51
|
Domínguez‐Andrés J, Ferreira AV, Jansen T, Smithers N, Prinjha RK, Furze RC, Netea MG. Bromodomain inhibitor I-BET151 suppresses immune responses during fungal-immune interaction. Eur J Immunol 2019; 49:2044-2050. [PMID: 31206650 PMCID: PMC6899658 DOI: 10.1002/eji.201848081] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/06/2019] [Accepted: 06/12/2019] [Indexed: 12/29/2022]
Abstract
Changes in the epigenetic landscape of immune cells are a crucial component of gene activation during the induction of inflammatory responses, therefore it has been hypothesized that epigenetic modulation could be employed to restore homeostasis in inflammatory scenarios. Fungal pathogens cause a large burden of morbidity and even mortality due to the hyperinflammatory processes that induce mucosal, allergic or systemic infections. Bromodomain and extraterminal domain (BET) proteins are considered as one as the most tantalizing pharmacological targets for the modulation of inflammatory responses at the epigenetic level. Nothing is known of the role of BET inhibitors on the inflammation induced by fungal pathogens. In the present study, we assessed the in vitro efficacy of the small molecular histone mimic BET inhibitor I-BET151 to modulate innate immune responses during fungal-immune interaction with the clinically relevant fungal pathogens Candida albicans and Aspergillus fumigatus. Our results prove that BET inhibitors (I-BETs) represent an important modulator of inflammation induced by fungal pathogens: both direct production of proinflammatory cytokines and the induction of trained immunity were inhibited by I-BET151. These modulatory effects are likely to have important potential implications in clinically relevant situations.
Collapse
Affiliation(s)
- Jorge Domínguez‐Andrés
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI)Radboud University Nijmegen Medical Centre6500HBNijmegenthe Netherlands
| | - Anaísa V Ferreira
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI)Radboud University Nijmegen Medical Centre6500HBNijmegenthe Netherlands
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS)Universidade do Porto4050‐313PortoPortugal
| | - Trees Jansen
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI)Radboud University Nijmegen Medical Centre6500HBNijmegenthe Netherlands
| | - Nicholas Smithers
- Epigenetics DPU, Immuno‐Inflammation Therapy AreaGlaxoSmithKline R&D, Medicines Research CentreStevenageSG1 2NYUK
| | - Rab K. Prinjha
- Epigenetics DPU, Immuno‐Inflammation Therapy AreaGlaxoSmithKline R&D, Medicines Research CentreStevenageSG1 2NYUK
| | - Rebecca C. Furze
- Epigenetics DPU, Immuno‐Inflammation Therapy AreaGlaxoSmithKline R&D, Medicines Research CentreStevenageSG1 2NYUK
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI)Radboud University Nijmegen Medical Centre6500HBNijmegenthe Netherlands
- Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES)University of Bonn53115BonnGermany
- Human Genomics LaboratoryCraiova University of Medicine and PharmacyCraiovaRomania
| |
Collapse
|
52
|
Gontijo AV, G Sampaio AD, Koga-Ito CY, Salvador MJ. Biopharmaceutical and antifungal properties of ellagic acid-cyclodextrin using an in vitro model of invasive candidiasis. Future Microbiol 2019; 14:957-967. [PMID: 31373226 DOI: 10.2217/fmb-2019-0107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To investigate biopharmaceutical and antifungal properties of pure and complexed ellagic acid. Materials & methods: Caco-2 cells cultured in a Transwell® inserts were infected with Candida albicans to develop an in vitro model. Ellagic acid was complexed with cyclodextrins. Microbial compositions, ellagic acid concentration as function of time and characterization studies of complexes were evaluated. Results: Ellagic acid presented ability to reduce C. albicans invasion, although this was not statistically significant. Its poor water solubility and absorption probably limited this ability. Water solubility was increased after complexation with hydroxypropyl-β-CD; however, ellagic acid/hydroxypropyl-β-CD did not improve the antifungal activity. Conclusion: Although ellagic acid presented a promising antifungal activity, its biopharmaceutical properties limit such activity and should be improved.
Collapse
Affiliation(s)
- Aline Vl Gontijo
- Department of Plant Biology, Institute of Biology, University of Campinas/UNICAMP, Campinas, SP, Brazil.,Department of Environmental Engineering & Oral Biopathology Graduate Program, São José dos Campos Institute of Science & Technology, São Paulo State University - UNESP, SP, Brazil
| | - Aline da G Sampaio
- Department of Environmental Engineering & Oral Biopathology Graduate Program, São José dos Campos Institute of Science & Technology, São Paulo State University - UNESP, SP, Brazil
| | - Cristiane Y Koga-Ito
- Department of Environmental Engineering & Oral Biopathology Graduate Program, São José dos Campos Institute of Science & Technology, São Paulo State University - UNESP, SP, Brazil
| | - Marcos J Salvador
- Department of Plant Biology, Institute of Biology, University of Campinas/UNICAMP, Campinas, SP, Brazil
| |
Collapse
|
53
|
Shivarathri R, Tscherner M, Zwolanek F, Singh NK, Chauhan N, Kuchler K. The Fungal Histone Acetyl Transferase Gcn5 Controls Virulence of the Human Pathogen Candida albicans through Multiple Pathways. Sci Rep 2019; 9:9445. [PMID: 31263212 PMCID: PMC6603162 DOI: 10.1038/s41598-019-45817-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/14/2019] [Indexed: 12/28/2022] Open
Abstract
Fungal virulence is regulated by a tight interplay of transcriptional control and chromatin remodelling. Despite compelling evidence that lysine acetylation modulates virulence of pathogenic fungi such as Candida albicans, the underlying mechanisms have remained largely unexplored. We report here that Gcn5, a paradigm lysyl-acetyl transferase (KAT) modifying both histone and non-histone targets, controls fungal morphogenesis - a key virulence factor of C. albicans. Our data show that genetic removal of GCN5 abrogates fungal virulence in mice, suggesting strongly diminished fungal fitness in vivo. This may at least in part arise from increased susceptibility to killing by macrophages, as well as by other phagocytes such as neutrophils or monocytes. Loss of GCN5 also causes hypersensitivity to the fungicidal drug caspofungin. Caspofungin hypersusceptibility requires the master regulator Efg1, working in concert with Gcn5. Moreover, Gcn5 regulates multiple independent pathways, including adhesion, cell wall-mediated MAP kinase signaling, hypersensitivity to host-derived oxidative stress, and regulation of the Fks1 glucan synthase, all of which play critical roles in virulence and antifungal susceptibility. Hence, Gcn5 regulates fungal virulence through multiple mechanisms, suggesting that specific inhibition of Gcn5 could offer new therapeutic strategies to combat invasive fungal infections.
Collapse
Affiliation(s)
- Raju Shivarathri
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, A-1030, Vienna, Austria
| | - Michael Tscherner
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, A-1030, Vienna, Austria
| | - Florian Zwolanek
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, A-1030, Vienna, Austria
| | | | - Neeraj Chauhan
- Public Health Research Institute, New Jersey Medical School, Rutgers The State University of New Jersey, Newark, NJ, 07103, USA.
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers The State University of New Jersey, Newark, NJ, 07103, USA.
| | - Karl Kuchler
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, A-1030, Vienna, Austria.
| |
Collapse
|
54
|
Shin SD, Shin A, Mayagoitia K, Wilson CG, Bellinger DL, Soriano S. Interferon downstream signaling is activated early in pre-symptomatic Niemann-Pick disease type C. Neurosci Lett 2019; 706:43-50. [DOI: 10.1016/j.neulet.2019.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/02/2019] [Accepted: 05/04/2019] [Indexed: 12/16/2022]
|
55
|
Leukotriene B4–type I interferon axis regulates macrophage-mediated disease tolerance to influenza infection. Nat Microbiol 2019; 4:1389-1400. [DOI: 10.1038/s41564-019-0444-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 04/03/2019] [Indexed: 11/09/2022]
|
56
|
Martins R, Carlos AR, Braza F, Thompson JA, Bastos-Amador P, Ramos S, Soares MP. Disease Tolerance as an Inherent Component of Immunity. Annu Rev Immunol 2019; 37:405-437. [PMID: 30673535 DOI: 10.1146/annurev-immunol-042718-041739] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pathogenic organisms exert a negative impact on host health, revealed by the clinical signs of infectious diseases. Immunity limits the severity of infectious diseases through resistance mechanisms that sense and target pathogens for containment, killing, or expulsion. These resistance mechanisms are viewed as the prevailing function of immunity. Under pathophysiologic conditions, however, immunity arises in response to infections that carry health and fitness costs to the host. Therefore, additional defense mechanisms are required to limit these costs, before immunity becomes operational as well as thereafter to avoid immunopathology. These are tissue damage control mechanisms that adjust the metabolic output of host tissues to different forms of stress and damage associated with infection. Disease tolerance is the term used to define this defense strategy, which does not exert a direct impact on pathogens but is essential to limit the health and fitness costs of infection. Under this argument, we propose that disease tolerance is an inherent component of immunity.
Collapse
Affiliation(s)
- Rui Martins
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal;
| | | | - Faouzi Braza
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal;
| | | | | | - Susana Ramos
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal;
| | | |
Collapse
|
57
|
Del Fresno C, Saz-Leal P, Enamorado M, Wculek SK, Martínez-Cano S, Blanco-Menéndez N, Schulz O, Gallizioli M, Miró-Mur F, Cano E, Planas A, Sancho D. DNGR-1 in dendritic cells limits tissue damage by dampening neutrophil recruitment. Science 2018; 362:351-356. [PMID: 30337411 DOI: 10.1126/science.aan8423] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 06/13/2018] [Accepted: 09/03/2018] [Indexed: 12/14/2022]
Abstract
Host injury triggers feedback mechanisms that limit tissue damage. Conventional type 1 dendritic cells (cDC1s) express dendritic cell natural killer lectin group receptor-1 (DNGR-1), encoded by the gene Clec9a, which senses tissue damage and favors cross-presentation of dead-cell material to CD8+ T cells. Here we find that DNGR-1 additionally reduces host-damaging inflammatory responses induced by sterile and infectious tissue injury in mice. DNGR-1 deficiency leads to exacerbated caerulein-induced necrotizing pancreatitis and increased pathology during systemic Candida albicans infection without affecting fungal burden. This effect is B and T cell-independent and attributable to increased neutrophilia in DNGR-1-deficient settings. Mechanistically, DNGR-1 engagement activates SHP-1 and inhibits MIP-2 (encoded by Cxcl2) production by cDC1s during Candida infection. This consequently restrains neutrophil recruitment and promotes disease tolerance. Thus, DNGR-1-mediated sensing of injury by cDC1s serves as a rheostat for the control of tissue damage, innate immunity, and immunopathology.
Collapse
Affiliation(s)
- Carlos Del Fresno
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| | - Paula Saz-Leal
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Michel Enamorado
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Stefanie K Wculek
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sarai Martínez-Cano
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Noelia Blanco-Menéndez
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Oliver Schulz
- Immunobiology Laboratory, The Francis Crick Institute, London, UK.
| | - Mattia Gallizioli
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Francesc Miró-Mur
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Eva Cano
- Chronic Disease Programme-CROSADIS, Instituto De Salud Carlos III, Madrid, Spain
| | - Anna Planas
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB-CSIC), Barcelona, Spain
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
58
|
Group A Streptococcal DNase Sda1 Impairs Plasmacytoid Dendritic Cells' Type 1 Interferon Response. J Invest Dermatol 2018; 139:1284-1293. [PMID: 30543898 DOI: 10.1016/j.jid.2018.11.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 01/19/2023]
Abstract
Group A Streptococcus causes severe invasive infections, including necrotizing fasciitis. The expression of an array of virulence factors targeting specific host immune functions impedes successful bacterial clearance. The virulence factor streptococcal DNase Sda1 was previously shown to interfere with the entrapment of bacteria through neutrophil extracellular traps and TLR9 signaling. In this study, we showed that plasmacytoid dendritic cells are recruited to the infected tissue during group A streptococcal necrotizing fasciitis. We found that the streptococcal DNase Sda1 impairs plasmacytoid dendritic cell recruitment by reducing IFN-1 levels at the site of infection. We found that streptococcal DNase Sda1 interferes with stabilization of the DNA by the host molecule HMGB1 protein, which may account for decreased IFN-1 levels at the site of infection.
Collapse
|
59
|
Guo Y, Chang Q, Cheng L, Xiong S, Jia X, Lin X, Zhao X. C-Type Lectin Receptor CD23 Is Required for Host Defense against Candida albicans and Aspergillus fumigatus Infection. THE JOURNAL OF IMMUNOLOGY 2018; 201:2427-2440. [DOI: 10.4049/jimmunol.1800620] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/06/2018] [Indexed: 12/27/2022]
|
60
|
Guiducci E, Lemberg C, Küng N, Schraner E, Theocharides APA, LeibundGut-Landmann S. Candida albicans-Induced NETosis Is Independent of Peptidylarginine Deiminase 4. Front Immunol 2018; 9:1573. [PMID: 30038623 PMCID: PMC6046457 DOI: 10.3389/fimmu.2018.01573] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/25/2018] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are the most abundant innate immune cells and the first line of defense against many pathogenic microbes, including the human fungal pathogen Candida albicans. Among the neutrophils' arsenal of effector functions, neutrophil extracellular traps (NETs) are thought to be of particular importance for trapping and killing the large fungal filaments by means of their web-like structures that consist of chromatin fibers decorated with proteolytic enzymes and host defense proteins. Peptidylarginine deiminase 4 (PAD4)-mediated citrullination of histones in activated neutrophils correlates with chromatin decondensation and extrusion and is widely accepted to act as an integral process of NET induction (NETosis). However, the requirement of PAD4-mediated histone citrullination for NET release during C. albicans infection remains unclear. In this study, we show that although PAD4-dependent neutrophil histone citrullination is readily induced by C. albicans, PAD4 is dispensable for NETosis in response to the fungus and other common NET-inducing stimuli. Moreover, PAD4 is not required for antifungal immunity during mucosal and systemic C. albicans infection. Our results demonstrate that PAD4 is dispensable for C. albicans-induced NETosis, and they highlight the limitations of using histone citrullination as a marker for NETs and PAD4-/- mice as a model of NET-deficiency.
Collapse
Affiliation(s)
- Eva Guiducci
- Section of Immunology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Christina Lemberg
- Section of Immunology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Noëmi Küng
- Section of Immunology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Elisabeth Schraner
- Institute of Virology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
61
|
Yan F, Auerbach D, Chai Y, Keller L, Tu Q, Hüttel S, Glemser A, Grab HA, Bach T, Zhang Y, Müller R. Biosynthesis and Heterologous Production of Vioprolides: Rational Biosynthetic Engineering and Unprecedented 4‐Methylazetidinecarboxylic Acid Formation. Angew Chem Int Ed Engl 2018; 57:8754-8759. [DOI: 10.1002/anie.201802479] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/08/2018] [Indexed: 11/07/2022]
Affiliation(s)
- Fu Yan
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Center for Infection Research, and Department of Pharmacy Saarland University Campus Building E8.1 66123 Saarbrücken Germany
| | - David Auerbach
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Center for Infection Research, and Department of Pharmacy Saarland University Campus Building E8.1 66123 Saarbrücken Germany
| | - Yi Chai
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Center for Infection Research, and Department of Pharmacy Saarland University Campus Building E8.1 66123 Saarbrücken Germany
| | - Lena Keller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Center for Infection Research, and Department of Pharmacy Saarland University Campus Building E8.1 66123 Saarbrücken Germany
| | - Qiang Tu
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Center for Infection Research, and Department of Pharmacy Saarland University Campus Building E8.1 66123 Saarbrücken Germany
| | - Stephan Hüttel
- Helmholtz Centre for Infection Research Department of Microbial Drugs Braunschweig Germany
| | - Amelie Glemser
- Helmholtz Centre for Infection Research Department of Microbial Drugs Braunschweig Germany
| | - Hanusch A. Grab
- Lehrstuhl für Organische Chemie I Technische Universität München Lichtenbergstrasse 4 85747 Garching Germany
| | - Thorsten Bach
- Lehrstuhl für Organische Chemie I Technische Universität München Lichtenbergstrasse 4 85747 Garching Germany
| | - Youming Zhang
- Shandong University-Helmholtz Joint Institute of Biotechnology State Key Laboratory of Microbial Technology School of Life Science Shandong University Qingdao P. R. China
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Center for Infection Research, and Department of Pharmacy Saarland University Campus Building E8.1 66123 Saarbrücken Germany
| |
Collapse
|
62
|
Yan F, Auerbach D, Chai Y, Keller L, Tu Q, Hüttel S, Glemser A, Grab HA, Bach T, Zhang Y, Müller R. Biosynthese und heterologe Expression der Vioprolide: rationale gentechnische Eingriffe in die Biosynthese und 4‐Methylazetidincarbonsäure‐Bildung. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201802479] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Fu Yan
- Helmholtz-Institut für Pharmazeutische Forschung Saarland (HIPS) Helmholtz-Zentrum für Infektionsforschung und Pharmazeutische Biotechnologie Universität des Saarlands Campus Gebäude E8.1 66123 Saarbrücken Deutschland
| | - David Auerbach
- Helmholtz-Institut für Pharmazeutische Forschung Saarland (HIPS) Helmholtz-Zentrum für Infektionsforschung und Pharmazeutische Biotechnologie Universität des Saarlands Campus Gebäude E8.1 66123 Saarbrücken Deutschland
| | - Yi Chai
- Helmholtz-Institut für Pharmazeutische Forschung Saarland (HIPS) Helmholtz-Zentrum für Infektionsforschung und Pharmazeutische Biotechnologie Universität des Saarlands Campus Gebäude E8.1 66123 Saarbrücken Deutschland
| | - Lena Keller
- Helmholtz-Institut für Pharmazeutische Forschung Saarland (HIPS) Helmholtz-Zentrum für Infektionsforschung und Pharmazeutische Biotechnologie Universität des Saarlands Campus Gebäude E8.1 66123 Saarbrücken Deutschland
| | - Qiang Tu
- Helmholtz-Institut für Pharmazeutische Forschung Saarland (HIPS) Helmholtz-Zentrum für Infektionsforschung und Pharmazeutische Biotechnologie Universität des Saarlands Campus Gebäude E8.1 66123 Saarbrücken Deutschland
| | - Stephan Hüttel
- Helmholtz-Zentrum für Infektionsforschung (HZI) Abteilung Mikrobielle Wirkstoffe Braunschweig Deutschland
| | - Amelie Glemser
- Helmholtz-Zentrum für Infektionsforschung (HZI) Abteilung Mikrobielle Wirkstoffe Braunschweig Deutschland
| | - Hanusch A. Grab
- Lehrstuhl für Organische Chemie I Technische Universität München Lichtenbergstraße 4 85747 Garching Deutschland
| | - Thorsten Bach
- Lehrstuhl für Organische Chemie I Technische Universität München Lichtenbergstraße 4 85747 Garching Deutschland
| | - Youming Zhang
- Shandong University-Helmholtz Joint Institute of Biotechnology State Key Laboratory of Microbial Technology School of Life Science Shandong University Qingdao VR China
| | - Rolf Müller
- Helmholtz-Institut für Pharmazeutische Forschung Saarland (HIPS) Helmholtz-Zentrum für Infektionsforschung und Pharmazeutische Biotechnologie Universität des Saarlands Campus Gebäude E8.1 66123 Saarbrücken Deutschland
| |
Collapse
|
63
|
Chen XP, Zheng H, Li WG, Chen GD, Lu JX. Bacteria-induced susceptibility to Candida albicans super-infection in mice via monocyte methyltransferase Setdb2. Cell Microbiol 2018; 20:e12860. [PMID: 29749709 DOI: 10.1111/cmi.12860] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/02/2018] [Accepted: 05/01/2018] [Indexed: 12/22/2022]
Abstract
Systemic bacterial infections are prone to secondary Candida albicans super-infection. However, the molecular mechanisms involved remain poorly understood. In this study, a model comprising sublethal cecal ligation and puncture plus C. albicans intravenous injection was applied to mimic the situation in super-infection. Compared with mice without systemic bacterial infection, mice with systemic bacterial infection had lower antifungal gene expression (including Il1b, Tnf, Il6, Ifnb, Ifng, Cxcl1, and Ccr2) in monocytes and less inflammatory monocytes and neutrophils infiltrating into the kidney when challenged with C. albicans. Further, lentivirus-mediated Setdb2-knockout and overexpression experiments verified that Setdb2 levels in monocytes correlated negatively with antifungal gene expression and survival rates. Transcriptional repression was probably achieved by Setdb2 through H3 methylation at lysine 9 in promoter regions of these antifungal genes.
Collapse
Affiliation(s)
- Xiao-Ping Chen
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Nosocomial Infection, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hao Zheng
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Nosocomial Infection, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wen-Ge Li
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Nosocomial Infection, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Guo-Dong Chen
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China
| | - Jin-Xing Lu
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Nosocomial Infection, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
64
|
Salazar F, Brown GD. Antifungal Innate Immunity: A Perspective from the Last 10 Years. J Innate Immun 2018; 10:373-397. [PMID: 29768268 DOI: 10.1159/000488539] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 03/11/2018] [Indexed: 01/02/2023] Open
Abstract
Fungal pathogens can rarely cause diseases in immunocompetent individuals. However, commensal and normally nonpathogenic environmental fungi can cause life-threatening infections in immunocompromised individuals. Over the last few decades, there has been a huge increase in the incidence of invasive opportunistic fungal infections along with a worrying increase in antifungal drug resistance. As a consequence, research focused on understanding the molecular and cellular basis of antifungal immunity has expanded tremendously in the last few years. This review will provide an overview of the most exciting recent advances in innate antifungal immunity, discoveries that are helping to pave the way for the development of new strategies that are desperately needed to combat these devastating diseases.
Collapse
|
65
|
Tucey TM, Verma J, Harrison PF, Snelgrove SL, Lo TL, Scherer AK, Barugahare AA, Powell DR, Wheeler RT, Hickey MJ, Beilharz TH, Naderer T, Traven A. Glucose Homeostasis Is Important for Immune Cell Viability during Candida Challenge and Host Survival of Systemic Fungal Infection. Cell Metab 2018; 27:988-1006.e7. [PMID: 29719235 PMCID: PMC6709535 DOI: 10.1016/j.cmet.2018.03.019] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/18/2017] [Accepted: 03/26/2018] [Indexed: 12/20/2022]
Abstract
To fight infections, macrophages undergo a metabolic shift whereby increased glycolysis fuels antimicrobial inflammation and killing of pathogens. Here we demonstrate that the pathogen Candida albicans turns this metabolic reprogramming into an Achilles' heel for macrophages. During Candida-macrophage interactions intertwined metabolic shifts occur, with concomitant upregulation of glycolysis in both host and pathogen setting up glucose competition. Candida thrives on multiple carbon sources, but infected macrophages are metabolically trapped in glycolysis and depend on glucose for viability: Candida exploits this limitation by depleting glucose, triggering rapid macrophage death. Using pharmacological or genetic means to modulate glucose metabolism of host and/or pathogen, we show that Candida infection perturbs host glucose homeostasis in the murine candidemia model and demonstrate that glucose supplementation improves host outcomes. Our results support the importance of maintaining glucose homeostasis for immune cell survival during Candida challenge and for host survival in systemic infection.
Collapse
Affiliation(s)
- Timothy M Tucey
- Infection and Immunity Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton 3800, VIC, Australia
| | - Jiyoti Verma
- Infection and Immunity Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton 3800, VIC, Australia
| | - Paul F Harrison
- Bioinformatics Platform, Monash University, Clayton 3800, VIC, Australia
| | - Sarah L Snelgrove
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton 3168, VIC, Australia
| | - Tricia L Lo
- Infection and Immunity Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton 3800, VIC, Australia
| | - Allison K Scherer
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Adele A Barugahare
- Bioinformatics Platform, Monash University, Clayton 3800, VIC, Australia
| | - David R Powell
- Bioinformatics Platform, Monash University, Clayton 3800, VIC, Australia
| | - Robert T Wheeler
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton 3168, VIC, Australia
| | - Traude H Beilharz
- Development and Stem Cells Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton 3800, VIC, Australia
| | - Thomas Naderer
- Infection and Immunity Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton 3800, VIC, Australia.
| | - Ana Traven
- Infection and Immunity Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton 3800, VIC, Australia.
| |
Collapse
|
66
|
Alsina-Beauchamp D, Escós A, Fajardo P, González-Romero D, Díaz-Mora E, Risco A, Martín-Serrano MA, Del Fresno C, Dominguez-Andrés J, Aparicio N, Zur R, Shpiro N, Brown GD, Ardavín C, Netea MG, Alemany S, Sanz-Ezquerro JJ, Cuenda A. Myeloid cell deficiency of p38γ/p38δ protects against candidiasis and regulates antifungal immunity. EMBO Mol Med 2018; 10:e8485. [PMID: 29661910 PMCID: PMC5938613 DOI: 10.15252/emmm.201708485] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 03/12/2018] [Accepted: 03/19/2018] [Indexed: 12/27/2022] Open
Abstract
Candida albicans is a frequent aetiologic agent of sepsis associated with high mortality in immunocompromised patients. Developing new antifungal therapies is a medical need due to the low efficiency and resistance to current antifungal drugs. Here, we show that p38γ and p38δ regulate the innate immune response to C. albicans We describe a new TAK1-TPL2-MKK1-ERK1/2 pathway in macrophages, which is activated by Dectin-1 engagement and positively regulated by p38γ/p38δ. In mice, p38γ/p38δ deficiency protects against C. albicans infection by increasing ROS and iNOS production and thus the antifungal capacity of neutrophils and macrophages, and by decreasing the hyper-inflammation that leads to severe host damage. Leucocyte recruitment to infected kidneys and production of inflammatory mediators are decreased in p38γ/δ-null mice, reducing septic shock. p38γ/p38δ in myeloid cells are critical for this effect. Moreover, pharmacological inhibition of p38γ/p38δ in mice reduces fungal burden, revealing that these p38MAPKs may be therapeutic targets for treating C. albicans infection in humans.
Collapse
Affiliation(s)
| | - Alejandra Escós
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Pilar Fajardo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Diego González-Romero
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Ester Díaz-Mora
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Ana Risco
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | | | - Carlos Del Fresno
- Immunobiology of Inflammation Laboratory Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Jorge Dominguez-Andrés
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Noelia Aparicio
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Rafal Zur
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Natalia Shpiro
- Medical Research Council Protein Phosphorylation Unit, Sir James Black Building, School of Life Sciences, University of Dundee, Dundee, UK
| | - Gordon D Brown
- Aberdeen Fungal Group, Institute of Medical Sciences, Medical Research Council Centre for Medical Mycology at the University of Aberdeen, Aberdeen, UK
| | - Carlos Ardavín
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Susana Alemany
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
| | | | - Ana Cuenda
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| |
Collapse
|
67
|
Stawowczyk M, Naseem S, Montoya V, Baker DP, Konopka J, Reich NC. Pathogenic Effects of IFIT2 and Interferon-β during Fatal Systemic Candida albicans Infection. mBio 2018; 9:e00365-18. [PMID: 29666281 PMCID: PMC5904408 DOI: 10.1128/mbio.00365-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 03/21/2018] [Indexed: 01/01/2023] Open
Abstract
A balanced immune response to infection is essential to prevent the pathology and tissue damage that can occur from an unregulated or hyperactive host defense. Interferons (IFNs) are critical mediators of the innate defense to infection, and in this study we evaluated the contribution of a specific gene coding for IFIT2 induced by type I IFNs in a murine model of disseminated Candida albicans Invasive candidiasis is a frequent challenge during immunosuppression or surgical medical interventions, and C. albicans is a common culprit that leads to high rates of mortality. When IFIT2 knockout mice were infected systemically with C. albicans, they were found to have improved survival and reduced fungal burden compared to wild-type mice. One of the mechanisms by which IFIT2 increases the pathological effects of invasive C. albicans appears to be suppression of NADPH oxidase activation. Loss of IFIT2 increases production of reactive oxygen species by leukocytes, and we demonstrate that IFIT2 is a binding partner of a critical regulatory subunit of NADPH oxidase, p67phox Since the administration of IFN has been used therapeutically to combat viral infections, cancer, and multiple sclerosis, we evaluated administration of IFN-β to mice prior to C. albicans infection. IFN-β treatment promoted pathology and death from C. albicans infection. We provide evidence that IFIT2 increases the pathological effects of invasive C. albicans and that administration of IFN-β has deleterious effects during infection.IMPORTANCE The attributable mortality associated with systemic C. albicans infections in health care settings is significant, with estimates greater than 40%. This life-threatening disease is common in patients with weakened immune systems, either due to disease or as a result of therapies. Type I interferons (IFN) are cytokines of the innate defense response that are used as immune modulators in the treatment of specific cancers, viral infections, and multiple sclerosis. In this study, we show using a murine model that the loss of a specific IFN-stimulated gene coding for IFIT2 improves survival following systemic C. albicans infection. This result infers a harmful effect of IFN during C. albicans infection and is supported by our finding that administration of IFN-β prior to invasive infection promotes fatal pathology. The findings contribute to our understanding of the innate immune response to C. albicans, and they suggest that IFN therapies present a risk factor for disseminated candidiasis.
Collapse
Affiliation(s)
- Marcin Stawowczyk
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York, USA
| | - Shamoon Naseem
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York, USA
| | - Valeria Montoya
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York, USA
| | | | - James Konopka
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York, USA
| | - Nancy C Reich
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
68
|
Hopke A, Brown AJP, Hall RA, Wheeler RT. Dynamic Fungal Cell Wall Architecture in Stress Adaptation and Immune Evasion. Trends Microbiol 2018; 26:284-295. [PMID: 29452950 PMCID: PMC5869159 DOI: 10.1016/j.tim.2018.01.007] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/23/2018] [Accepted: 01/25/2018] [Indexed: 12/28/2022]
Abstract
Deadly infections from opportunistic fungi have risen in frequency, largely because of the at-risk immunocompromised population created by advances in modern medicine and the HIV/AIDS pandemic. This review focuses on dynamics of the fungal polysaccharide cell wall, which plays an outsized role in fungal pathogenesis and therapy because it acts as both an environmental barrier and as the major interface with the host immune system. Human fungal pathogens use architectural strategies to mask epitopes from the host and prevent immune surveillance, and recent work elucidates how biotic and abiotic stresses present during infection can either block or enhance masking. The signaling components implicated in regulating fungal immune recognition can teach us how cell wall dynamics are controlled, and represent potential targets for interventions designed to boost or dampen immunity.
Collapse
Affiliation(s)
- Alex Hopke
- Molecular & Biomedical Sciences, University of Maine, Orono, ME, USA; Current address: BioMEMS Resource Center, Division of Surgery, Innovation and Bioengineering, Department of Surgery, Massachusetts General Hospital, Shriners Burns Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Alistair J P Brown
- MRC Centre for Medical Mycology, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
| | - Rebecca A Hall
- Institute of Microbiology and Infection, and School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Robert T Wheeler
- Molecular & Biomedical Sciences, University of Maine, Orono, ME, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA.
| |
Collapse
|
69
|
Zhong J, Peng L, Wang B, Zhang H, Li S, Yang R, Deng Y, Huang H, Yuan J. Tacrolimus interacts with voriconazole to reduce the severity of fungal keratitis by suppressing IFN-related inflammatory responses and concomitant FK506 and voriconazole treatment suppresses fungal keratitis. Mol Vis 2018; 24. [PMID: 29527115 PMCID: PMC5836723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
PURPOSE To investigate the expression and roles of type I and II interferons (IFNs) in fungal keratitis, as well as the therapeutic effects of tacrolimus (FK506) and voriconazole on this condition. METHODS The mRNA and protein expression levels of type I (IFN-α/β) and II (IFN-γ) IFNs, as well as of related downstream inflammatory cytokines (interleukin (IL)-1α, IL-6, IL-12, and IL-17), were detected in macrophages, neutrophils, lymphocytes, and corneal epithelial cells (A6(1) cells) stimulated with zymosan (10 mg/ml) for 8 or 24 h. A fungal keratitis mouse model was generated through intrastromal injection of Aspergillus fumigatus, and the mice were then divided into four groups: group I, the PBS group; group II, the voriconazole group; group III, the FK506 group; and group IV, the voriconazole plus 0.05% FK506 group. Corneal damage was evaluated with clinical scoring and histological examination. In addition, the mRNA and protein expression levels of type I (IFN-α/β) and type II (IFN-γ) IFNs, as well as related inflammatory cytokines, were determined at different time points using quantitative real-time PCR (qRT-PCR) and western blotting. RESULTS After zymosan stimulation of mouse neutrophils, lymphocytes, macrophages, and A6(1) cells, the IFN mRNA and protein expression levels were markedly increased until 24 h, peaking at 8 h (p<0.001). The mRNA and protein expression levels of inflammatory cytokines (IL-1α, IL-6, IL-12, and IL-17) were also upregulated after zymosan stimulation. Moreover, type I (IFN-α/β) and type II (IFN-γ) IFN expression levels were increased and positively correlated with the progression of fungal keratitis in vivo. FK506 administered with voriconazole reduced the pathological infiltration of inflammatory cells into the cornea and downregulated the expression levels of IFNs and related inflammatory cytokines. CONCLUSIONS In conclusion, this study demonstrated that type I and II IFN levels were markedly increased in fungal keratitis and that FK506 combined with voriconazole decreased the severity of fungal keratitis by suppressing type I and II IFNs and their related inflammatory responses.
Collapse
|
70
|
Schleicher U, Liese J, Justies N, Mischke T, Haeberlein S, Sebald H, Kalinke U, Weiss S, Bogdan C. Type I Interferon Signaling Is Required for CpG-Oligodesoxynucleotide-Induced Control of Leishmania major, but Not for Spontaneous Cure of Subcutaneous Primary or Secondary L. major Infection. Front Immunol 2018; 9:79. [PMID: 29459858 PMCID: PMC5807663 DOI: 10.3389/fimmu.2018.00079] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/11/2018] [Indexed: 01/11/2023] Open
Abstract
We previously showed that in mice infected with Leishmania major type I interferons (IFNs) initiate the innate immune response to the parasite at day 1 and 2 of infection. Here, we investigated which type I IFN subtypes are expressed during the first 8 weeks of L. major infection and whether type I IFNs are essential for a protective immune response and clinical cure of the disease. In self-healing C57BL/6 mice infected with a high dose of L. major, IFN-α4, IFN-α5, IFN-α11, IFN-α13, and IFN-β mRNA were most prominently regulated during the course of infection. In C57BL/6 mice deficient for IFN-β or the IFN-α/β-receptor chain 1 (IFNAR1), development of skin lesions and parasite loads in skin, draining lymph node, and spleen was indistinguishable from wild-type (WT) mice. In line with the clinical findings, C57BL/6 IFN-β−/−, IFNAR1−/−, and WT mice exhibited similar mRNA expression levels of IFN-γ, interleukin (IL)-4, IL-12, IL-13, inducible nitric oxide synthase, and arginase 1 during the acute and late phase of the infection. Also, myeloid dendritic cells from WT and IFNAR1−/− mice produced comparable amounts of IL-12p40/p70 protein upon exposure to L. major in vitro. In non-healing BALB/c WT mice, the mRNAs of IFN-α subtypes (α2, α4, α5, α6, and α9) were rapidly induced after high-dose L. major infection. However, genetic deletion of IFNAR1 or IFN-β did not alter the progressive course of infection seen in WT BALB/c mice. Finally, we tested whether type I IFNs and/or IL-12 are required for the prophylactic effect of CpG-oligodesoxynucleotides (ODN) in BALB/c mice. Local and systemic administration of CpG-ODN 1668 protected WT and IFN-β−/− mice equally well from progressive leishmaniasis. By contrast, the protective effect of CpG-ODN 1668 was lost in BALB/c IFNAR1−/− (despite a sustained suppression of IL-4) and in BALB/c IL-12p35−/− mice. From these data, we conclude that IFN-β and IFNAR1 signaling are dispensable for a curative immune response to L. major in C57BL/6 mice and irrelevant for disease development in BALB/c mice, whereas IL-12 and IFN-α subtypes are essential for the disease prevention by CpG-ODNs in this mouse strain.
Collapse
Affiliation(s)
- Ulrike Schleicher
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Jan Liese
- Abteilung Mikrobiologie und Hygiene, Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Nicole Justies
- Abteilung Mikrobiologie und Hygiene, Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Thomas Mischke
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Simone Haeberlein
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Heidi Sebald
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ulrich Kalinke
- Institut für Experimentelle Infektionsforschung, TWINCORE, Zentrum für Experimentelle und Klinische Infektionsforschung, eine Gemeinschaftseinrichtung vom Helmholtz Zentrum für Infektionsforschung und der Medizinischen Hochschule Hannover, Hannover, Germany
| | - Siegfried Weiss
- Abteilung für Molekulare Immunologie, Helmholtz Zentrum für Infektionsforschung, Braunschweig, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
71
|
Modulation of the Fungal-Host Interaction by the Intra-Species Diversity of C. albicans. Pathogens 2018; 7:pathogens7010011. [PMID: 29342100 PMCID: PMC5874737 DOI: 10.3390/pathogens7010011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 01/09/2023] Open
Abstract
The incidence of human infections caused by the opportunistic fungal pathogen Candida albicans is on the rise due to increasing numbers of immunosuppressed patients. The importance of the immune system in preventing overgrowth of the colonizing fungus and thereby limiting infection is well recognized and host protective mechanisms widely investigated. Only recently, it was recognized that the natural diversity in the fungal species could also influence the outcome of the interaction between the fungus and the host. C. albicans strain-specific differences are complex and their regulation at the genomic, genetic, and epigenetic level and by environmental factors is only partially understood. In this review, we provide an overview of the natural diversity of C. albicans and discuss how it impacts host-fungal interactions and thereby affects the balance between commensalism versus disease.
Collapse
|
72
|
Carpino N, Naseem S, Frank DM, Konopka JB. Modulating Host Signaling Pathways to Promote Resistance to Infection by Candida albicans. Front Cell Infect Microbiol 2017; 7:481. [PMID: 29201860 PMCID: PMC5696602 DOI: 10.3389/fcimb.2017.00481] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/06/2017] [Indexed: 12/29/2022] Open
Abstract
Candida albicans is a common human fungal pathogen capable of causing serious systemic infections that can progress to become lethal. Current therapeutic approaches have limited effectiveness, especially once a systemic infection is established, in part due to the lack of an effective immune response. Boosting the immune response to C. albicans has been the goal of immunotherapy, but it has to be done selectively to prevent deleterious hyperinflammation (sepsis). Although an efficient inflammatory response is necessary to fight infection, the typical response to C. albicans results in collateral damage to tissues thereby exacerbating the pathological effects of infection. For this reason, identifying specific ways of modulating the immune system holds promise for development of new improved therapeutic approaches. This review will focus on recent studies that provide insight using mutant strains of mice that are more resistant to bloodstream infection by C. albicans. These mice are deficient in signal transduction proteins including the Jnk1 MAP kinase, the Cbl-b E3 ubiquitin ligase, or the Sts phosphatases. Interestingly, the mutant mice display a different response to C. albicans that results in faster clearance of infection without hyper-inflammation and collateral damage. A common underlying theme between the resistant mouse strains is loss of negative regulatory proteins that are known to restrain activation of cell surface receptor-initiated signaling cascades. Understanding the cellular and molecular mechanisms that promote resistance to C. albicans in mice will help to identify new approaches for improving antifungal therapy.
Collapse
Affiliation(s)
- Nick Carpino
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, United States
| | - Shamoon Naseem
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, United States
| | - David M Frank
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, United States
| | - James B Konopka
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
73
|
Transcriptomic Signatures of Tacaribe Virus-Infected Jamaican Fruit Bats. mSphere 2017; 2:mSphere00245-17. [PMID: 28959737 PMCID: PMC5615131 DOI: 10.1128/msphere.00245-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/07/2017] [Indexed: 12/18/2022] Open
Abstract
As reservoir hosts of viruses associated with human disease, little is known about the interactions between bats and viruses. Using Jamaican fruit bats infected with Tacaribe virus (TCRV) as a model, we characterized the gene expression responses to infection in different tissues and identified pathways involved with the response to infection. This report is the most detailed gene discovery work in the species to date and the first to describe immune gene expression responses in bats during a pathogenic viral infection. Tacaribe virus (TCRV) is a mammalian arenavirus that was first isolated from artibeus bats in the 1950s. Subsequent experimental infection of Jamaican fruit bats (Artibeus jamaicensis) caused a disease similar to that of naturally infected bats. Although substantial attention has focused on bats as reservoir hosts of viruses that cause human disease, little is known about the interactions between bats and their pathogens. We performed a transcriptome-wide study to illuminate the response of Jamaican fruit bats experimentally infected with TCRV. Differential gene expression analysis of multiple tissues revealed global and organ-specific responses associated with innate antiviral responses, including interferon alpha/beta and Toll-like receptor signaling, activation of complement cascades, and cytokine signaling, among others. Genes encoding proteins involved in adaptive immune responses, such as gamma interferon signaling and costimulation of T cells by the CD28 family, were also altered in response to TCRV infection. Immunoglobulin gene expression was also elevated in the spleens of infected bats, including IgG, IgA, and IgE isotypes. These results indicate an active innate and adaptive immune response to TCRV infection occurred but did not prevent fatal disease. This de novo assembly provides a high-throughput data set of the Jamaican fruit bat and its host response to TCRV infection, which remains a valuable tool to understand the molecular signatures involved in antiviral responses in bats. IMPORTANCE As reservoir hosts of viruses associated with human disease, little is known about the interactions between bats and viruses. Using Jamaican fruit bats infected with Tacaribe virus (TCRV) as a model, we characterized the gene expression responses to infection in different tissues and identified pathways involved with the response to infection. This report is the most detailed gene discovery work in the species to date and the first to describe immune gene expression responses in bats during a pathogenic viral infection.
Collapse
|
74
|
Inflammatory Ly6C high Monocytes Protect against Candidiasis through IL-15-Driven NK Cell/Neutrophil Activation. Immunity 2017. [PMID: 28636955 DOI: 10.1016/j.immuni.2017.05.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neutrophils play a crucial role in defense against systemic candidiasis, a disease associated with a high mortality rate in patients receiving immunosuppressive therapy, although the early immune mechanisms that boost the candidacidal activity of neutrophils remain to be defined in depth. Here, we used a murine model of systemic candidiasis to explore the role of inflammatory Ly6Chigh monocytes in NK cell-mediated neutrophil activation during the innate immune response against C. albicans. We found that efficient anti-Candida immunity required a collaborative response between the spleen and kidney, which relied on type I interferon-dependent IL-15 production by spleen inflammatory Ly6Chigh monocytes to drive efficient activation and GM-CSF release by spleen NK cells; this in turn was necessary to boost the Candida killing potential of kidney neutrophils. Our findings unveil a role for IL-15 as a critical mediator in defense against systemic candidiasis and hold promise for the design of IL-15-based antifungal immunotherapies.
Collapse
|
75
|
Armstrong-James D, Brown GD, Netea MG, Zelante T, Gresnigt MS, van de Veerdonk FL, Levitz SM. Immunotherapeutic approaches to treatment of fungal diseases. THE LANCET. INFECTIOUS DISEASES 2017; 17:e393-e402. [PMID: 28774700 DOI: 10.1016/s1473-3099(17)30442-5] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 11/20/2016] [Accepted: 02/09/2017] [Indexed: 12/15/2022]
Abstract
Fungal infections cause morbidity worldwide and are associated with an unacceptably high mortality despite the availability of antifungal drugs. The incidence of mycoses is rising because of the HIV pandemic and because immunomodulatory drugs are increasingly used to treat autoimmune diseases and cancer. New classes of antifungal drugs have only been partly successful in improving the prognosis for patients with fungal infection. Adjunctive host-directed therapy is therefore believed to be the only option to further improve patient outcomes. Recent advances in the understanding of complex interactions between fungi and host have led to the design and exploration of novel therapeutic strategies in cytokine therapy, vaccines, and cellular immunotherapy, each of which might become viable adjuncts to existing antifungal regimens. In this report, we discuss immunotherapeutic approaches-the rationale behind their design, the challenges in their use, and the progress that is so urgently needed to overcome the devastating effect of fungal diseases.
Collapse
Affiliation(s)
- Darius Armstrong-James
- Fungal Pathogens Laboratory, National Heart and Lung Institute, Imperial College London, UK.
| | - Gordon D Brown
- Aberdeen Fungal Group, MRC Centre for Medical Mycology, University of Aberdeen, Aberdeen, UK
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Teresa Zelante
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Mark S Gresnigt
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Frank L van de Veerdonk
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Stuart M Levitz
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
76
|
Abstract
Pathogenic fungi cause a wide range of syndromes in immune-competent and immune-compromised individuals, with life-threatening disease primarily seen in humans with HIV/AIDS and in patients receiving immunosuppressive therapies for cancer, autoimmunity, and end-organ failure. The discovery that specific primary immune deficiencies manifest with fungal infections and the development of animal models of mucosal and invasive mycoses have facilitated insight into fungus-specific recognition, signaling, effector pathways, and adaptive immune responses. Progress in deciphering the molecular and cellular basis of immunity against fungi is guiding preclinical studies into vaccine and immune reconstitution strategies for vulnerable patient groups. Furthermore, recent work has begun to address the role of endogenous fungal communities in human health and disease. In this review, we summarize a contemporary understanding of protective immunity against fungi.
Collapse
Affiliation(s)
- Michail S Lionakis
- Fungal Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Iliyan D Iliev
- Jill Roberts Institute for Research in IBD, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Tobias M Hohl
- Infectious Disease Service, Department of Medicine, and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
77
|
Wu SY, Huang JH, Chen WY, Chan YC, Lin CH, Chen YC, Liu FT, Wu-Hsieh BA. Cell Intrinsic Galectin-3 Attenuates Neutrophil ROS-Dependent Killing of Candida by Modulating CR3 Downstream Syk Activation. Front Immunol 2017; 8:48. [PMID: 28217127 PMCID: PMC5289966 DOI: 10.3389/fimmu.2017.00048] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/12/2017] [Indexed: 12/27/2022] Open
Abstract
Invasive candidiasis is a leading cause of nosocomial bloodstream infection. Neutrophils are the important effector cells in host resistance to candidiasis. To investigate the modulation of neutrophil fungicidal function will advance our knowledge on the control of candidiasis. While recombinant galectin-3 enhances neutrophil phagocytosis of Candida, we found that intracellular galectin-3 downregulates neutrophil fungicidal functions. Co-immunoprecipitation and immunofluorescence staining reveal that cytosolic gal3 physically interacts with Syk in neutrophils after Candida stimulation. Gal3−/− neutrophils have higher level of Syk activation as well as greater abilities to generate reactive oxygen species (ROS) and kill Candida than gal3+/+ cells. While galectin-3 deficiency modulates neutrophil and macrophage activation and the recruitment of monocytes and dendritic cells, the deficiency does not affect the numbers of infiltrating neutrophils or macrophages. Galectin-3 deficiency ameliorates systemic candidiasis by reducing fungal burden, renal pathology, and mortality. Adoptive transfer experiments demonstrate that cell intrinsic galectin-3 negatively regulates neutrophil effector functions against candidiasis. Reducing galectin-3 expression or activity by siRNA or gal3 inhibitor TD139 enhances human neutrophil ROS production. Mice treated with TD139 have enhanced ability to clear the fungus. Our work unravels the mechanism by which galectin-3 regulates Syk-dependent neutrophil fungicidal functions and raises the possibility that blocking gal3 in neutrophils may be a promising therapeutic strategy for treating systemic candidiasis.
Collapse
Affiliation(s)
- Sheng-Yang Wu
- Graduate Institute of Immunology, National Taiwan University College of Medicine , Taipei , Taiwan
| | - Juin-Hua Huang
- Graduate Institute of Immunology, National Taiwan University College of Medicine , Taipei , Taiwan
| | - Wen-Yu Chen
- Graduate Institute of Immunology, National Taiwan University College of Medicine , Taipei , Taiwan
| | - Yi-Chen Chan
- Institute of Biological Chemistry, Academia Sinica , Taipei , Taiwan
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica , Taipei , Taiwan
| | - Yee-Chun Chen
- Department of Internal Medicine, National Taiwan University Hospital , Taipei , Taiwan
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica , Taipei , Taiwan
| | - Betty A Wu-Hsieh
- Graduate Institute of Immunology, National Taiwan University College of Medicine , Taipei , Taiwan
| |
Collapse
|
78
|
Differential Effects of Vitamins A and D on the Transcriptional Landscape of Human Monocytes during Infection. Sci Rep 2017; 7:40599. [PMID: 28094291 PMCID: PMC5240108 DOI: 10.1038/srep40599] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 12/06/2016] [Indexed: 12/16/2022] Open
Abstract
Vitamin A and vitamin D are essential nutrients with a wide range of pleiotropic effects in humans. Beyond their well-documented roles in cellular differentiation, embryogenesis, tissue maintenance and bone/calcium homeostasis, both vitamins have attracted considerable attention due to their association with-immunological traits. Nevertheless, our knowledge of their immunomodulatory potential during infection is restricted to single gene-centric studies, which do not reflect the complexity of immune processes. In the present study, we performed a comprehensive RNA-seq-based approach to define the whole immunomodulatory role of vitamins A and D during infection. Using human monocytes as host cells, we characterized the differential role of both vitamins upon infection with three different pathogens: Aspergillus fumigatus, Candida albicans and Escherichia coli. Both vitamins showed an unexpected ability to counteract the pathogen-induced transcriptional responses. Upon infection, we identified 346 and 176 immune-relevant genes that were regulated by atRA and vitD, respectively. This immunomodulatory activity was dependent on the inflammatory stimulus, allowing us to distinguish regulatory patterns which were specific for each stimulatory setting. Moreover, we explored possible direct and indirect mechanisms of vitamin-mediated regulation of the immune response. Our findings highlight the importance of vitamin-monitoring in critically ill patients. Moreover, our results underpin the potential of atRA and vitD as therapeutic options for anti-inflammatory treatment.
Collapse
|
79
|
Mayer-Barber KD, Yan B. Clash of the Cytokine Titans: counter-regulation of interleukin-1 and type I interferon-mediated inflammatory responses. Cell Mol Immunol 2017; 14:22-35. [PMID: 27264686 PMCID: PMC5214938 DOI: 10.1038/cmi.2016.25] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/25/2016] [Accepted: 04/26/2016] [Indexed: 02/07/2023] Open
Abstract
Over the past decades the notion of 'inflammation' has been extended beyond the original hallmarks of rubor (redness), calor (heat), tumor (swelling) and dolor (pain) described by Celsus. We have gained a more detailed understanding of the cellular players and molecular mediators of inflammation which is now being applied and extended to areas of biomedical research such as cancer, obesity, heart disease, metabolism, auto-inflammatory disorders, autoimmunity and infectious diseases. Innate cytokines are often central components of inflammatory responses. Here, we discuss how the type I interferon and interleukin-1 cytokine pathways represent distinct and specialized categories of inflammatory responses and how these key mediators of inflammation counter-regulate each other.
Collapse
Affiliation(s)
- Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bo Yan
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
80
|
Wheeler ML, Limon JJ, Underhill DM. Immunity to Commensal Fungi: Detente and Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 12:359-385. [PMID: 28068483 DOI: 10.1146/annurev-pathol-052016-100342] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fungi are ubiquitous in our environment, and a healthy immune system is essential to maintain adequate protection from fungal infections. When this protection breaks down, superficial and invasive fungal infections cause diseases that range from irritating to life-threatening. Millions of people worldwide develop invasive infections during their lives, and mortality for these infections often exceeds 50%. Nevertheless, we are normally colonized with many of the same disease-causing fungi (e.g., on the skin or in the gut). Recent research is dramatically expanding our understanding of the mechanisms by which our immune systems interact with these organisms in health and disease. In this review, we discuss what is currently known about where and how the immune system interacts with common fungi.
Collapse
Affiliation(s)
- Matthew L Wheeler
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, and Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California 90048; , ,
| | - Jose J Limon
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, and Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California 90048; , ,
| | - David M Underhill
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, and Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California 90048; , , .,Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| |
Collapse
|
81
|
|
82
|
Rodríguez M, Márquez S, de la Rosa JV, Alonso S, Castrillo A, Sánchez Crespo M, Fernández N. Fungal pattern receptors down-regulate the inflammatory response by a cross-inhibitory mechanism independent of interleukin-10 production. Immunology 2016; 150:184-198. [PMID: 27709605 DOI: 10.1111/imm.12678] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 12/19/2022] Open
Abstract
Cyclic AMP regulatory element binding protein and signal transducer and activator of transcription 3 (STAT3) may control inflammation by several mechanisms, one of the best characterized is the induction of the expression of the anti-inflammatory cytokine interleukin-10 (IL-10). STAT3 also down-regulates the production of pro-inflammatory cytokines induced by immunoreceptor tyrosine-based activation motif (ITAM)-coupled receptors, a mechanism termed cross-inhibition. Because signalling via ITAM-dependent mechanisms is a hallmark of fungal pattern receptors, STAT3 activation might be involved in the cross-inhibition associated with invasive fungal infections. The fungal surrogate zymosan produced the phosphorylation of Y705-STAT3 and the expression of Ifnb1 and Socs3, but did not induce the interferon (IFN)-signature cytokines Cxcl9 and Cxcl10 in bone marrow-derived dendritic cells. Unlike lipopolysaccharide (LPS), zymosan induced IL-10 and phosphorylated Y705-STAT3 to a similar extent in Irf3 and Ifnar1 knockout and wild-type mice. Human dendritic cells showed similar results, although the induction of IFNB1 was less prominent. These results indicate that LPS and zymosan activate STAT3 through different routes. Whereas type I IFN is the main effector of LPS effect, the mechanism involved in Y705-STAT3 phosphorylation by zymosan is more complex, cannot be associated with type I IFN, IL-6 or granulocyte-macrophage colony-stimulating factor, and seems dependent on several factors given that it was partially inhibited by the platelet-activating factor antagonist WEB2086 and high concentrations of COX inhibitors, p38 mitogen-activate protein kinase inhibitors, and blockade of tumour necrosis factor-α function. Altogether, these results indicate that fungal pattern receptors share with other ITAM-coupled receptors the capacity to produce cross-inhibition through a mechanism involving STAT3 and induction of SOCS3 and IL-10, but that cannot be explained through type I IFN signalling.
Collapse
Affiliation(s)
- Mario Rodríguez
- Departamento de Bioquímica y Biología Molecular, y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
| | - Saioa Márquez
- Departamento de Bioquímica y Biología Molecular, y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
| | - Juan Vladimir de la Rosa
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-Universidad Autónoma de Madrid, Madrid, Spain.,Unidad Asociada de Biomedicina CSIC-Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.,Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la ULPGC, Las Palmas de Gran Canaria, Spain
| | - Sara Alonso
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Valladolid, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-Universidad Autónoma de Madrid, Madrid, Spain.,Unidad Asociada de Biomedicina CSIC-Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.,Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la ULPGC, Las Palmas de Gran Canaria, Spain
| | - Mariano Sánchez Crespo
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Valladolid, Spain
| | - Nieves Fernández
- Departamento de Bioquímica y Biología Molecular, y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain.,Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Valladolid, Spain
| |
Collapse
|
83
|
Hebecker B, Vlaic S, Conrad T, Bauer M, Brunke S, Kapitan M, Linde J, Hube B, Jacobsen ID. Dual-species transcriptional profiling during systemic candidiasis reveals organ-specific host-pathogen interactions. Sci Rep 2016; 6:36055. [PMID: 27808111 PMCID: PMC5093689 DOI: 10.1038/srep36055] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/11/2016] [Indexed: 11/15/2022] Open
Abstract
Candida albicans is a common cause of life-threatening fungal bloodstream infections. In the murine model of systemic candidiasis, the kidney is the primary target organ while the fungal load declines over time in liver and spleen. To better understand these organ-specific differences in host-pathogen interaction, we performed gene expression profiling of murine kidney, liver and spleen and determined the fungal transcriptome in liver and kidney. We observed a delayed transcriptional immune response accompanied by late induction of fungal stress response genes in the kidneys. In contrast, early upregulation of the proinflammatory response in the liver was associated with a fungal transcriptome resembling response to phagocytosis, suggesting that phagocytes contribute significantly to fungal control in the liver. Notably, C. albicans hypha-associated genes were upregulated in the absence of visible filamentation in the liver, indicating an uncoupling of gene expression and morphology and a morphology-independent effect by hypha-associated genes in this organ. Consistently, integration of host and pathogen transcriptional data in an inter-species gene regulatory network indicated connections of C. albicans cell wall remodelling and metabolism to the organ-specific immune responses.
Collapse
Affiliation(s)
- Betty Hebecker
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology (Hans Knöll Institute), Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Sebastian Vlaic
- Department of General, Visceral and Vascular Surgery, Experimental Transplantation Surgery, Jena University Hospital, Jena, Germany.,Research Group Systems Biology/Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology (Hans Knöll Institute), Jena, Germany.,Department of Bioinformatics, Friedrich-Schiller-University Jena, Germany
| | - Theresia Conrad
- Research Group Systems Biology/Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology (Hans Knöll Institute), Jena, Germany
| | - Michael Bauer
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Department of Anaesthesiology and Intensive Care Therapy, Jena University Hospital, Jena, Germany
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology (Hans Knöll Institute), Jena, Germany
| | - Mario Kapitan
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology (Hans Knöll Institute), Jena, Germany
| | - Jörg Linde
- Research Group Systems Biology/Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology (Hans Knöll Institute), Jena, Germany
| | - Bernhard Hube
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology (Hans Knöll Institute), Jena, Germany.,Friedrich-Schiller-University, Jena, Germany
| | - Ilse D Jacobsen
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology (Hans Knöll Institute), Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Friedrich-Schiller-University, Jena, Germany
| |
Collapse
|
84
|
Channappanavar R, Fehr AR, Vijay R, Mack M, Zhao J, Meyerholz DK, Perlman S. Dysregulated Type I Interferon and Inflammatory Monocyte-Macrophage Responses Cause Lethal Pneumonia in SARS-CoV-Infected Mice. Cell Host Microbe 2016; 19:181-93. [PMID: 26867177 PMCID: PMC4752723 DOI: 10.1016/j.chom.2016.01.007] [Citation(s) in RCA: 1160] [Impact Index Per Article: 128.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/30/2015] [Accepted: 01/22/2016] [Indexed: 02/08/2023]
Abstract
Highly pathogenic human respiratory coronaviruses cause acute lethal disease characterized by exuberant inflammatory responses and lung damage. However, the factors leading to lung pathology are not well understood. Using mice infected with SARS (severe acute respiratory syndrome)-CoV, we show that robust virus replication accompanied by delayed type I interferon (IFN-I) signaling orchestrates inflammatory responses and lung immunopathology with diminished survival. IFN-I remains detectable until after virus titers peak, but early IFN-I administration ameliorates immunopathology. This delayed IFN-I signaling promotes the accumulation of pathogenic inflammatory monocyte-macrophages (IMMs), resulting in elevated lung cytokine/chemokine levels, vascular leakage, and impaired virus-specific T cell responses. Genetic ablation of the IFN-αβ receptor (IFNAR) or IMM depletion protects mice from lethal infection, without affecting viral load. These results demonstrate that IFN-I and IMM promote lethal SARS-CoV infection and identify IFN-I and IMMs as potential therapeutic targets in patients infected with pathogenic coronavirus and perhaps other respiratory viruses. SARS-CoV causes a lethal respiratory infection in BALB/c mice Robust SARS-CoV replication and delayed IFN-I signaling promote disease IFN-I induces influx of pathogenic inflammatory monocytes and vascular leakage Disease severity is ameliorated in the absence of IFN signaling
Collapse
Affiliation(s)
| | - Anthony R Fehr
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | - Rahul Vijay
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Matthias Mack
- Department of Internal Medicine, University Hospital Regensburg, Regensburg 93042, Germany
| | - Jincun Zhao
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - David K Meyerholz
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Stanley Perlman
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA; Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
85
|
Park SJ, Cho HR, Kwon B. Roles of IL-33 in Resistance and Tolerance to Systemic Candida albicans Infections. Immune Netw 2016; 16:159-64. [PMID: 27340384 PMCID: PMC4917399 DOI: 10.4110/in.2016.16.3.159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/11/2016] [Accepted: 04/12/2016] [Indexed: 12/11/2022] Open
Abstract
IL-33 is a multifunctional cytokine that is released in response to a variety of intrinsic and extrinsic stimuli. The role of IL-33 in Candida albicans infections is just beginning to be revealed. This cytokine has beneficial effects on host defense against systemic C. albicans infections, and it promotes resistance mechanisms by which the immune system eliminates the invading fungal pathogens; and it also elevates host tolerance by reducing the inflammatory response and thereby, potentially, tissue damage. Thus, IL-33 is classified as a cytokine that has evolved functionally to protect the host from damage by pathogens and immunopathology.
Collapse
Affiliation(s)
- Sang Jun Park
- Department of Surgery, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan 44033, Korea.; Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan 44033, Korea
| | - Hong Rae Cho
- Department of Surgery, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan 44033, Korea.; Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan 44033, Korea
| | - Byungsuk Kwon
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan 44033, Korea.; School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| |
Collapse
|
86
|
Mallick EM, Bergeron AC, Jones SK, Newman ZR, Brothers KM, Creton R, Wheeler RT, Bennett RJ. Phenotypic Plasticity Regulates Candida albicans Interactions and Virulence in the Vertebrate Host. Front Microbiol 2016; 7:780. [PMID: 27303374 PMCID: PMC4880793 DOI: 10.3389/fmicb.2016.00780] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/09/2016] [Indexed: 12/18/2022] Open
Abstract
Phenotypic diversity is critical to the lifestyles of many microbial species, enabling rapid responses to changes in environmental conditions. In the human fungal pathogen Candida albicans, cells exhibit heritable switching between two phenotypic states, white and opaque, which yield differences in mating, filamentous growth, and interactions with immune cells in vitro. Here, we address the in vivo virulence properties of the two cell states in a zebrafish model of infection. Multiple attributes were compared including the stability of phenotypic states, filamentation, virulence, dissemination, and phagocytosis by immune cells, and phenotypes equated across three different host temperatures. Importantly, we found that both white and opaque cells could establish a lethal systemic infection. The relative virulence of the two cell types was temperature dependent; virulence was similar at 25°C, but at higher temperatures (30 and 33°C) white cells were significantly more virulent than opaque cells. Despite the difference in virulence, fungal burden, and dissemination were similar between cells in the two states. Additionally, both white and opaque cells exhibited robust filamentation during infection and blocking filamentation resulted in decreased virulence, establishing that this program is critical for pathogenesis in both cell states. Interactions between C. albicans cells and immune cells differed between white and opaque states. Macrophages and neutrophils preferentially phagocytosed white cells over opaque cells in vitro, and neutrophils showed preferential phagocytosis of white cells in vivo. Together, these studies distinguish the properties of white and opaque cells in a vertebrate host, and establish that the two cell types demonstrate both important similarities and key differences during infection.
Collapse
Affiliation(s)
- Emily M Mallick
- Department of Molecular Microbiology and Immunology, Brown University Providence, RI, USA
| | - Audrey C Bergeron
- Department of Molecular and Biomedical Sciences, University of Maine Orono, ME, USA
| | - Stephen K Jones
- Department of Molecular Microbiology and Immunology, Brown University Providence, RI, USA
| | - Zachary R Newman
- Department of Molecular and Biomedical Sciences, University of Maine Orono, ME, USA
| | - Kimberly M Brothers
- Department of Molecular and Biomedical Sciences, University of Maine Orono, ME, USA
| | - Robbert Creton
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University Providence, RI, USA
| | - Robert T Wheeler
- Department of Molecular and Biomedical Sciences, University of Maine Orono, ME, USA
| | - Richard J Bennett
- Department of Molecular Microbiology and Immunology, Brown University Providence, RI, USA
| |
Collapse
|
87
|
Campione E, Gaziano R, Marino D, Orlandi A. Fungistatic activity of all-trans retinoic acid against Aspergillus fumigatus and Candida albicans. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:1551-5. [PMID: 27199548 PMCID: PMC4857806 DOI: 10.2147/dddt.s93985] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Fungal infections are a major complication in hematologic and neoplastic patients causing severe morbidity and mortality. Aspergillus fumigatus and Candida albicans are among the most invasive opportunistic pathogens in immunocompromised patients, and classic antifungal drugs are frequently unsuccessful in these patients. Recent reports hypothesize that the antifungal efficacy of all-trans retinoic acid (ATRA) is mainly related to its strong capacity to stimulate monocyte-mediated immunity, but no consideration was given to its potential direct fungistatic activity. Moreover, ATRA offers the opportunity for systemic therapy. METHODS AND RESULTS We investigated the efficacy of ATRA at different concentrations for its antifungal activity against opportunistic A. fumigatus and C. albicans obtained from clinical samples according to standard protocols. A fungistatic activity of ATRA on A. fumigatus and C. albicans at 0.5-1 mM concentration was documented up to 7 days. CONCLUSION This is the first evidence of a direct and strong fungistatic activity of ATRA against A. fumigatus and C. albicans. The potential adjuvant therapeutic application of ATRA might be useful in the treatment and/or prevention of systemic mycoses in immunocompromised patients. The discovery of a direct fungistatic activity, in association with its reported immunomodulatory properties, makes ATRA an excellent candidate for new combined antifungal strategies for systemic mycoses in immunocompromised and cancer patients.
Collapse
Affiliation(s)
- Elena Campione
- Department of Dermatology, University of Rome Tor Vergata, Rome, Italy
| | - Roberta Gaziano
- Department of Microbiology, University of Rome Tor Vergata, Rome, Italy
| | - Daniele Marino
- Department of Microbiology, University of Rome Tor Vergata, Rome, Italy
| | - Augusto Orlandi
- Department of Anatomic Pathology, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
88
|
Opportunities for the development of novel therapies based on host-microbial interactions. Pharmacol Res 2016; 112:68-83. [PMID: 27107789 DOI: 10.1016/j.phrs.2016.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/07/2016] [Accepted: 04/08/2016] [Indexed: 12/21/2022]
Abstract
Immune responses are fundamental for protecting against most infectious agents. However, there is now much evidence to suggest that the pathogenesis and tissue damage after infection are not usually related to the direct action of the replication of microorganisms, but instead to altered immune responses triggered after the contact with the pathogen. This review article discusses several mechanisms necessary for the host to protect against microbial infection and focuses in aspects that cause altered inflammation and drive immunopathology. These basic findings can ultimately reveal pathways amenable to host-directed therapy in adjunct to antimicrobial therapy for future improved control measures for many infectious diseases. Therefore, modulating the effects of inflammatory pathways may represent a new therapy during infection outcome and disease.
Collapse
|
89
|
Zheng NX, Wang Y, Hu DD, Yan L, Jiang YY. The role of pattern recognition receptors in the innate recognition of Candida albicans. Virulence 2016; 6:347-61. [PMID: 25714264 DOI: 10.1080/21505594.2015.1014270] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Candida albicans is both a commensal microorganism in healthy individuals and a major fungal pathogen causing high mortality in immunocompromised patients. Yeast-hypha morphological transition is a well known virulence trait of C. albicans. Host innate immunity to C. albicans critically requires pattern recognition receptors (PRRs). In this review, we summarize the PRRs involved in the recognition of C. albicans in epithelial cells, endothelial cells, and phagocytic cells separately. We figure out the differential recognition of yeasts and hyphae, the findings on PRR-deficient mice, and the discoveries on human PRR-related single nucleotide polymorphisms (SNPs).
Collapse
Affiliation(s)
- Nan-Xin Zheng
- a Changzheng Hospital ; Second Military Medical University ; Shanghai , China
| | | | | | | | | |
Collapse
|
90
|
Smeekens SP, van de Veerdonk FL, Netea MG. An Omics Perspective on Candida Infections: Toward Next-Generation Diagnosis and Therapy. Front Microbiol 2016; 7:154. [PMID: 26909070 PMCID: PMC4754423 DOI: 10.3389/fmicb.2016.00154] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 01/29/2016] [Indexed: 12/21/2022] Open
Abstract
Candida species can cause severe infections associated with high morbidity and mortality. Therefore, it is essential to gain more insight into the anti-fungal host defense response. The advent of omics technology and development of advanced systems biology tools has permitted to approach this in an unbiased and quantitative manner. This review summarizes the insights gained on anti-Candida immunity from genetic-, transcriptome-, proteome-, metabolome-, microbiome-, mycobiome-, and computational systems biology studies and discusses practical aspects and future perspectives.
Collapse
Affiliation(s)
- S P Smeekens
- Internal Medicine, Radboud University Medical Center Nijmegen, Netherlands
| | | | - M G Netea
- Internal Medicine, Radboud University Medical Center Nijmegen, Netherlands
| |
Collapse
|
91
|
Khan NS, Kasperkovitz PV, Timmons AK, Mansour MK, Tam JM, Seward MW, Reedy JL, Puranam S, Feliu M, Vyas JM. Dectin-1 Controls TLR9 Trafficking to Phagosomes Containing β-1,3 Glucan. THE JOURNAL OF IMMUNOLOGY 2016; 196:2249-61. [PMID: 26829985 DOI: 10.4049/jimmunol.1401545] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 01/03/2016] [Indexed: 12/23/2022]
Abstract
Dectin-1 and TLR9 play distinct roles in the recognition and induction of innate immune responses to Aspergillus fumigatus and Candida albicans. Dectin-1 is a receptor for the major fungal cell wall carbohydrate β-1,3 glucan that induces inflammatory cytokines and controls phagosomal maturation through spleen tyrosine kinase activation. TLR9 is an endosomal TLR that also modulates the inflammatory cytokine response to fungal pathogens. In this study, we demonstrate that β-1,3 glucan beads are sufficient to induce dynamic redistribution and accumulation of cleaved TLR9 to phagosomes. Trafficking of TLR9 to A. fumigatus and C. albicans phagosomes requires Dectin-1 recognition. Inhibition of phagosomal acidification blocks TLR9 accumulation on phagosomes containing β-1,3 glucan beads. Dectin-1-mediated spleen tyrosine kinase activation is required for TLR9 trafficking to β-1,3 glucan-, A. fumigatus-, and C. albicans-containing phagosomes. In addition, Dectin-1 regulates TLR9-dependent gene expression. Collectively, our study demonstrates that recognition of β-1,3 glucan by Dectin-1 triggers TLR9 trafficking to β-1,3 glucan-containing phagosomes, which may be critical in coordinating innate antifungal defense.
Collapse
Affiliation(s)
- Nida S Khan
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114; Biomedical Engineering and Biotechnology, University of Massachusetts, Lowell, MA 01854
| | | | - Allison K Timmons
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114
| | - Michael K Mansour
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114; Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - Jenny M Tam
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114
| | - Michael W Seward
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114
| | - Jennifer L Reedy
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114; Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - Sravanthi Puranam
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114
| | - Marianela Feliu
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114; Nutrition and Metabolism, Boston University, Boston, MA 02118; and
| | - Jatin M Vyas
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114; Department of Medicine, Harvard Medical School, Boston, MA 02115; Program in Immunology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
92
|
Inhibiting the immunoproteasome exacerbates the pathogenesis of systemic Candida albicans infection in mice. Sci Rep 2016; 6:19434. [PMID: 26776888 PMCID: PMC4726078 DOI: 10.1038/srep19434] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 12/14/2015] [Indexed: 12/01/2022] Open
Abstract
Apart from its role in MHC class I antigen processing, the immunoproteasome has recently been implicated in the modulation of T helper cell differentiation under polarizing conditions in vitro and in the pathogenesis of autoimmune diseases in vivo. In this study, we investigated the influence of LMP7 on T helper cell differentiation in response to the fungus Candida albicans. We observed a strong effect of ONX 0914, an LMP7-selective inhibitor of the immunoproteasome, on IFN-γ and IL-17A production by murine splenocytes and human peripheral blood mononuclear cells (PBMCs) stimulated with C. albicans in vitro. Using a murine model of systemic candidiasis, we could confirm reduced generation of IFN-γ- and IL-17A-producing cells in ONX 0914 treated mice in vivo. Interestingly, ONX 0914 treatment resulted in increased susceptibility to systemic candidiasis, which manifested at very early stages of infection. Mice treated with ONX 0914 showed markedly increased kidney and brain fungal burden which resulted in enhanced neutrophil recruitment and immunopathology. Together, these results strongly suggest a role of the immunoproteasome in promoting proinflammatory T helper cells in response to C. albicans but also in affecting the innate antifungal immunity in a T helper cell-independent manner.
Collapse
|
93
|
Uyangaa E, Kim JH, Patil AM, Choi JY, Kim SB, Eo SK. Distinct Upstream Role of Type I IFN Signaling in Hematopoietic Stem Cell-Derived and Epithelial Resident Cells for Concerted Recruitment of Ly-6Chi Monocytes and NK Cells via CCL2-CCL3 Cascade. PLoS Pathog 2015; 11:e1005256. [PMID: 26618488 PMCID: PMC4664252 DOI: 10.1371/journal.ppat.1005256] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 10/09/2015] [Indexed: 12/24/2022] Open
Abstract
Type I interferon (IFN-I)-dependent orchestrated mobilization of innate cells in inflamed tissues is believed to play a critical role in controlling replication and CNS-invasion of herpes simplex virus (HSV). However, the crucial regulators and cell populations that are affected by IFN-I to establish the early environment of innate cells in HSV-infected mucosal tissues are largely unknown. Here, we found that IFN-I signaling promoted the differentiation of CCL2-producing Ly-6Chi monocytes and IFN-γ/granzyme B-producing NK cells, whereas deficiency of IFN-I signaling induced Ly-6Clo monocytes producing CXCL1 and CXCL2. More interestingly, recruitment of Ly-6Chi monocytes preceded that of NK cells with the levels peaked at 24 h post-infection in IFN-I–dependent manner, which was kinetically associated with the CCL2-CCL3 cascade response. Early Ly-6Chi monocyte recruitment was governed by CCL2 produced from hematopoietic stem cell (HSC)-derived leukocytes, whereas NK cell recruitment predominantly depended on CC chemokines produced by resident epithelial cells. Also, IFN-I signaling in HSC-derived leukocytes appeared to suppress Ly-6Ghi neutrophil recruitment to ameliorate immunopathology. Finally, tissue resident CD11bhiF4/80hi macrophages and CD11chiEpCAM+ dendritic cells appeared to produce initial CCL2 for migration-based self-amplification of early infiltrated Ly-6Chi monocytes upon stimulation by IFN-I produced from infected epithelial cells. Ultimately, these results decipher a detailed IFN-I–dependent pathway that establishes orchestrated mobilization of Ly-6Chi monocytes and NK cells through CCL2-CCL3 cascade response of HSC-derived leukocytes and epithelium-resident cells. Therefore, this cascade response of resident–to-hematopoietic–to-resident cells that drives cytokine–to-chemokine–to-cytokine production to recruit orchestrated innate cells is critical for attenuation of HSV replication in inflamed tissues. Herpes simplex virus type 1 and 2 (HSV-1 and HSV-2) are the most common cause of genital ulceration in humans worldwide with lifelong latent infection after peripheral replication in mucosal tissues. Furthermore, acquisition of human immunodeficiency virus (HIV) is increased in HSV-infected individuals, underscoring the contribution of this virus in facilitating increased susceptibility to other microbial pathogens. Therefore, it is imperative to characterize the host defense to HSV infection and identify key components that regulate virus resistance, in order to devise therapeutic strategy. Although type I interferon (IFN-I)-dependent orchestrated mobilization of innate cells in inflamed tissues is considered a key player to control replication and CNS-invasion of HSV, the regulators and cell population that are affected by IFN-I to establish the orchestrated environment of innate cells in HSV-infected tissues are largely unknown. In the present study, we demonstrate that IFN-I signal governs the sequential recruitment of Ly-6Chi monocytes and then NK cells into mucosal tissues, depending on CCL2-CCL3 cascade mediated by HSC-derived leukocytes and epithelial resident cells, respectively. Also, tissue resident CD11bhiF4/80hi macrophages and CD11chiEpCAM+ dendritic cells were involved in producing the initial CCL2 for migration-based self-amplification of rapidly infiltrated Ly-6Chi monocytes through stimulation by IFN-I produced from infected epithelial cells. This study deciphers detailed IFN-I-dependent pathway that establishes orchestrated mobilization of Ly-6Chi monocytes and NK cells through CCL2-CCL3 cascade.
Collapse
Affiliation(s)
- Erdenebileg Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Republic of Korea
| | - Jin Hyoung Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Republic of Korea
| | - Ajit Mahadev Patil
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Republic of Korea
| | - Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Republic of Korea
| | - Seong Bum Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Republic of Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Republic of Korea
- Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, Republic of Korea
- * E-mail:
| |
Collapse
|
94
|
Tscherner M, Zwolanek F, Jenull S, Sedlazeck FJ, Petryshyn A, Frohner IE, Mavrianos J, Chauhan N, von Haeseler A, Kuchler K. The Candida albicans Histone Acetyltransferase Hat1 Regulates Stress Resistance and Virulence via Distinct Chromatin Assembly Pathways. PLoS Pathog 2015; 11:e1005218. [PMID: 26473952 PMCID: PMC4608838 DOI: 10.1371/journal.ppat.1005218] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/21/2015] [Indexed: 01/14/2023] Open
Abstract
Human fungal pathogens like Candida albicans respond to host immune surveillance by rapidly adapting their transcriptional programs. Chromatin assembly factors are involved in the regulation of stress genes by modulating the histone density at these loci. Here, we report a novel role for the chromatin assembly-associated histone acetyltransferase complex NuB4 in regulating oxidative stress resistance, antifungal drug tolerance and virulence in C. albicans. Strikingly, depletion of the NuB4 catalytic subunit, the histone acetyltransferase Hat1, markedly increases resistance to oxidative stress and tolerance to azole antifungals. Hydrogen peroxide resistance in cells lacking Hat1 results from higher induction rates of oxidative stress gene expression, accompanied by reduced histone density as well as subsequent increased RNA polymerase recruitment. Furthermore, hat1Δ/Δ cells, despite showing growth defects in vitro, display reduced susceptibility to reactive oxygen-mediated killing by innate immune cells. Thus, clearance from infected mice is delayed although cells lacking Hat1 are severely compromised in killing the host. Interestingly, increased oxidative stress resistance and azole tolerance are phenocopied by the loss of histone chaperone complexes CAF-1 and HIR, respectively, suggesting a central role for NuB4 in the delivery of histones destined for chromatin assembly via distinct pathways. Remarkably, the oxidative stress phenotype of hat1Δ/Δ cells is a species-specific trait only found in C. albicans and members of the CTG clade. The reduced azole susceptibility appears to be conserved in a wider range of fungi. Thus, our work demonstrates how highly conserved chromatin assembly pathways can acquire new functions in pathogenic fungi during coevolution with the host. Candida albicans is the most prevalent fungal pathogen infecting humans, causing life-threatening infections in immunocompromised individuals. Host immune surveillance imposes stress conditions upon C. albicans, to which it has to adapt quickly to escape host killing. This can involve regulation of specific genes requiring disassembly and reassembly of histone proteins, around which DNA is wrapped to form the basic repeat unit of eukaryotic chromatin—the nucleosome. Here, we discover a novel function for the chromatin assembly-associated histone acetyltransferase complex NuB4 in oxidative stress response, antifungal drug tolerance as well as in fungal virulence. The NuB4 complex modulates the induction kinetics of hydrogen peroxide-induced genes. Furthermore, NuB4 negatively regulates susceptibility to killing by immune cells and thereby slowing the clearing from infected mice in vivo. Remarkably, the oxidative stress resistance seems restricted to C. albicans and closely related species, which might have acquired this function during coevolution with the host.
Collapse
Affiliation(s)
- Michael Tscherner
- Department for Medical Biochemistry, Medical University of Vienna, Max F. Perutz Laboratories, Campus Vienna Biocenter, Vienna, Austria
| | - Florian Zwolanek
- Department for Medical Biochemistry, Medical University of Vienna, Max F. Perutz Laboratories, Campus Vienna Biocenter, Vienna, Austria
| | - Sabrina Jenull
- Department for Medical Biochemistry, Medical University of Vienna, Max F. Perutz Laboratories, Campus Vienna Biocenter, Vienna, Austria
| | - Fritz J. Sedlazeck
- Center for Integrative Bioinformatics Vienna, Max F. Perutz Laboratories, University of Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Andriy Petryshyn
- Department for Medical Biochemistry, Medical University of Vienna, Max F. Perutz Laboratories, Campus Vienna Biocenter, Vienna, Austria
| | - Ingrid E. Frohner
- Department for Medical Biochemistry, Medical University of Vienna, Max F. Perutz Laboratories, Campus Vienna Biocenter, Vienna, Austria
| | - John Mavrianos
- Public Health Research Institute, New Jersey Medical School - Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
| | - Neeraj Chauhan
- Public Health Research Institute, New Jersey Medical School - Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
| | - Arndt von Haeseler
- Center for Integrative Bioinformatics Vienna, Max F. Perutz Laboratories, University of Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Karl Kuchler
- Department for Medical Biochemistry, Medical University of Vienna, Max F. Perutz Laboratories, Campus Vienna Biocenter, Vienna, Austria
- * E-mail:
| |
Collapse
|
95
|
Netea MG, Joosten LAB, van der Meer JWM, Kullberg BJ, van de Veerdonk FL. Immune defence against Candida fungal infections. Nat Rev Immunol 2015; 15:630-42. [DOI: 10.1038/nri3897] [Citation(s) in RCA: 325] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
96
|
Buechler MB, Gessay GM, Srivastava S, Campbell DJ, Hamerman JA. Hematopoietic and nonhematopoietic cells promote Type I interferon- and TLR7-dependent monocytosis during low-dose LCMV infection. Eur J Immunol 2015; 45:3064-72. [PMID: 26289159 DOI: 10.1002/eji.201445331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 07/27/2015] [Accepted: 08/14/2015] [Indexed: 12/24/2022]
Abstract
Release of inflammatory monocytes from the bone marrow (BM) into the blood is an important physiological response to infection, but the mechanisms regulating this phenomenon during viral infection are not completely defined. Here, we show that low-dose infection with lymphocytic choriomeningitis virus (LCMV) caused rapid, transient inflammatory monocytosis that required type I interferon (IFN) and Toll-like receptor (TLR) 7 signaling. Type I IFN and TLR7 signals were critical for induction of IFN-stimulated gene expression and CCR2 ligand upregulation in the BM microenvironment in response to LCMV infection. Experiments utilizing BM chimeric mice demonstrated that type I IFN and TLR7 signaling on either hematopoietic or nonhematopoietic cells was sufficient to initiate monocytosis in response to LCMV infection. BM plasmacytoid dendritic cells (pDCs) generated type I IFN directly ex vivo, suggesting that pDCs are a hematopoietic contributor of type I IFN in the BM early during LCMV infection. Overall, we describe novel roles for type I IFN and TLR7 signaling in nonhematopoietic cells and BM pDCs in directing IFN-stimulated gene and CCR2 ligand expression in the BM to initiate an increase in blood inflammatory monocytes during viral infection.
Collapse
Affiliation(s)
- Matthew B Buechler
- Department of Immunology, University of Washington, Seattle, WA, USA.,Immunology Research Program, Benaroya Research Institute (BRI) at Virginia Mason, Seattle, WA, USA
| | - Griffin M Gessay
- Immunology Research Program, Benaroya Research Institute (BRI) at Virginia Mason, Seattle, WA, USA
| | - Shivani Srivastava
- Department of Immunology, University of Washington, Seattle, WA, USA.,Immunology Research Program, Benaroya Research Institute (BRI) at Virginia Mason, Seattle, WA, USA
| | - Daniel J Campbell
- Department of Immunology, University of Washington, Seattle, WA, USA.,Immunology Research Program, Benaroya Research Institute (BRI) at Virginia Mason, Seattle, WA, USA
| | - Jessica A Hamerman
- Department of Immunology, University of Washington, Seattle, WA, USA.,Immunology Research Program, Benaroya Research Institute (BRI) at Virginia Mason, Seattle, WA, USA
| |
Collapse
|
97
|
Sionov E, Mayer-Barber KD, Chang YC, Kauffman KD, Eckhaus MA, Salazar AM, Barber DL, Kwon-Chung KJ. Type I IFN Induction via Poly-ICLC Protects Mice against Cryptococcosis. PLoS Pathog 2015; 11:e1005040. [PMID: 26252005 PMCID: PMC4529209 DOI: 10.1371/journal.ppat.1005040] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/22/2015] [Indexed: 01/10/2023] Open
Abstract
Cryptococcus neoformans is the most common cause of fungal meningoencephalitis in AIDS patients. Depletion of CD4 cells, such as occurs during advanced AIDS, is known to be a critical risk factor for developing cryptococcosis. However, the role of HIV-induced innate inflammation in susceptibility to cryptococcosis has not been evaluated. Thus, we sought to determine the role of Type I IFN induction in host defense against cryptococci by treatment of C. neoformans (H99) infected mice with poly-ICLC (pICLC), a dsRNA virus mimic. Unexpectedly, pICLC treatment greatly extended survival of infected mice and reduced fungal burdens in the brain. Protection from cryptococcosis by pICLC-induced Type I IFN was mediated by MDA5 rather than TLR3. PICLC treatment induced a large, rapid and sustained influx of neutrophils and Ly6Chigh monocytes into the lung while suppressing the development of eosinophilia. The pICLC-mediated protection against H99 was CD4 T cell dependent and analysis of CD4 T cell polyfunctionality showed a reduction in IL-5 producing CD4 T cells, marginal increases in Th1 cells and dramatic increases in RORγt+ Th17 cells in pICLC treated mice. Moreover, the protective effect of pICLC against H99 was diminished in IFNγ KO mice and by IL-17A neutralization with blocking mAbs. Furthermore, pICLC treatment also significantly extended survival of C. gattii infected mice with reduced fungal loads in the lungs. These data demonstrate that induction of type I IFN dramatically improves host resistance against the etiologic agents of cryptococcosis by beneficial alterations in both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Edward Sionov
- Molecular Microbiology Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Katrin D Mayer-Barber
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Yun C Chang
- Molecular Microbiology Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Keith D Kauffman
- T-Lymphocyte Biology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Michael A Eckhaus
- Division of Veterinary Resources, Office of Research Services, Office of the Director, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | | | - Daniel L Barber
- T-Lymphocyte Biology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Kyung J Kwon-Chung
- Molecular Microbiology Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| |
Collapse
|
98
|
Jiang TT, Chaturvedi V, Ertelt JM, Xin L, Clark DR, Kinder JM, Way SS. Commensal enteric bacteria lipopolysaccharide impairs host defense against disseminated Candida albicans fungal infection. Mucosal Immunol 2015; 8:886-95. [PMID: 25492473 PMCID: PMC4465067 DOI: 10.1038/mi.2014.119] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/30/2014] [Indexed: 02/04/2023]
Abstract
Commensal enteric bacteria maintain systemic immune responsiveness that protects against disseminated or localized infection in extra-intestinal tissues caused by pathogenic microbes. However, as shifts in infection susceptibility after commensal bacteria eradication have primarily been probed using viruses, the broader applicability to other pathogen types remains undefined. In sharp contrast to diminished antiviral immunity, we show commensal bacteria eradication bolsters protection against disseminated Candida albicans fungal infection. Enhanced antifungal immunity reflects more robust systemic expansion of Ly6G(hi)Ly6C(int) neutrophils, and their mobilization into infected tissues among antibiotic-treated compared with commensal bacteria-replete control mice. Reciprocally, depletion of neutrophils from expanded levels or intestinal lipopolysaccharide reconstitution overrides the antifungal protective benefits conferred by commensal bacteria eradication. This discordance in antifungal compared with antiviral immunity highlights intrinsic differences in how commensal bacteria control responsiveness for specific immune cell subsets, because pathogen-specific CD8(+) T cells that protect against viruses were suppressed similarly after C. albicans and influenza A virus infection. Thus, positive calibration of antiviral immunity by commensal bacteria is counterbalanced by restrained activation of other immune components that confer antifungal immunity.
Collapse
Affiliation(s)
- Tony T. Jiang
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA,Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Vandana Chaturvedi
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - James M. Ertelt
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Lijun Xin
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Dayna R. Clark
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jeremy M. Kinder
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sing Sing Way
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA,for correspondence, Dr. Sing Sing Way 3333 Burnet Avenue, MLC 7017 Cincinnati, OH 45229, USA Phone, 513-636-7603 Fax, 513-636-7655
| |
Collapse
|
99
|
Stifter SA, Feng CG. Interfering with immunity: detrimental role of type I IFNs during infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:2455-65. [PMID: 25747907 DOI: 10.4049/jimmunol.1402794] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Type I IFNs are known to inhibit viral replication and mediate protection against viral infection. However, recent studies revealed that these cytokines play a broader and more fundamental role in host responses to infections beyond their well-established antiviral function. Type I IFN induction, often associated with microbial evasion mechanisms unique to virulent microorganisms, is now shown to increase host susceptibility to a diverse range of pathogens, including some viruses. This article presents an overview of the role of type I IFNs in infections with bacterial, fungal, parasitic, and viral pathogens and discusses the key mechanisms mediating the regulatory function of type I IFNs in pathogen clearance and tissue inflammation.
Collapse
Affiliation(s)
- Sebastian A Stifter
- Immunology and Host Defense Group, Department of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney 2006, New South Wales, Australia; and Mycobacterial Research Program, Centenary Institute, Sydney 2050, New South Wales, Australia
| | - Carl G Feng
- Immunology and Host Defense Group, Department of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney 2006, New South Wales, Australia; and Mycobacterial Research Program, Centenary Institute, Sydney 2050, New South Wales, Australia
| |
Collapse
|
100
|
Goritzka M, Makris S, Kausar F, Durant LR, Pereira C, Kumagai Y, Culley FJ, Mack M, Akira S, Johansson C. Alveolar macrophage-derived type I interferons orchestrate innate immunity to RSV through recruitment of antiviral monocytes. ACTA ACUST UNITED AC 2015; 212:699-714. [PMID: 25897172 PMCID: PMC4419339 DOI: 10.1084/jem.20140825] [Citation(s) in RCA: 219] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 03/24/2015] [Indexed: 12/24/2022]
Abstract
Goritzka et al. describe a role for recruited inflammatory monocytes in antiviral immunity and protection from RSV infection in mice. The authors demonstrate that this is critically dependent on the production of type I IFNs by alveolar macrophages triggered via RIG-I–like receptors, thus highlighting an important cell-extrinsic mechanism of type I IFN–mediated antiviral activity. Type I interferons (IFNs) are important for host defense from viral infections, acting to restrict viral production in infected cells and to promote antiviral immune responses. However, the type I IFN system has also been associated with severe lung inflammatory disease in response to respiratory syncytial virus (RSV). Which cells produce type I IFNs upon RSV infection and how this directs immune responses to the virus, and potentially results in pathological inflammation, is unclear. Here, we show that alveolar macrophages (AMs) are the major source of type I IFNs upon RSV infection in mice. AMs detect RSV via mitochondrial antiviral signaling protein (MAVS)–coupled retinoic acid–inducible gene 1 (RIG-I)–like receptors (RLRs), and loss of MAVS greatly compromises innate immune restriction of RSV. This is largely attributable to loss of type I IFN–dependent induction of monocyte chemoattractants and subsequent reduced recruitment of inflammatory monocytes (infMo) to the lungs. Notably, the latter have potent antiviral activity and are essential to control infection and lessen disease severity. Thus, infMo recruitment constitutes an important and hitherto underappreciated, cell-extrinsic mechanism of type I IFN–mediated antiviral activity. Dysregulation of this system of host antiviral defense may underlie the development of RSV-induced severe lung inflammation.
Collapse
Affiliation(s)
- Michelle Goritzka
- Centre for Respiratory Infection, Respiratory Infections Section, National Heart and Lung Institute, Imperial College London, London W2 1PG, England, UK
| | - Spyridon Makris
- Centre for Respiratory Infection, Respiratory Infections Section, National Heart and Lung Institute, Imperial College London, London W2 1PG, England, UK
| | - Fahima Kausar
- Centre for Respiratory Infection, Respiratory Infections Section, National Heart and Lung Institute, Imperial College London, London W2 1PG, England, UK
| | - Lydia R Durant
- Centre for Respiratory Infection, Respiratory Infections Section, National Heart and Lung Institute, Imperial College London, London W2 1PG, England, UK
| | - Catherine Pereira
- Centre for Respiratory Infection, Respiratory Infections Section, National Heart and Lung Institute, Imperial College London, London W2 1PG, England, UK
| | - Yutaro Kumagai
- Laboratory of Host Defense, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Fiona J Culley
- Centre for Respiratory Infection, Respiratory Infections Section, National Heart and Lung Institute, Imperial College London, London W2 1PG, England, UK
| | - Matthias Mack
- University Hospital Regensburg, 93042 Regensburg, Germany
| | - Shizuo Akira
- Laboratory of Host Defense, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Cecilia Johansson
- Centre for Respiratory Infection, Respiratory Infections Section, National Heart and Lung Institute, Imperial College London, London W2 1PG, England, UK
| |
Collapse
|