributes to increased clearance of intracellular pathogens, it seems that NLRC4 inflammasome activation decreases the clearance of P. aeruginosa, a mainly extracellular pathogen. OBJECTIVES We sought to determine the underlying mechanisms of this effect of the activation of NLRC4 by P. aeruginosa. METHODS We established acute lung injury in wild-type and Nlrc4(-/-) mice using sublethal intranasal inocula of P. aeruginosa strain CHA expressing or not a functional T3SS. We studied 96-hour survival, lung injury, bacterial clearance from the lungs, cytokine secretion in bronchoalveolar lavage, lung antimicrobial peptide expression by quantitative polymerase chain reaction, and flow cytometry analysis of lung cells. MEASUREMENTS AND MAIN RESULTS Nlrc4(-/-) mice showed enhanced bacterial clearance and decreased lung injury contributing to increased survival against extracellular P. aeruginosa strain expressing a functional T3SS. The mechanism involved decreased NLRC4-inflammasome-driven IL-18 secretion attenuating lung injury caused by excessive neutrophil recruitment. Additionally, in the lungs of Nlrc4(-/-) mice secretion of IL-17 by innate immune cells was increased and responsible for increased expression of lung epithelial antimicrobial peptides. Furthermore, IL-18 secretion was found to repress IL-17 and IL-17-driven lung antimicrobial peptide expression. CONCLUSIONS We report a new role of the T3SS apparatus itself, independently of exotoxin translocation. Through NLRC4 inflammasome activation, the T3SS promotes IL-18 secretion, which dampens a beneficial IL-17-mediated antimicrobial host response.
Collapse
Affiliation(s)
- Emmanuel Faure
- 1 Pseudomonas aeruginosa Host-Pathogen Translational Research Group, Université Droit et Santé de Lille, Lille, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Clinical consequences of targeting IL-17 and TH17 in autoimmune and allergic disorders. Curr Allergy Asthma Rep 2014; 13:587-95. [PMID: 23760974 DOI: 10.1007/s11882-013-0361-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The TH17 lineage of T cells and its canonical cytokine IL-17 have been the focus of many recent studies in autoimmune, allergic, and infectious disease. In this review, we will briefly discuss the current knowledge about the role of these cells and IL-17 in a spectrum of disorders. It is clear that IL-17 plays pathogenic roles in certain conditions while the same pathway is critically important to immunity in others. Targeting of TH17 cells or IL-17 therapeutically may impart many benefits, but this approach is not without potentially serious implications regarding host defense. These issues will be discussed herein as we evaluate pharmacological approaches targeting this pathway that are just beginning to be fully tested in human disease.
Collapse
|
37
|
PrtR homeostasis contributes to Pseudomonas aeruginosa pathogenesis and resistance against ciprofloxacin. Infect Immun 2014; 82:1638-47. [PMID: 24491574 DOI: 10.1128/iai.01388-13] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that causes acute and chronic infections in humans. Pyocins are bacteriocins produced by P. aeruginosa that are usually released through lysis of the producer strains. Expression of pyocin genes is negatively regulated by PrtR, which gets cleaved under SOS response, leading to upregulation of pyocin synthetic genes. Previously, we demonstrated that PrtR is required for the expression of type III secretion system (T3SS), which is an important virulence component of P. aeruginosa. In this study, we demonstrate that mutation in prtR results in reduced bacterial colonization in a mouse acute pneumonia model. Examination of bacterial and host cells in the bronchoalveolar lavage fluids from infected mice revealed that expression of PrtR is induced by reactive oxygen species (ROS) released by neutrophils. We further demonstrate that treatment with hydrogen peroxide or ciprofloxacin, known to induce the SOS response and pyocin production, resulted in an elevated PrtR mRNA level. Overexpression of PrtR by a tac promoter repressed the endogenous prtR promoter activity, and electrophoretic mobility shift assay revealed that PrtR binds to its own promoter, suggesting an autorepressive mechanism of regulation. A high level of PrtR expressed from a plasmid resulted in increased T3SS gene expression during infection and higher resistance against ciprofloxacin. Overall, our results suggest that the autorepression of PrtR contributes to the maintenance of a relatively stable level of PrtR, which is permissive to T3SS gene expression in the presence of ROS while increasing bacterial tolerance to stresses, such as ciprofloxacin, by limiting pyocin production.
Collapse
|
38
|
Abstract
The Th17 pathway has recently been shown to play a critical role in host defense, allergic responses and autoimmune inflammation. Th17 cells predominantly produce IL-17 and IL-22, which are two cytokines with broad effects in the lung and other tissues. This review summarizes not only what is currently known about the molecular regulation of this pathway and Th17-related cytokine signaling, but also the roles of these cytokines in pathogen immunity and asthma. In the last 5 years, the Th17 field has rapidly grown and research has revealed that the Th17 pathway is essential in lung pathogenesis in response to exogenous stimuli. As work in the field continues, it is expected that many exciting therapeutic advances will be made for a broad range of diseases.
Collapse
Affiliation(s)
- Michelle L Manni
- Department of Pediatrics, Division of Pulmonary Medicine, Allergy, and Immunology, Children’s Hospital of Pittsburgh of UPMC, One Children’s Hospital, Dr, 9127 Rangos, 4401 Penn Ave., Pittsburgh, PA 15224, USA
| | - Keven M Robinson
- Department of Pediatrics, Division of Pulmonary Medicine, Allergy, and Immunology, Children’s Hospital of Pittsburgh of UPMC, One Children’s Hospital, Dr, 9127 Rangos, 4401 Penn Ave., Pittsburgh, PA 15224, USA
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - John F Alcorn
- Department of Pediatrics, Division of Pulmonary Medicine, Allergy, and Immunology, Children’s Hospital of Pittsburgh of UPMC, One Children’s Hospital, Dr, 9127 Rangos, 4401 Penn Ave., Pittsburgh, PA 15224, USA
| |
Collapse
|
39
|
Abstract
Evidence has increasingly shown that the lungs are a major site of immune regulation. A robust and highly regulated immune response in the lung protects the host from pathogen infection, whereas an inefficient or deleterious response can lead to various pulmonary diseases. Many cell types, such as epithelial cells, dendritic cells, macrophages, neutrophils, eosinophils, and B and T lymphocytes, contribute to lung immunity. This review focuses on the recent advances in understanding how T lymphocytes mediate pulmonary host defenses against bacterial, viral, and fungal pathogens.
Collapse
Affiliation(s)
- Kong Chen
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15201, USA
| | | |
Collapse
|
40
|
Suryawanshi A, Cao Z, Thitiprasert T, Zaidi TS, Panjwani N. Galectin-1-mediated suppression of Pseudomonas aeruginosa-induced corneal immunopathology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:6397-409. [PMID: 23686486 PMCID: PMC3689592 DOI: 10.4049/jimmunol.1203501] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Corneal infection with Pseudomonas aeruginosa leads to a severe immunoinflammatory lesion, often causing vision impairment and blindness. Although past studies have indicated a critical role for CD4(+) T cells, particularly Th1 cells, in corneal immunopathology, the relative contribution of recently discovered Th17 and regulatory T cells is undefined. In this study, we demonstrate that after corneal P. aeruginosa infection, both Th1 and Th17 cells infiltrate the cornea with increased representation of Th17 cells. In addition to Th1 and Th17 cells, regulatory T cells also migrate into the cornea during early as well as late stages of corneal pathology. Moreover, using galectin-1 (Gal-1), an immunomodulatory carbohydrate-binding molecule, we investigated whether shifting the balance among various CD4(+) T cell subsets can modulate P. aeruginosa-induced corneal immunopathology. We demonstrate in this study that local recombinant Gal-1 (rGal-1) treatment by subconjunctival injections significantly diminishes P. aeruginosa-mediated corneal inflammation through multiple mechanisms. Specifically, in our study, rGal-1 treatment significantly diminished corneal infiltration of total CD45(+) T cells, neutrophils, and CD4(+) T cells. Furthermore, rGal-1 treatment significantly reduced proinflammatory Th17 cell response in the cornea as well as local draining lymph nodes. Also, rGal-1 therapy promoted anti-inflammatory Th2 and IL-10 response in secondary lymphoid organs. Collectively, our results indicate that corneal P. aeruginosa infection induces a strong Th17-mediated corneal pathology, and treatment with endogenously derived protein such as Gal-1 may be of therapeutic value for the management of bacterial keratitis, a prevalent cause of vision loss and blindness in humans worldwide.
Collapse
Affiliation(s)
- Amol Suryawanshi
- New England Eye Center and Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02111
| | - Zhiyi Cao
- New England Eye Center and Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02111
| | - Thananya Thitiprasert
- New England Eye Center and Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02111
| | - Tanveer S. Zaidi
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115
| | - Noorjahan Panjwani
- New England Eye Center and Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02111
- Department of Biochemistry, Tufts University School of Medicine, Boston, MA 02111
| |
Collapse
|
41
|
Liu J, Qu H, Li Q, Ye L, Ma G, Wan H. The responses of γδ T-cells against acute Pseudomonas aeruginosa pulmonary infection in mice via interleukin-17. Pathog Dis 2013; 68:44-51. [PMID: 23620413 DOI: 10.1111/2049-632x.12043] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 03/19/2013] [Accepted: 04/03/2013] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-17-producing T-lymphocytes play a crucial role in inflammation, yet the potential roles of the cells in acute bacterial pulmonary infection remain unclear. Here, we investigated the role of IL-17-producing γδ T-cells in a mouse model of acute Pseudomonas aeruginosa pulmonary infection. Results showed that augmentation of IL-17, IL-22 and IL-23 was associated with the development of acute bacterial pulmonary infection. However, IL-17 was markedly reduced following the blockade of γδ T-cell activity in vivo. The levels of the chemokines, including granulocyte colony-stimulating factor (G-CSF), keratinocyte chemoattractant (KC), macrophage inflammatory protein-1α (MIP-1) and macrophage inflammatory protein (MIP-2), were also noticeably decreased in the anti-γδ T Cell Receptor(TCR) mice after 8 h infection. Following the depletion of γδ T-cells, the bacterial load was consistently increased. Anti-TCRγδ-treated mice had changes similar to those in the the anti-IL-17-treated mice. The mRNA and protein levels of IL-22 and IL-23, and the mRNA level of RORγt were all markedly decreased in the anti-TCRγδ mice. Overall, our results demonstrated that at the early stage of acute P. aeruginosa pulmonary infection, γδ T-cells are the major source of IL-17 and play a pivotal role in the host immune response and defense against bacteria.
Collapse
Affiliation(s)
- Jialin Liu
- Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | |
Collapse
|
42
|
Kumar P, Chen K, Kolls JK. Th17 cell based vaccines in mucosal immunity. Curr Opin Immunol 2013; 25:373-80. [PMID: 23669353 DOI: 10.1016/j.coi.2013.03.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 03/29/2013] [Indexed: 12/18/2022]
Abstract
Vaccination is proven to be effective in controlling many infections including small pox, influenza and hepatitis, but strain-specific factors may limit vaccine efficacy. All of these vaccines work through the generation of neutralizing antibodies but for some pathogens there may be roles for serotype-independent immunity. Recently several groups using murine vaccine models have shown that induced T helper cell responses including Th17 responses have shown the potential for CD4+ T-cell dependent vaccine responses. Th17 mediated protective responses involve the recruitment of neutrophils, release of anti-microbial peptides and IL-17-driven Th1 immunity. These effector mechanisms provide immunity against a range of pathogens including the recently described antibiotic-resistant metallo-beta-lactamase 1 Klebsiella pneumoniae. Continued elucidation of the mechanism of Th17 responses and identification of effective adjuvants for inducing robust non pathogenic Th17 responses may lead to successful Th17 based vaccines. Here we summarize the recent advances in understanding the role of Th17 in vaccine induced immunity. We also discuss the current status and future challenges in Th17-based mucosal vaccine development.
Collapse
Affiliation(s)
- Pawan Kumar
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | |
Collapse
|
43
|
Cohen TS, Prince AS. Activation of inflammasome signaling mediates pathology of acute P. aeruginosa pneumonia. J Clin Invest 2013; 123:1630-7. [PMID: 23478406 DOI: 10.1172/jci66142] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 01/17/2013] [Indexed: 01/03/2023] Open
Abstract
The respiratory tract is exceptionally well defended against infection from inhaled bacteria, with multiple proinflammatory signaling cascades recruiting phagocytes to clear airway pathogens. However, organisms that efficiently activate damaging innate immune responses, such as those mediated by the inflammasome and caspase-1, may cause pulmonary damage and interfere with bacterial clearance. The extracellular, opportunistic pathogen Pseudomonas aeruginosa expresses not only pathogen-associated molecular patterns that activate NF-κB signaling in epithelial and immune cells, but also flagella that activate the NLRC4 inflammasome. We demonstrate that induction of inflammasome signaling, ascribed primarily to the alveolar macrophage, impaired P. aeruginosa clearance and was associated with increased apoptosis/pyroptosis and mortality in a murine model of acute pneumonia. Strategies that limited inflammasome activation, including infection by fliC mutants, depletion of macrophages, deletion of NLRC4, reduction of IL-1β and IL-18 production, inhibition of caspase-1, and inhibition of downstream signaling in IL-1R- or IL-18R-null mice, all resulted in enhanced bacterial clearance and diminished pathology. These results demonstrate that the inflammasome provides a potential target to limit the pathological consequences of acute P. aeruginosa pulmonary infection.
Collapse
Affiliation(s)
- Taylor S Cohen
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | | |
Collapse
|
44
|
Rieber N, Brand A, Hector A, Graepler-Mainka U, Ost M, Schäfer I, Wecker I, Neri D, Wirth A, Mays L, Zundel S, Fuchs J, Handgretinger R, Stern M, Hogardt M, Döring G, Riethmüller J, Kormann M, Hartl D. Flagellin Induces Myeloid-Derived Suppressor Cells: Implications forPseudomonas aeruginosaInfection in Cystic Fibrosis Lung Disease. THE JOURNAL OF IMMUNOLOGY 2012; 190:1276-84. [DOI: 10.4049/jimmunol.1202144] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
45
|
Hartl D, Gaggar A, Bruscia E, Hector A, Marcos V, Jung A, Greene C, McElvaney G, Mall M, Döring G. Innate immunity in cystic fibrosis lung disease. J Cyst Fibros 2012; 11:363-82. [PMID: 22917571 DOI: 10.1016/j.jcf.2012.07.003] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 06/29/2012] [Accepted: 07/02/2012] [Indexed: 12/16/2022]
Abstract
Chronic lung disease determines the morbidity and mortality of cystic fibrosis (CF) patients. The pulmonary immune response in CF is characterized by an early and non-resolving activation of the innate immune system, which is dysregulated at several levels. Here we provide a comprehensive overview of innate immunity in CF lung disease, involving (i) epithelial dysfunction, (ii) pathogen sensing, (iii) leukocyte recruitment, (iv) phagocyte impairment, (v) mechanisms linking innate and adaptive immunity and (iv) the potential clinical relevance. Dissecting the complex network of innate immune regulation and associated pro-inflammatory cascades in CF lung disease may pave the way for novel immune-targeted therapies in CF and other chronic infective lung diseases.
Collapse
Affiliation(s)
- D Hartl
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Host response signature to Staphylococcus aureus alpha-hemolysin implicates pulmonary Th17 response. Infect Immun 2012; 80:3161-9. [PMID: 22733574 DOI: 10.1128/iai.00191-12] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Staphylococcus aureus pneumonia causes significant morbidity and mortality. Alpha-hemolysin (Hla), a pore-forming cytotoxin of S. aureus, has been identified through animal models of pneumonia as a critical virulence factor that induces lung injury. In spite of considerable molecular knowledge of how this cytotoxin injures the host, the precise host response to Hla in the context of infection remains poorly understood. We employed whole-genome expression profiling of infected lungs to define the host response to wild-type S. aureus compared with the response to an Hla-deficient isogenic mutant in experimental pneumonia. These data provide a complete expression profile at 4 and at 24 h postinfection, revealing a unique response to the toxin-expressing strain. Gene ontogeny analysis revealed significant differences in the extracellular matrix and cardiomyopathy pathways, both of which govern cellular interactions in the tissue microenvironment. Evaluation of individual transcript responses to Hla-secreting staphylococci was notable for upregulation of host cytokine and chemokine genes, including the p19 subunit of interleukin-23. Consistent with this observation, the cellular immune response to infection was characterized by a prominent Th17 response to the wild-type pathogen. These findings define specific host mRNA responses to Hla-producing S. aureus, coupling the pulmonary Th17 response to the secretion of this cytotoxin. Expression profiling to define the host response to a single virulence factor proved to be a valuable tool in identifying pathways for further investigation in S. aureus pneumonia. This approach may be broadly applicable to the study of bacterial toxins, defining host pathways that can be targeted to mitigate toxin-induced disease.
Collapse
|
47
|
Song L, Weng D, Liu F, Chen Y, Li C, Dong L, Tang W, Chen J. Tregs promote the differentiation of Th17 cells in silica-induced lung fibrosis in mice. PLoS One 2012; 7:e37286. [PMID: 22615967 PMCID: PMC3352873 DOI: 10.1371/journal.pone.0037286] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 04/17/2012] [Indexed: 01/01/2023] Open
Abstract
Background Silicosis is an occupational lung disease caused by inhalation of silica dust and characterized by lung inflammation and fibrosis. Previous study showed that Tregs regulate the process of silicosis by modulating the maintenance of immune homeostasis in the lung. Th17 cells share reciprocal developmental pathway with Tregs and play a pivotal role in the immunopathogenesis of many lung diseases by recruiting and activating neutrophils, but the regulatory function of Tregs on Th17 response in silica induced lung fibrosis remains to be explored. Methodology/Principal Findings To evaluate the role of Th17 and IL-17 in the development of silicosis and their interaction with Tregs, Treg-depleted mice model was generated and exposed to silica to establish experimental model of silica-induced lung fibrosis. Here we showed that silica increased Th17 response in lung fibrosis. Tregs depletion enhanced the neutrophils accumulation and attenuated Th17 response in silica induced lung fibrosis. Both mRNA and protein results showed that Tregs exerted its modulatory function on Th17 cells and IL-17 by regulating TGF-β1 and IL-1β. Conclusion/Significance Our study suggested that Tregs could promote Th17 cells differentiation by regulating TGF-β1 and IL-1β in silica induced lung fibrosis of mice, which further the understanding of the progress of silicosis and provide a new insight in the regulatory mechanism of Th17 by Tregs in lung inflammation.
Collapse
Affiliation(s)
- Laiyu Song
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Dong Weng
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Fangwei Liu
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Ying Chen
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Cuiying Li
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Lei Dong
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Wen Tang
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Jie Chen
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, People's Republic of China
- * E-mail:
| |
Collapse
|