1
|
Qi WH, Hu LF, Gu YJ, Zhang XY, Jiang XM, Li WJ, Qi JS, Xiao GS, Jie H. Integrated mRNA-miRNA transcriptome profiling of blood immune responses potentially related to pulmonary fibrosis in forest musk deer. Front Immunol 2024; 15:1404108. [PMID: 38873601 PMCID: PMC11169664 DOI: 10.3389/fimmu.2024.1404108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/18/2024] [Indexed: 06/15/2024] Open
Abstract
Background Forest musk deer (FMD, Moschus Berezovskii) is a critically endangered species world-widely, the death of which can be caused by pulmonary disease in the farm. Pulmonary fibrosis (PF) was a huge threat to the health and survival of captive FMD. MicroRNAs (miRNAs) and messenger RNAs (mRNAs) have been involved in the regulation of immune genes and disease development. However, the regulatory profiles of mRNAs and miRNAs involved in immune regulation of FMD are unclear. Methods In this study, mRNA-seq and miRNA-seq in blood were performed to constructed coexpression regulatory networks between PF and healthy groups of FMD. The hub immune- and apoptosis-related genes in the PF blood of FMD were explored through Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Further, protein-protein interaction (PPI) network of immune-associated and apoptosis-associated key signaling pathways were constructed based on mRNA-miRNA in the PF blood of the FMD. Immune hub DEGs and immune hub DEmiRNAs were selected for experimental verification using RT-qPCR. Results A total of 2744 differentially expressed genes (DEGs) and 356 differentially expressed miRNAs (DEmiRNAs) were identified in the PF blood group compared to the healthy blood group. Among them, 42 DEmiRNAs were negatively correlated with 20 immune DEGs from a total of 57 correlations. The DEGs were significantly associated with pathways related to CD molecules, immune disease, immune system, cytokine receptors, T cell receptor signaling pathway, Th1 and Th2 cell differentiation, cytokine-cytokine receptor interaction, intestinal immune network for IgA production, and NOD-like receptor signaling pathway. There were 240 immune-related DEGs, in which 186 immune-related DEGs were up-regulated and 54 immune-related DEGs were down-regulated. In the protein-protein interaction (PPI) analysis of immune-related signaling pathway, TYK2, TLR2, TLR4, IL18, CSF1, CXCL13, LCK, ITGB2, PIK3CB, HCK, CD40, CD86, CCL3, CCR7, IL2RA, TLR3, and IL4R were identified as the hub immune genes. The mRNA-miRNA coregulation analysis showed that let-7d, miR-324-3p, miR-760, miR-185, miR-149, miR-149-5p, and miR-1842-5p are key miRNAs that target DEGs involved in immune disease, immune system and immunoregulation. Conclusion The development and occurrence of PF were significantly influenced by the immune-related and apoptosis-related genes present in PF blood. mRNAs and miRNAs associated with the development and occurrence of PF in the FMD.
Collapse
Affiliation(s)
- Wen-Hua Qi
- College of Biological and Food Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Li-Fan Hu
- College of Environmental and Chemical Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Yu-Jiawei Gu
- College of Biological and Food Engineering, Chongqing Three Gorges University, Chongqing, China
| | | | - Xue-Mei Jiang
- College of Biological and Food Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Wu-Jiao Li
- Department of Laboratory Medicine, Shenzhen Children’s Hospital, Shenzhen, China
| | - Jun-Sheng Qi
- College of Biological and Food Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Guo-Sheng Xiao
- College of Biological and Food Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Hang Jie
- Jinfo Mountain Forest Ecosystem Field Scientific Observation and Research Station of Chongqing, Chongqing Institute of Medicinal Plant Cultivation, Chongqing, China
| |
Collapse
|
2
|
Kembou-Ringert JE, Steinhagen D, Thompson KD, Daly JM, Adamek M. Immune responses to Tilapia lake virus infection: what we know and what we don't know. Front Immunol 2023; 14:1240094. [PMID: 37622112 PMCID: PMC10445761 DOI: 10.3389/fimmu.2023.1240094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/20/2023] [Indexed: 08/26/2023] Open
Abstract
Tilapia lake virus (TiLV) is a novel contagious pathogen associated with a lethal disease affecting and decimating tilapia populations on several continents across the globe. Fish viral diseases, such as Tilapia lake virus disease (TiLVD), represent a serious threat to tilapia aquaculture. Therefore, a better understanding of the innate immune responses involved in establishing an antiviral state can help shed light on TiLV disease pathogenesis. Moreover, understanding the adaptive immune mechanisms involved in mounting protection against TiLV could greatly assist in the development of vaccination strategies aimed at controlling TiLVD. This review summarizes the current state of knowledge on the immune responses following TiLV infection. After describing the main pathological findings associated with TiLVD, both the innate and adaptive immune responses and mechanisms to TiLV infection are discussed, in both disease infection models and in vitro studies. In addition, our work, highlights research questions, knowledge gaps and research areas in the immunology of TiLV infection where further studies are needed to better understand how disease protection against TiLV is established.
Collapse
Affiliation(s)
- Japhette E. Kembou-Ringert
- Department of Infection, Immunity and Inflammation, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Dieter Steinhagen
- Fish Disease Research Unit, Institute for Parasitology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Kim D. Thompson
- Moredun Research Institute, Pentlands Science Park, Penicuik, United Kingdom
| | - Janet M. Daly
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, United Kingdom
| | - Mikolaj Adamek
- Fish Disease Research Unit, Institute for Parasitology, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
3
|
Resiliac J, Rohlfing M, Santoro J, Hussain SRA, Grayson MH. Low-Dose Lipopolysaccharide Protects from Lethal Paramyxovirus Infection in a Macrophage- and TLR4-Dependent Process. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:348-355. [PMID: 36480273 PMCID: PMC9851983 DOI: 10.4049/jimmunol.2200604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/14/2022] [Indexed: 01/03/2023]
Abstract
Respiratory diseases are a major public health burden and a leading cause of death and disability in the world. Understanding antiviral immune responses is crucial to alleviate morbidity and mortality associated with these respiratory viral infections. Previous data from human and animal studies suggested that pre-existing atopy may provide some protection against severe disease from a respiratory viral infection. However, the mechanism(s) of protection is not understood. Low-dose LPS has been shown to drive an atopic phenotype in mice. In addition, LPS has been shown in vitro to have an antiviral effect. We examined the effect of LPS treatment on mortality to the murine parainfluenza virus Sendai virus. Low-dose LPS treatment 24 h before inoculation with a normally lethal dose of Sendai virus greatly reduced death. This protection was associated with a reduced viral titer and reduced inflammatory cytokine production in the airways. The administration of LPS was associated with a marked increase in lung neutrophils and macrophages. Depletion of neutrophils failed to reverse the protective effect of LPS; however, depletion of macrophages reversed the protective effect of LPS. Further, we demonstrate that the protective effect of LPS depends on type I IFN and TLR4-MyD88 signaling. Together, these studies demonstrate pretreatment with low-dose LPS provides a survival advantage against a severe respiratory viral infection through a macrophage-, TLR4-, and MyD88-dependent pathway.
Collapse
Affiliation(s)
- Jenny Resiliac
- The Ohio State University College of Medicine, Biomedical Sciences Graduate Program, Columbus, Ohio
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Michelle Rohlfing
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Jennifer Santoro
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Syed-Rehan A. Hussain
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Mitchell H. Grayson
- Abigail Wexner Research Institute at Nationwide Children’s Hospital, Center for Clinical and Translational Research, Columbus, Ohio
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children’s Hospital and The Ohio State University, Columbus, OH
| |
Collapse
|
4
|
Manna S, McAuley J, Jacobson J, Nguyen CD, Ullah MA, Sebina I, Williamson V, Mulholland EK, Wijburg O, Phipps S, Satzke C. Synergism and Antagonism of Bacterial-Viral Coinfection in the Upper Respiratory Tract. mSphere 2022; 7:e0098421. [PMID: 35044807 PMCID: PMC8769199 DOI: 10.1128/msphere.00984-21] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 12/28/2021] [Indexed: 01/03/2023] Open
Abstract
Streptococcus pneumoniae (the pneumococcus) is a leading cause of pneumonia in children under 5 years of age. Coinfection by pneumococci and respiratory viruses enhances disease severity. Little is known about pneumococcal coinfections with respiratory syncytial virus (RSV). Here, we developed a novel infant mouse model of coinfection using pneumonia virus of mice (PVM), a murine analogue of RSV, to examine the dynamics of coinfection in the upper respiratory tract, an anatomical niche that is essential for host-to-host transmission and progression to disease. Coinfection increased damage to the nasal tissue and increased production of the chemokine CCL3. Nasopharyngeal pneumococcal density and shedding in nasal secretions were increased by coinfection. In contrast, coinfection reduced PVM loads in the nasopharynx, an effect that was independent of pneumococcal strain and the order of infection. We showed that this "antagonistic" effect was absent using either ethanol-killed pneumococci or a pneumococcal mutant deficient in capsule production and incapable of nasopharyngeal carriage. Colonization with a pneumococcal strain naturally unable to produce capsule also reduced viral loads. The pneumococcus-mediated reduction in PVM loads was caused by accelerated viral clearance from the nasopharynx. Although these synergistic and antagonistic effects occurred with both wild-type pneumococcal strains used in this study, the magnitude of the effects was strain dependent. Lastly, we showed that pneumococci can also antagonize influenza virus. Taken together, our study has uncovered multiple novel facets of bacterial-viral coinfection. Our findings have important public health implications, including for bacterial and viral vaccination strategies in young children. IMPORTANCE Respiratory bacterial-viral coinfections (such as pneumococci and influenza virus) are often synergistic, resulting in enhanced disease severity. Although colonization of the nasopharynx is the precursor to disease and transmission, little is known about bacterial-viral interactions that occur within this niche. In this study, we developed a novel mouse model to examine pneumococcal-viral interactions in the nasopharynx with pneumonia virus of mice (PVM) and influenza. We found that PVM infection benefits pneumococci by increasing their numbers in the nasopharynx and shedding of these bacteria in respiratory secretions. In contrast, we discovered that pneumococci decrease PVM numbers by accelerating viral clearance. We also report a similar effect of pneumococci on influenza. By showing that coinfections lead to both synergistic and antagonistic outcomes, our findings challenge the existing dogma in the field. Our work has important applications and implications for bacterial and viral vaccines that target these microbes.
Collapse
Affiliation(s)
- Sam Manna
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Julie McAuley
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jonathan Jacobson
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Cattram D. Nguyen
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Md. Ashik Ullah
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Ismail Sebina
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Victoria Williamson
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - E. Kim Mulholland
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Odilia Wijburg
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Simon Phipps
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Catherine Satzke
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
5
|
Karkossa I, Bannuscher A, Hellack B, Wohlleben W, Laloy J, Stan MS, Dinischiotu A, Wiemann M, Luch A, Haase A, von Bergen M, Schubert K. Nanomaterials induce different levels of oxidative stress, depending on the used model system: Comparison of in vitro and in vivo effects. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 801:149538. [PMID: 34428663 DOI: 10.1016/j.scitotenv.2021.149538] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 06/13/2023]
Abstract
The immense diversity and constant development of nanomaterials (NMs) increase the need for a facilitated risk assessment, which requires knowledge of the modes of action (MoAs) of NMs. This necessitates a comprehensive data basis, which can be obtained using omics. Furthermore, the establishment of suitable in vitro test systems is essential to follow the 3R concept and to cope with the high number of NMs. In the present study, we aimed to compare NM effects in vitro and in vivo using a multi-omics approach. We applied an integrated data analysis strategy based on proteomics and metabolomics to four silica NMs and one titanium dioxide-based NM. For the in vitro investigations, rat alveolar epithelial cells (RLE-6TN) and rat alveolar macrophages (NR8383) were treated with different doses of NMs, and the results were compared with the effects on rat lungs after short-term inhalations and instillations. Since reactive oxygen species (ROS) production has been described as a critical biological effect of NMs, we focused on different levels of oxidative stress. Thus, we found opposite changes in proteins and metabolites related to the production of reduced glutathione in alveolar epithelial cells and alveolar macrophages, demonstrating that the MoAs of NMs depend on the model system used. Interestingly, in vivo, pathways related to inflammation were more affected than oxidative stress responses. Hence, the assignment of the observed effects to levels of oxidative stress was also different in vitro and in vivo. However, the overall classification of "active" and "passive" NMs was consistent in vitro and in vivo, suggesting that both cell lines tested are suitable for the assessment of NM toxicity. In summary, the results presented here highlight the need to carefully review model systems to decipher the extent to which they can replace in vivo assays.
Collapse
Affiliation(s)
- Isabel Karkossa
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Anne Bannuscher
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany; Adolphe Merkle Institute (AMI), University of Fribourg, Fribourg, Switzerland
| | - Bryan Hellack
- Institute of Energy and Environmental Technology (IUTA) e.V., Duisburg, Germany; German Environment Agency (UBA), Dessau, Germany
| | | | - Julie Laloy
- Department of Pharmacy, Namur Nanosafety Centre, University of Namur, Namur, Belgium
| | - Miruna S Stan
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Martin Wiemann
- IBE R&D Institute for Lung Health gGmbH, Münster, Germany
| | - Andreas Luch
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Andrea Haase
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany; Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | - Kristin Schubert
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany.
| |
Collapse
|
6
|
Prokop JW, Hartog NL, Chesla D, Faber W, Love CP, Karam R, Abualkheir N, Feldmann B, Teng L, McBride T, Leimanis ML, English BK, Holsworth A, Frisch A, Bauss J, Kalpage N, Derbedrossian A, Pinti RM, Hale N, Mills J, Eby A, VanSickle EA, Pageau SC, Shankar R, Chen B, Carcillo JA, Sanfilippo D, Olivero R, Bupp CP, Rajasekaran S. High-Density Blood Transcriptomics Reveals Precision Immune Signatures of SARS-CoV-2 Infection in Hospitalized Individuals. Front Immunol 2021; 12:694243. [PMID: 34335605 PMCID: PMC8322982 DOI: 10.3389/fimmu.2021.694243] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/30/2021] [Indexed: 12/27/2022] Open
Abstract
The immune response to COVID-19 infection is variable. How COVID-19 influences clinical outcomes in hospitalized patients needs to be understood through readily obtainable biological materials, such as blood. We hypothesized that a high-density analysis of host (and pathogen) blood RNA in hospitalized patients with SARS-CoV-2 would provide mechanistic insights into the heterogeneity of response amongst COVID-19 patients when combined with advanced multidimensional bioinformatics for RNA. We enrolled 36 hospitalized COVID-19 patients (11 died) and 15 controls, collecting 74 blood PAXgene RNA tubes at multiple timepoints, one early and in 23 patients after treatment with various therapies. Total RNAseq was performed at high-density, with >160 million paired-end, 150 base pair reads per sample, representing the most sequenced bases per sample for any publicly deposited blood PAXgene tube study. There are 770 genes significantly altered in the blood of COVID-19 patients associated with antiviral defense, mitotic cell cycle, type I interferon signaling, and severe viral infections. Immune genes activated include those associated with neutrophil mechanisms, secretory granules, and neutrophil extracellular traps (NETs), along with decreased gene expression in lymphocytes and clonal expansion of the acquired immune response. Therapies such as convalescent serum and dexamethasone reduced many of the blood expression signatures of COVID-19. Severely ill or deceased patients are marked by various secondary infections, unique gene patterns, dysregulated innate response, and peripheral organ damage not otherwise found in the cohort. High-density transcriptomic data offers shared gene expression signatures, providing unique insights into the immune system and individualized signatures of patients that could be used to understand the patient's clinical condition. Whole blood transcriptomics provides patient-level insights for immune activation, immune repertoire, and secondary infections that can further guide precision treatment.
Collapse
Affiliation(s)
- Jeremy W. Prokop
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Nicholas L. Hartog
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Allergy & Immunology, Spectrum Health, Grand Rapids, MI, United States
| | - Dave Chesla
- Office of Research, Spectrum Health, Grand Rapids, MI, United States
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - William Faber
- Physical Sciences, Grand Rapids Community College, Grand Rapids, MI, United States
| | - Chanise P. Love
- Office of Research, Spectrum Health, Grand Rapids, MI, United States
| | | | | | | | - Li Teng
- Ambry Genetics, Aliso Viejo, CA, United States
| | | | - Mara L. Leimanis
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Pediatric Intensive Care Unit, Helen DeVos Children’s Hospital, Grand Rapids, MI, United States
| | - B. Keith English
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Amanda Holsworth
- Allergy & Immunology, Spectrum Health, Grand Rapids, MI, United States
| | - Austin Frisch
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Jacob Bauss
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Nathisha Kalpage
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Aram Derbedrossian
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Ryan M. Pinti
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Nicole Hale
- The Department of Chemistry and Biochemistry, Calvin University, Grand Rapids, MI, United States
| | - Joshua Mills
- Department of Biology, Grand Valley State University, Allendale, MI, United States
| | - Alexandra Eby
- Department of Science, Davenport University, Grand Rapids, MI, United States
| | | | - Spencer C. Pageau
- Office of Research, Spectrum Health, Grand Rapids, MI, United States
| | - Rama Shankar
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Bin Chen
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Joseph A. Carcillo
- Department of Critical Care Medicine and Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Dominic Sanfilippo
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Pediatric Intensive Care Unit, Helen DeVos Children’s Hospital, Grand Rapids, MI, United States
| | - Rosemary Olivero
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Infectious Disease, Helen DeVos Children’s Hospital, Grand Rapids, MI, United States
| | - Caleb P. Bupp
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Medical Genetics, Spectrum Health Medical Genetics, Grand Rapids, MI, United States
| | - Surender Rajasekaran
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Office of Research, Spectrum Health, Grand Rapids, MI, United States
- Pediatric Intensive Care Unit, Helen DeVos Children’s Hospital, Grand Rapids, MI, United States
| |
Collapse
|
7
|
Kim HW, Seo SM, Kim JY, Lee JH, Lee HW, Choi YK. C1qa deficiency in mice increases susceptibility to mouse hepatitis virus A59 infection. J Vet Sci 2021; 22:e36. [PMID: 34056877 PMCID: PMC8170211 DOI: 10.4142/jvs.2021.22.e36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/26/2021] [Accepted: 04/15/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Mouse hepatitis virus (MHV) A59 is a highly infectious pathogen and starts in the respiratory tract and progresses to systemic infection in laboratory mice. The complement system is an important part of the host immune response to viral infection. It is not clear the role of the classical complement pathway in MHV infection. OBJECTIVES The purpose of this study was to determine the importance of the classical pathway in coronavirus pathogenesis by comparing C1qa KO mice and wild-type mice. METHODS We generated a C1qa KO mouse using CRISPR/Cas9 technology and compared the susceptibility to MHV A59 infection between C1qa KO and wild-type mice. Histopathological and immunohistochemical changes, viral loads, and chemokine expressions in both mice were measured. RESULTS MHV A59-infected C1qa KO mice showed severe histopathological changes, such as hepatocellular necrosis and interstitial pneumonia, compared to MHV A59-infected wild-type mice. Virus copy numbers in the olfactory bulb, liver, and lungs of C1qa KO mice were significantly higher than those of wild-type mice. The increase in viral copy numbers in C1qa KO mice was consistent with the histopathologic changes in organs. These results indicate that C1qa deficiency enhances susceptibility to MHV A59 systemic infection in mice. In addition, this enhanced susceptibility effect is associated with dramatic elevations in spleen IFN-γ, MIP-1 α, and MCP-1 in C1qa KO mice. CONCLUSIONS These data suggest that C1qa deficiency enhances susceptibility to MHV A59 systemic infection, and activation of the classical complement pathway may be important for protecting the host against MHV A59 infection.
Collapse
Affiliation(s)
- Han Woong Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- Regenerative Dental Medicine Institute, Hysensbio, Gwacheon 13814, Korea
| | - Sun Min Seo
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Jun Young Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- Green Cross Corporation, Yongin 16924, Korea
| | - Jae Hoon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Han Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Yang Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
8
|
Sood N, Verma DK, Paria A, Yadav SC, Yadav MK, Bedekar MK, Kumar S, Swaminathan TR, Mohan CV, Rajendran KV, Pradhan PK. Transcriptome analysis of liver elucidates key immune-related pathways in Nile tilapia Oreochromis niloticus following infection with tilapia lake virus. FISH & SHELLFISH IMMUNOLOGY 2021; 111:208-219. [PMID: 33577877 DOI: 10.1016/j.fsi.2021.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 06/12/2023]
Abstract
Nile tilapia (Oreochromis niloticus) is one of the most important aquaculture species farmed worldwide. However, the recent emergence of tilapia lake virus (TiLV) disease, also known as syncytial hepatitis of tilapia, has threatened the global tilapia industry. To gain more insight regarding the host response against the disease, the transcriptional profiles of liver in experimentally-infected and control tilapia were compared. Analysis of RNA-Seq data identified 4640 differentially expressed genes (DEGs), which were involved among others in antigen processing and presentation, MAPK, apoptosis, necroptosis, chemokine signaling, interferon, NF-kB, acute phase response and JAK-STAT pathways. Enhanced expression of most of the DEGs in the above pathways suggests an attempt by tilapia to resist TiLV infection. However, upregulation of some of the key genes such as BCL2L1 in apoptosis pathway; NFKBIA in NF-kB pathway; TRFC in acute phase response; and SOCS, EPOR, PI3K and AKT in JAK-STAT pathway and downregulation of the genes, namely MAP3K7 in MAPK pathway; IFIT1 in interferon; and TRIM25 in NF-kB pathway suggested that TiLV was able to subvert the host immune response to successfully establish the infection. The study offers novel insights into the cellular functions that are affected following TiLV infection and will serve as a valuable genomic resource towards our understanding of susceptibility of tilapia to TiLV infection.
Collapse
Affiliation(s)
- Neeraj Sood
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Dev Kumar Verma
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Anutosh Paria
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Shrish Chandra Yadav
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Manoj Kumar Yadav
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Megha Kadam Bedekar
- ICAR-Central Institute of Fisheries Education, Versova, Andheri (W), Mumbai, 400 061, Maharashtra, India
| | - Saurav Kumar
- ICAR-Central Institute of Fisheries Education, Versova, Andheri (W), Mumbai, 400 061, Maharashtra, India
| | - Thangaraj Raja Swaminathan
- Peninsular and Marine Fish Genetic Resources Centre, ICAR-NBFGR, CMFRI Campus, Kochi, 682 018, Kerala, India
| | | | - K V Rajendran
- ICAR-Central Institute of Fisheries Education, Versova, Andheri (W), Mumbai, 400 061, Maharashtra, India
| | - Pravata Kumar Pradhan
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India.
| |
Collapse
|
9
|
Spivack K, Muzzelo C, Hall M, Warga E, Neely C, Slepian H, Cunningham A, Tucker M, Elmer J. Enhancement of transgene expression by the β-catenin inhibitor iCRT14. Plasmid 2021; 114:102556. [PMID: 33472046 DOI: 10.1016/j.plasmid.2021.102556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/08/2020] [Accepted: 10/14/2020] [Indexed: 11/30/2022]
Abstract
The innate immune response is an essential defense mechanism that allows cells to detect pathogen-associated molecular patterns (PAMPs) like endotoxin or cytosolic DNA and then induce the expression of defensive genes that restrict the replication of viruses and other pathogens. However, the therapeutic DNA used in some gene therapy treatments can also trigger the innate immune response, which activates host cell genes that may inhibit transgene expression. The goal of this study was to enhance transgene expression by inhibiting key components of the innate immune response with small molecule inhibitors (iCRT14, curcumin, Amlexanox, H-151, SC-514, & VX-702). Most of the inhibitors significantly increased transgene (luciferase) expression at least 2-fold, but the β-catenin/TCF4 inhibitor iCRT14 showed the highest enhancement (16 to 35-fold) in multiple cell lines (PC-3, MCF7, & MB49) without significantly decreasing cellular proliferation. Alternatively, cloning a β-catenin/TCF4 binding motif (TCAAAG) into the EF1α promoter also enhanced transgene expression up to 8-fold. To further investigate the role of β-catenin/TCF4 in transgene expression, mRNA-sequencing experiments were conducted to identify host cell genes that were upregulated following transfection with PEI but down-regulated after the addition of iCRT14. As expected, transfection with plasmid DNA activated the innate immune response and upregulated hundreds (687) of defensive genes, but only 7 of those genes were down-regulated in the presence of iCRT14 (e.g., PTGS2 & PLA1A). Altogether, these results show that transgene expression can be enhanced by inhibiting the innate immune response with SMIs like iCRT14, which inhibits β-catenin/TCF4 to prevent the expression of specific host cell genes.
Collapse
Affiliation(s)
- Kyle Spivack
- Villanova University, Department of Chemical & Biological Engineering, United States
| | - Christine Muzzelo
- Villanova University, Department of Chemical & Biological Engineering, United States
| | - Matthew Hall
- Villanova University, Department of Chemical & Biological Engineering, United States
| | - Eric Warga
- Villanova University, Department of Chemical & Biological Engineering, United States
| | - Christopher Neely
- Villanova University, Department of Chemical & Biological Engineering, United States
| | - Holly Slepian
- Villanova University, Department of Chemical & Biological Engineering, United States
| | - Alyssa Cunningham
- Villanova University, Department of Chemical & Biological Engineering, United States
| | - Matthew Tucker
- Villanova University, Department of Chemical & Biological Engineering, United States
| | - Jacob Elmer
- Villanova University, Department of Chemical & Biological Engineering, United States.
| |
Collapse
|
10
|
Sebina I, Phipps S. The Contribution of Neutrophils to the Pathogenesis of RSV Bronchiolitis. Viruses 2020; 12:E808. [PMID: 32726921 PMCID: PMC7472258 DOI: 10.3390/v12080808] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Acute viral bronchiolitis causes significant mortality in the developing world, is the number one cause of infant hospitalisation in the developed world, and is associated with the later development of chronic lung diseases such as asthma. A vaccine against respiratory syncytial virus (RSV), the leading cause of viral bronchiolitis in infancy, remains elusive, and hence new therapeutic modalities are needed to limit disease severity. However, much remains unknown about the underlying pathogenic mechanisms. Neutrophilic inflammation is the predominant phenotype observed in infants with both mild and severe disease, however, a clear understanding of the beneficial and deleterious effects of neutrophils is lacking. In this review, we describe the multifaceted roles of neutrophils in host defence and antiviral immunity, consider their contribution to bronchiolitis pathogenesis, and discuss whether new approaches that target neutrophil effector functions will be suitable for treating severe RSV bronchiolitis.
Collapse
Affiliation(s)
- Ismail Sebina
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston 4006, Australia;
| | | |
Collapse
|
11
|
Wilson GJ, Fukuoka A, Love SR, Kim J, Pingen M, Hayes AJ, Graham GJ. Chemokine receptors coordinately regulate macrophage dynamics and mammary gland development. Development 2020; 147:dev187815. [PMID: 32467242 PMCID: PMC7328164 DOI: 10.1242/dev.187815] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/20/2020] [Indexed: 12/22/2022]
Abstract
Macrophages are key regulators of developmental processes, including those involved in mammary gland development. We have previously demonstrated that the atypical chemokine receptor ACKR2 contributes to the control of ductal epithelial branching in the developing mammary gland by regulating macrophage dynamics. ACKR2 is a chemokine-scavenging receptor that mediates its effects through collaboration with inflammatory chemokine receptors (iCCRs). Here, we reveal reciprocal regulation of branching morphogenesis in the mammary gland, whereby stromal ACKR2 modulates levels of the shared ligand CCL7 to control the movement of a key population of CCR1-expressing macrophages to the ductal epithelium. In addition, oestrogen, which is essential for ductal elongation during puberty, upregulates CCR1 expression on macrophages. The age at which girls develop breasts is decreasing, which raises the risk of diseases including breast cancer. This study presents a previously unknown mechanism controlling the rate of mammary gland development during puberty and highlights potential therapeutic targets.
Collapse
MESH Headings
- Animals
- Chemokine CCL3/deficiency
- Chemokine CCL3/genetics
- Chemokine CCL3/metabolism
- Chemokine CCL5/deficiency
- Chemokine CCL5/genetics
- Chemokine CCL5/metabolism
- Epithelium/metabolism
- Estradiol/pharmacology
- Female
- Lectins, C-Type/metabolism
- Macrophages/cytology
- Macrophages/metabolism
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/metabolism
- Mannose Receptor
- Mannose-Binding Lectins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Morphogenesis
- Receptors, CCR1/deficiency
- Receptors, CCR1/genetics
- Receptors, CCR1/metabolism
- Receptors, Cell Surface/metabolism
- Receptors, Chemokine/deficiency
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Gillian J Wilson
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Ayumi Fukuoka
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Samantha R Love
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Jiwon Kim
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
- Department of Physiology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Marieke Pingen
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Alan J Hayes
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Gerard J Graham
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
12
|
Yi X, Zhang J, Liu H, Yi T, Ou Y, Liu M, Zhu L, Chen H, Zhang J. Suppressed Immune-Related Profile Rescues Abortion-Prone Fetuses: A Novel Insight Into the CBA/J × DBA/2J Mouse Model. Reprod Sci 2019; 26:1485-1492. [PMID: 30791861 DOI: 10.1177/1933719119828042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The adverse clinical result and poor treatment outcome in recurrent spontaneous abortion (RSA) make it necessary to understand the pathogenic mechanism. The mating combination CBA/J × DBA/2 has been widely used as an abortion-prone model compared to DBA/2-mated CBA/J mice. Here, we used RNA-seq to get a comprehensive catalogue of genes differentially expressed between survival placenta in abortion-prone model and control. Five hundred twenty-four differentially expressed genes were obtained followed by clustering analysis, Gene Ontology analysis, and pathway analysis. We paid more attention to immune-related genes namely "immune response" and "immune system process" including 33 downregulated genes and 28 upregulated genes. Twenty-one genes contribute to suppressing immune system and 7 are against it. Six genes were validated by reverse transcription-polymerase chain reaction, namely Ccr1l1, Tlr4, Tgf-β1, Tyro3, Gzmb, and Il-1β. Furthermore, Tlr4, Tgf-β1, and Il-1β were analyzed by Western blot. Such immune profile gives us a better understanding of the complicated immune processing in RSA and immunosuppression can rescue pregnancy loss.
Collapse
Affiliation(s)
- Xiaochun Yi
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jie Zhang
- Department of Rehabilitation Medicine, Guangdong Women and Children Hospital, Guangzhou, People's Republic of China
| | - Huixiang Liu
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Tianxia Yi
- Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Yuhua Ou
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Meilan Liu
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Liqiong Zhu
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hui Chen
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jianping Zhang
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
13
|
Eguchi R, Karim MB, Hu P, Sato T, Ono N, Kanaya S, Altaf-Ul-Amin M. An integrative network-based approach to identify novel disease genes and pathways: a case study in the context of inflammatory bowel disease. BMC Bioinformatics 2018; 19:264. [PMID: 30005591 PMCID: PMC6043997 DOI: 10.1186/s12859-018-2251-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 06/18/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND There are different and complicated associations between genes and diseases. Finding the causal associations between genes and specific diseases is still challenging. In this work we present a method to predict novel associations of genes and pathways with inflammatory bowel disease (IBD) by integrating information of differential gene expression, protein-protein interaction and known disease genes related to IBD. RESULTS We downloaded IBD gene expression data from NCBI's Gene Expression Omnibus, performed statistical analysis to determine differentially expressed genes, collected known IBD genes from DisGeNet database, which were used to construct a IBD related PPI network with HIPPIE database. We adapted our graph-based clustering algorithm DPClusO to cluster the disease PPI network. We evaluated the statistical significance of the identified clusters in the context of determining the richness of IBD genes using Fisher's exact test and predicted novel genes related to IBD. We showed 93.8% of our predictions are correct in the context of other databases and published literatures related to IBD. CONCLUSIONS Finding disease-causing genes is necessary for developing drugs with synergistic effect targeting many genes simultaneously. Here we present an approach to identify novel disease genes and pathways and discuss our approach in the context of IBD. The approach can be generalized to find disease-associated genes for other diseases.
Collapse
Affiliation(s)
- Ryohei Eguchi
- Graduate School of Science and Technology & NAIST Data Science Center, Nara Institute of Science and Technology, Nara, Japan
| | - Mohammand Bozlul Karim
- Graduate School of Science and Technology & NAIST Data Science Center, Nara Institute of Science and Technology, Nara, Japan
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Canada.,George and Fay Yee Centre for Healthcare Innovation, University of Manitoba, Winnipeg, Canada.,Department of Electrical and Computer Engineering, University of Manitoba, Winnipeg, Canada
| | - Tetsuo Sato
- Graduate School of Science and Technology & NAIST Data Science Center, Nara Institute of Science and Technology, Nara, Japan.,Department of Radiological Technology, Gunma Prefectural College of Health Sciences, Gunma, Japan
| | - Naoaki Ono
- Graduate School of Science and Technology & NAIST Data Science Center, Nara Institute of Science and Technology, Nara, Japan
| | - Shigehiko Kanaya
- Graduate School of Science and Technology & NAIST Data Science Center, Nara Institute of Science and Technology, Nara, Japan
| | - Md Altaf-Ul-Amin
- Graduate School of Science and Technology & NAIST Data Science Center, Nara Institute of Science and Technology, Nara, Japan.
| |
Collapse
|
14
|
Chan EC, Ren C, Xie Z, Jude J, Barker T, Koziol-White CA, Ma M, Panettieri RA, Wu D, Rosenberg HF, Druey KM. Regulator of G protein signaling 5 restricts neutrophil chemotaxis and trafficking. J Biol Chem 2018; 293:12690-12702. [PMID: 29929985 DOI: 10.1074/jbc.ra118.002404] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/14/2018] [Indexed: 01/09/2023] Open
Abstract
Neutrophils are white blood cells that are mobilized to damaged tissues and to sites of pathogen invasion, providing the first line of host defense. Chemokines displayed on the surface of blood vessels promote migration of neutrophils to these sites, and tissue- and pathogen-derived chemoattractant signals, including N-formylmethionylleucylphenylalanine (fMLP), elicit further migration to sites of infection. Although nearly all chemoattractant receptors use heterotrimeric G proteins to transmit signals, many of the mechanisms lying downstream of chemoattractant receptors that either promote or limit neutrophil motility are incompletely defined. Here, we show that regulator of G protein signaling 5 (RGS5), a protein that modulates G protein activity, is expressed in both human and murine neutrophils. We detected significantly more neutrophils in the airways of Rgs5-/- mice than WT counterparts following acute respiratory virus infection and in the peritoneum in response to injection of thioglycollate, a biochemical proinflammatory stimulus. RGS5-deficient neutrophils responded with increased chemotaxis elicited by the chemokines CXC motif chemokine ligand 1 (CXCL1), CXCL2, and CXCL12 but not fMLP. Moreover, adhesion of these cells was increased in the presence of both CXCL2 and fMLP. In summary, our results indicate that RGS5 deficiency increases chemotaxis and adhesion, leading to more efficient neutrophil mobilization to inflamed tissues in mice. These findings suggest that RGS5 expression and activity in neutrophils determine their migrational patterns in the complex microenvironments characteristic of inflamed tissues.
Collapse
Affiliation(s)
- Eunice C Chan
- Molecular Signal Transduction Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Chunguang Ren
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Zhihui Xie
- Molecular Signal Transduction Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Joseph Jude
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers New Jersey School of Medicine, Rutgers, New Jersey 07103
| | - Tolga Barker
- Molecular Signal Transduction Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Cynthia A Koziol-White
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers New Jersey School of Medicine, Rutgers, New Jersey 07103
| | - Michelle Ma
- Inflammation Immunobiology Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers New Jersey School of Medicine, Rutgers, New Jersey 07103
| | - Dianqing Wu
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Helene F Rosenberg
- Inflammation Immunobiology Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Kirk M Druey
- Molecular Signal Transduction Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892.
| |
Collapse
|
15
|
Wujak L, Schnieder J, Schaefer L, Wygrecka M. LRP1: A chameleon receptor of lung inflammation and repair. Matrix Biol 2017; 68-69:366-381. [PMID: 29262309 DOI: 10.1016/j.matbio.2017.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 12/17/2022]
Abstract
The lung displays a remarkable capability to regenerate following injury. Considerable effort has been made thus far to understand the cardinal processes underpinning inflammation and reconstruction of lung tissue. However, the factors determining the resolution or persistence of inflammation and efficient wound healing or aberrant remodeling remain largely unknown. Low density lipoprotein receptor-related protein 1 (LRP1) is an endocytic/signaling cell surface receptor which controls cellular and molecular mechanisms driving the physiological and pathological inflammatory reactions and tissue remodeling in several organs. In this review, we will discuss the impact of LRP1 on the consecutive steps of the inflammatory response and its role in the balanced tissue repair and aberrant remodeling in the lung.
Collapse
Affiliation(s)
- Lukasz Wujak
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Jennifer Schnieder
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Liliana Schaefer
- Goethe University School of Medicine, University Hospital, Theodor-Stern Kai 7, 60590 Frankfurt am Main, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany; Member of the German Center for Lung Research (DZL), Germany.
| |
Collapse
|
16
|
Percopo CM, Ma M, Rosenberg HF. Administration of immunobiotic Lactobacillus plantarum delays but does not prevent lethal pneumovirus infection in Rag1-/- mice. J Leukoc Biol 2017; 102:905-913. [PMID: 28619948 DOI: 10.1189/jlb.3ab0217-050rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/10/2017] [Accepted: 05/11/2017] [Indexed: 12/11/2022] Open
Abstract
Administration of immunobiotic Lactobacillus plantarum (Lp) directly to the respiratory mucosa promotes cross-protection against lethal pneumovirus infection via B-cell-independent mechanisms. In this study, we examined Lp-mediated cross protection in Rag1-/- mice which cannot clear virus from lung tissue. Although Lp was initially protective, Rag1-/- mice ultimately succumbed to a delayed lethal outcome associated with local production of the proinflammatory cytokines CCL1, -2, and -7, granulocyte recruitment, and ongoing virus replication. By contrast, CD8null mice, which are fully capable of clearing virus, are protected by Lp with no delayed lethal outcome, granulocyte recruitment to the airways, or induction of CCL7. Repeated administration of Lp to virus-infected Rag1-/- mice had no impact on delayed mortality. Moreover, administration of Lp to the respiratory mucosa resulted in no induction of IFN-α or -β in Rag1-/- or wild-type mice, and IFN-abR gene deletion had no impact on Lp-mediated protection. Overall, our findings indicate that although Lp administered to the respiratory tract has substantial impact on lethal virus-induced inflammation in situ, endogenous virus clearance mechanisms are needed to promote sustained protection. Our results suggest that a larger understanding of the mechanisms and mediators that limit acute virus-induced inflammation may yield new and useful therapeutic modalities.
Collapse
Affiliation(s)
- Caroline M Percopo
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Michelle Ma
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Helene F Rosenberg
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
17
|
Taylor G. Animal models of respiratory syncytial virus infection. Vaccine 2017; 35:469-480. [PMID: 27908639 PMCID: PMC5244256 DOI: 10.1016/j.vaccine.2016.11.054] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/12/2016] [Accepted: 11/16/2016] [Indexed: 11/26/2022]
Abstract
Human respiratory syncytial virus (hRSV) is a major cause of respiratory disease and hospitalisation of infants, worldwide, and is also responsible for significant morbidity in adults and excess deaths in the elderly. There is no licensed hRSV vaccine or effective therapeutic agent. However, there are a growing number of hRSV vaccine candidates that have been developed targeting different populations at risk of hRSV infection. Animal models of hRSV play an important role in the preclinical testing of hRSV vaccine candidates and although many have shown efficacy in preclinical studies, few have progressed to clinical trials or they have had only limited success. This is, at least in part, due to the lack of animal models that fully recapitulate the pathogenesis of hRSV infection in humans. This review summarises the strengths and limitations of animal models of hRSV, which include those in which hRSV is used to infect non-human mammalian hosts, and those in which non-human pneumoviruses, such as bovine (b)RSV and pneumonia virus of mice (PVM) are studied in their natural host. Apart from chimpanzees, other non-human primates (NHP) are only semi-permissive for hRSV replication and experimental infection with large doses of virus result in little or no clinical signs of disease, and generally only mild pulmonary pathology. Other animal models such as cotton rats, mice, ferrets, guinea pigs, hamsters, chinchillas, and neonatal lambs are also only semi-permissive for hRSV. Nevertheless, mice and cotton rats have been of value in the development of monoclonal antibody prophylaxis for infants at high risk of severe hRSV infection and have provided insights into mechanisms of immunity to and pathogenesis of hRSV. However, the extent to which they predict hRSV vaccine efficacy and safety is unclear and several hRSV vaccine candidates that are completely protective in rodent models are poorly effective in chimpanzees and other NHP, such as African Green monkeys. Furthermore, interpretation of findings from many rodent and NHP models of vaccine-enhanced hRSV disease has been confounded by sensitisation to non-viral antigens present in the vaccine and challenge virus. Studies of non-human pneumoviruses in their native hosts are more likely to reflect the pathogenesis of natural hRSV infection, and experimental infection of calves with bRSV and of mice with PVM result in clinical disease and extensive pulmonary pathology. These animal models have not only been of value in studies on mechanisms of immunity to and the pathogenesis of pneumovirus infections but have also been used to evaluate hRSV vaccine concepts. Furthermore, the similarities between the epidemiology of bRSV in calves and hRSV in infants and the high level of genetic and antigenic similarity between bRSV and hRSV, make the calf model of bRSV infection a relevant model for preclinical evaluation of hRSV vaccine candidates which contain proteins that are conserved between hRSV and bRSV.
Collapse
Affiliation(s)
- Geraldine Taylor
- The Pirbright Institute, Ash Road, Pirbright, Woking Surrey GU24 0NF, United Kingdom.
| |
Collapse
|
18
|
Kubo H, Hoshi M, Mouri A, Tashita C, Yamamoto Y, Nabeshima T, Saito K. Absence of kynurenine 3-monooxygenase reduces mortality of acute viral myocarditis in mice. Immunol Lett 2016; 181:94-100. [PMID: 27889626 DOI: 10.1016/j.imlet.2016.11.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/28/2016] [Accepted: 11/22/2016] [Indexed: 12/12/2022]
Abstract
Infection of the encephalomyocarditis virus (EMCV) in mice is an established model for viral myocarditis. Previously, we have demonstrated that indoleamine 2,3-dioxygenase (IDO), an L-tryptophan - kynurenine pathway (KP) enzyme, affects acute viral myocarditis. However, the roles of KP metabolites in EMCV infection remain unclear. Kynurenine 3-monooxygenase (KMO) is one of the key regulatory enzymes, which metabolizes kynurenine to 3-hydroxykynurenine in the KP. Therefore, we examined the role of KMO in acute viral infection by comparing between KMO-/- mice and KMO+/+ mice. KMO deficiency resulted in suppressed mortality after EMCV infection. The number of infiltrating cells and F4/80+ cells in KMO-/- mice was suppressed compared with those in KMO+/+ mice. KMO-/- mice showed significantly increased levels of serum KP metabolites, and induction of KMO expression upon EMCV infection was involved in its effect on mortality through EMCV suppression. Furthermore, KMO-/- mice showed significantly suppression of CCL2, CCL3 and CCL4 on day 2 and CXCL1 on day 4 after infection. These results suggest that increased KP metabolites reduced chemokine production, resulting in suppressed mortality upon KMO knockdown in EMCV infection. KP metabolites may thus provide an effective strategy for treating acute viral myocarditis.
Collapse
Affiliation(s)
- Hisako Kubo
- Human Health Sciences, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto 606-8507, Japan; Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Sciences, Aichi 470-1192, Japan
| | - Masato Hoshi
- Department of Biochemical and Analytical Sciences, Fujita Health University Graduate School of Health Sciences, Aichi 470-1192, Japan.
| | - Akihiro Mouri
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Aichi 470-1192, Japan
| | - Chieko Tashita
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Sciences, Aichi 470-1192, Japan; Department of Medical Technology, Gifu University of Medical Science, Gifu 501-3892, Japan
| | - Yasuko Yamamoto
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Sciences, Aichi 470-1192, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Aichi 470-1192, Japan; Japanese Drug Organization of Appropriate Use and Research, Aichi 468-0069, Japan; Aino University, Osaka, Ibaragi 567-0012, Japan
| | - Kuniaki Saito
- Human Health Sciences, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto 606-8507, Japan; Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Sciences, Aichi 470-1192, Japan; Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Aichi 470-1192, Japan
| |
Collapse
|
19
|
Brenner TA, Rice TA, Anderson ED, Percopo CM, Rosenberg HF. Immortalized MH-S cells lack defining features of primary alveolar macrophages and do not support mouse pneumovirus replication. Immunol Lett 2016; 172:106-12. [PMID: 26916143 DOI: 10.1016/j.imlet.2016.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/12/2016] [Accepted: 02/17/2016] [Indexed: 12/15/2022]
Abstract
The SV-40-transformed MH-S cell line maintains some, but not all, features of primary alveolar macrophages (AMs) from BALB/c mice. We show here that MH-S cells produce inflammatory cytokines IL-6 and CXCL10 in response to challenge with Gram-positive Lactobacillus reuteri, and to TLR2 and NOD2 ligands Pam3CSK4 and MDP, respectively. In contrast, although wild-type AMs are infected in vivo by pneumonia virus of mice (PVM), no virus replication was detected in MH-S cells. Interestingly, the surface immunophenotype of MH-S cells (CD11c(+)Siglec F(-)) differs from that of wild-type AMs (CD11c(+) Siglec F(+)) and is similar to that of immature AMs isolated from granulocyte macrophage-colony stimulating factor (GM-CSF) gene-deleted mice; AMs from GM-CSF(-/-) mice also support PVM replication. However, MH-S cells do not express the GM-CSF receptor alpha chain (CD116) and do not respond to GM-CSF. Due to these unusual features, MH-S cells should be used with caution as experimental models of AMs.
Collapse
Affiliation(s)
- Todd A Brenner
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Tyler A Rice
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Erik D Anderson
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Caroline M Percopo
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Helene F Rosenberg
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
20
|
Priming of the Respiratory Tract with Immunobiotic Lactobacillus plantarum Limits Infection of Alveolar Macrophages with Recombinant Pneumonia Virus of Mice (rK2-PVM). J Virol 2015; 90:979-91. [PMID: 26537680 DOI: 10.1128/jvi.02279-15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 10/27/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Pneumonia virus of mice (PVM) is a natural rodent pathogen that replicates in bronchial epithelial cells and reproduces many clinical and pathological features of the more severe forms of disease associated with human respiratory syncytial virus. In order to track virus-target cell interactions during acute infection in vivo, we developed rK2-PVM, bacterial artificial chromosome-based recombinant PVM strain J3666 that incorporates the fluorescent tag monomeric Katushka 2 (mKATE2). The rK2-PVM pathogen promotes lethal infection in BALB/c mice and elicits characteristic cytokine production and leukocyte recruitment to the lung parenchyma. Using recombinant virus, we demonstrate for the first time PVM infection of both dendritic cells (DCs; CD11c(+) major histocompatibility complex class II(+)) and alveolar macrophages (AMs; CD11c(+) sialic acid-binding immunoglobulin-like lectin F(+)) in vivo and likewise detect mKATE2(+) DCs in mediastinal lymph nodes from infected mice. AMs support both active virus replication and production of infectious virions. Furthermore, we report that priming of the respiratory tract with immunobiotic Lactobacillus plantarum, a regimen that results in protection against the lethal inflammatory sequelae of acute respiratory virus infection, resulted in differential recruitment of neutrophils, DCs, and lymphocytes to the lungs in response to rK2-PVM and a reduction from ∼ 40% to <10% mKATE2(+) AMs in association with a 2-log drop in the release of infectious virions. In contrast, AMs from L. plantarum-primed mice challenged with virus ex vivo exhibited no differential susceptibility to rK2-PVM. Although the mechanisms underlying Lactobacillus-mediated viral suppression remain to be fully elucidated, this study provides insight into the cellular basis of this response. IMPORTANCE Pneumonia virus of mice (PVM) is a natural mouse pathogen that serves as a model for severe human respiratory syncytial virus disease. We have developed a fully functional recombinant PVM strain with a fluorescent reporter protein (rK2-PVM) that permits us to track infection of target cells in vivo. With rK2-PVM, we demonstrate infection of leukocytes in the lung, notably, dendritic cells and alveolar macrophages. Alveolar macrophages undergo productive infection and release infectious virions. We have shown previously that administration of immunobiotic Lactobacillus directly to the respiratory mucosa protects mice from the lethal sequelae of PVM infection in association with profound suppression of the virus-induced inflammatory response. We show here that Lactobacillus administration also limits infection of leukocytes in vivo and results in diminished release of infectious virions from alveolar macrophages. This is the first study to provide insight into the cellular basis of the antiviral impact of immunobiotic L. plantarum.
Collapse
|
21
|
IL-21-mediated non-canonical pathway for IL-1β production in conventional dendritic cells. Nat Commun 2015; 6:7988. [PMID: 26269257 PMCID: PMC4555999 DOI: 10.1038/ncomms8988] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/03/2015] [Indexed: 01/10/2023] Open
Abstract
The canonical pathway for IL-1β production requires TLR-mediated NF-κB-dependent Il1b gene induction, followed by caspase-containing inflammasome-mediated processing of pro-IL-1β. Here we show that IL-21 unexpectedly induces IL-1β production in conventional dendritic cells (cDCs) via a STAT3-dependent but NF-κB-independent pathway. IL-21 does not induce Il1b expression in CD4+ T cells, with differential histone marks present in these cells versus cDCs. IL-21-induced IL-1β processing in cDCs does not require caspase-1 or caspase-8 but depends on IL-21-mediated death and activation of serine protease(s). Moreover, STAT3-dependent IL-1β expression in cDCs at least partially explains the IL-21-mediated pathologic response occurring during infection with pneumonia virus of mice. These results demonstrate lineage-restricted IL-21-induced IL-1β via a non-canonical pathway and provide evidence for its importance in vivo. The proinflammatory cytokine interleukin-1ß (IL-1ß) plays an important role in host defence against pathogens. Here the authors report a non-canonical pathway for IL-1 ß production in conventional dendritic cells that is induced by IL-21 via STAT3-dependent mechanism.
Collapse
|
22
|
Yang XQ, Wang L, Li HT, Liu D. Immune responses of porcine airway epithelial cells to poly(I:C), a synthetic analogue of viral double-stranded RNA. CANADIAN JOURNAL OF ANIMAL SCIENCE 2015. [DOI: 10.4141/cjas2013-145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Yang, X.-q., Wang, L., Li, H.-t. and Liu, D. 2015. Immune responses of porcine airway epithelial cells to poly(I:C), a synthetic analogue of viral double-stranded RNA. Can. J. Anim. Sci. 95: 13–20. Swine respiratory disease (SRD) is one of the most economically important diseases affecting the pig industry. The main infectious agents that cause SRD are viruses, but the molecular pathogenesis of viral SRD has not been extensively studied. Here, using digital gene expression tag profiling, the global transcriptional responses to poly(I:C), a synthetic analogue of viral double-stranded RNA, was analyzed in porcine airway epithelial cells (PAECs). The profiling analysis revealed numerous differentially expressed genes (DEGs), including unknown sequences in the porcine nucleotide databases. Gene ontology enrichment analysis showed that DEGs were mainly enriched in response to stress (GO: 0006950), of which, defense response is one sub-process. Poly(I:C) challenge induced a general inflammation response as indicated by marked upregulation of a variety of pathogen recognition receptors, interferon-stimulated genes, proinflammatory cytokines, and chemokines, together with the significant downregulation of anti-inflammatory molecules. Furthermore, the antiapoptotic pathway was triggered, as demonstrated by the significant suppression of molecules involved in the induction of apoptosis, together with the significant stimulation of putative inhibitor of apoptosis. The results indicate that PAECs initiated defense against poly(I:C) challenge through the inflammation responses, whereas poly(I:C) can utilize antiapoptotic pathway to evade host defense.
Collapse
Affiliation(s)
- Xiu-qin Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Liang Wang
- Heilongjiang Academy of Agricultural sciences, Harbin, China
| | - Hai-tao Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Di Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
- Heilongjiang Academy of Agricultural sciences, Harbin, China
| |
Collapse
|
23
|
Fulkerson PC, Schollaert KL, Bouffi C, Rothenberg ME. IL-5 triggers a cooperative cytokine network that promotes eosinophil precursor maturation. THE JOURNAL OF IMMUNOLOGY 2014; 193:4043-52. [PMID: 25230753 DOI: 10.4049/jimmunol.1400732] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Eosinophils originate in the bone marrow from an eosinophil lineage-committed, IL-5Rα-positive, hematopoietic progenitor (eosinophil progenitor). Indeed, IL-5 is recognized as a critical regulator of eosinophilia and has effects on eosinophil progenitors, eosinophil precursors, and mature eosinophils. However, substantial levels of eosinophils remain after IL-5 neutralization or genetic deletion, suggesting that there are alternative pathways for promoting eosinophilia. In this study, we investigated the contributory role of IL-5 accessory cytokines on the final stages of eosinophil differentiation. IL-5 stimulation of low-density bone marrow cells resulted in expression of a panel of cytokines and cytokine receptors, including several ligand-receptor pairs. Notably, IL-4 and IL-4Rα were expressed by eosinophil precursors and mature eosinophils. Signaling through IL-4Rα promoted eosinophil maturation when IL-5 was present, but IL-4 stimulation in the absence of IL-5 resulted in impaired eosinophil survival, suggesting that IL-4 cooperates with IL-5 to promote eosinophil differentiation. In contrast, CCL3, an eosinophil precursor-produced chemokine that signals through CCR1, promotes terminal differentiation of CCR1-positive eosinophil precursors in the absence of IL-5, highlighting an autocrine loop capable of sustaining eosinophil differentiation. These findings suggest that brief exposure to IL-5 is sufficient to initiate a cytokine cooperative network that promotes eosinophil differentiation of low-density bone marrow cells independent of further IL-5 stimulation.
Collapse
Affiliation(s)
- Patricia C Fulkerson
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Kaila L Schollaert
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Carine Bouffi
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| |
Collapse
|
24
|
van den Berg E, Bem RA, Bos AP, Lutter R, van Woensel JBM. The effect of TIP on pneumovirus-induced pulmonary edema in mice. PLoS One 2014; 9:e102749. [PMID: 25047452 PMCID: PMC4105480 DOI: 10.1371/journal.pone.0102749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 06/20/2014] [Indexed: 01/14/2023] Open
Abstract
Background Pulmonary edema plays a pivotal role in the pathophysiology of respiratory syncytial virus (RSV)-induced respiratory failure. In this study we determined whether treatment with TIP (AP301), a synthetic cyclic peptide that mimics the lectin-like domain of human TNF, decreases pulmonary edema in a mouse model of severe human RSV infection. TIP is currently undergoing clinical trials as a therapy for pulmonary permeability edema and has been shown to decrease pulmonary edema in different lung injury models. Methods C57BL/6 mice were infected with pneumonia virus of mice (PVM) and received TIP or saline (control group) by intratracheal instillation on day five (early administration) or day seven (late administration) after infection. In a separate set of experiments the effect of multiple dose administration of TIP versus saline was tested. Pulmonary edema was determined by the lung wet-to-dry (W/D) weight ratio and was assessed at different time-points after the administration of TIP. Secondary outcomes included clinical scores and lung cellular response. Results TIP did not have an effect on pulmonary edema in different dose regimens at different time points during PVM infection. In addition, TIP administration did not affect clinical severity scores or lung cellular response. Conclusion In this murine model of severe RSV infection TIP did not affect pulmonary edema nor course of disease.
Collapse
Affiliation(s)
- Elske van den Berg
- Pediatric Intensive Care Unit, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands
- * E-mail:
| | - Reinout A. Bem
- Pediatric Intensive Care Unit, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands
| | - Albert P. Bos
- Pediatric Intensive Care Unit, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands
| | - Rene Lutter
- Department of Respiratory Medicine and Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands
| | - Job B. M. van Woensel
- Pediatric Intensive Care Unit, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Chemokine (C-C Motif) receptor 1 is required for efficient recruitment of neutrophils during respiratory infection with modified vaccinia virus Ankara. J Virol 2014; 88:10840-50. [PMID: 25008920 DOI: 10.1128/jvi.01524-14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED Modified vaccinia virus Ankara (MVA) serves as a versatile platform in vaccine development. This highly attenuated orthopoxvirus, which cannot replicate in mammalian cells, triggers strong innate immune responses, including cell migration. Previously, we have shown that induction of chemokine (C-C motif) ligand 2 (CCL2) by MVA is necessary for the recruitment of monocytes and T cells, but not neutrophils, to the lung. Here, we identified neutrophil-attracting chemokines produced by MVA-infected primary murine lung fibroblasts and murine bone marrow-derived macrophages. We demonstrate that MVA, but not vaccinia virus (VACV) strain WR, induces chemokine expression, which is independent of Toll-like receptor 2 (TLR2) signaling. Additionally, we show that both chemokine (C-C motif) receptor 1 (CCR1) and chemokine (C-X-C motif) receptor 2 (CXCR2) are involved in MVA-induced neutrophil chemotaxis in vitro. Finally, intranasal infection of Ccr1(-/-) mice with MVA, as well as application of the CCR1 antagonist J-113863, revealed a role for CCR1 in leukocyte recruitment, including neutrophils, into the lung. IMPORTANCE Rapid attraction of leukocytes to the site of inoculation is unique to MVA in comparison to other VACV strains. The findings here extend current knowledge about the regulation of MVA-induced leukocyte migration, particularly regarding neutrophils, which could potentially be exploited to improve other VACV strains currently in development as oncolytic viruses and viral vectors. Additionally, the data presented here indicate that the inflammatory response may vary depending on the cell type infected by MVA, highlighting the importance of the site of vaccine application. Moreover, the rapid recruitment of neutrophils and other leukocytes can directly contribute to the induction of adaptive immune responses elicited by MVA inoculation. Thus, a better understanding of leukocyte migration upon MVA infection is particularly relevant for further development and use of MVA-based vaccines and vectors.
Collapse
|
26
|
Rowley SM, Kuriakose T, Dockery LM, Tran-Nguyen T, Gingerich AD, Wei L, Watford WT. Tumor progression locus 2 (Tpl2) kinase promotes chemokine receptor expression and macrophage migration during acute inflammation. J Biol Chem 2014; 289:15788-97. [PMID: 24713702 DOI: 10.1074/jbc.m114.559344] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In autoimmune diseases, the accumulation of activated leukocytes correlates with inflammation and disease progression, and, therefore, the disruption of leukocyte trafficking is an active area of research. The serine/threonine protein kinase Tpl2 (MAP3K8) regulates leukocyte inflammatory responses and is also being investigated for therapeutic inhibition during autoimmunity. Here we addressed the contribution of Tpl2 to the regulation of macrophage chemokine receptor expression and migration in vivo using a mouse model of Tpl2 ablation. LPS stimulation of bone marrow-derived macrophages induced early CCR1 chemokine receptor expression but repressed CCR2 and CCR5 expression. Notably, early induction of CCR1 expression by LPS was dependent upon a signaling pathway involving Tpl2, PI3K, and ERK. On the contrary, Tpl2 was required to maintain the basal expression of CCR2 and CCR5 as well as to stabilize CCR5 mRNA expression. Consistent with impairments in chemokine receptor expression, tpl2(-/-) macrophages were defective in trafficking to the peritoneal cavity following thioglycollate-induced inflammation. Overall, this study demonstrates a Tpl2-dependent mechanism for macrophage expression of select chemokine receptors and provides further insight into how Tpl2 inhibition may be used therapeutically to disrupt inflammatory networks in vivo.
Collapse
Affiliation(s)
- Sean M Rowley
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602 and
| | - Teneema Kuriakose
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602 and
| | - Lee M Dockery
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602 and
| | - Thi Tran-Nguyen
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602 and
| | - Aaron D Gingerich
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602 and
| | - Lai Wei
- the Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Wendy T Watford
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602 and
| |
Collapse
|
27
|
Animal model of respiratory syncytial virus: CD8+ T cells cause a cytokine storm that is chemically tractable by sphingosine-1-phosphate 1 receptor agonist therapy. J Virol 2014; 88:6281-93. [PMID: 24672024 DOI: 10.1128/jvi.00464-14] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED The cytokine storm is an intensified, dysregulated, tissue-injurious inflammatory response driven by cytokine and immune cell components. The cytokine storm during influenza virus infection, whereby the amplified innate immune response is primarily responsible for pulmonary damage, has been well characterized. Now we describe a novel event where virus-specific T cells induce a cytokine storm. The paramyxovirus pneumonia virus of mice (PVM) is a model of human respiratory syncytial virus (hRSV). Unexpectedly, when C57BL/6 mice were infected with PVM, the innate inflammatory response was undetectable until day 5 postinfection, at which time CD8(+) T cells infiltrated into the lung, initiating a cytokine storm by their production of gamma interferon (IFN-γ) and tumor necrosis factor alpha (TNF-α). Administration of an immunomodulatory sphingosine-1-phosphate (S1P) receptor 1 (S1P1R) agonist significantly inhibited PVM-elicited cytokine storm by blunting the PVM-specific CD8(+) T cell response, resulting in diminished pulmonary disease and enhanced survival. IMPORTANCE A dysregulated overly exuberant immune response, termed a "cytokine storm," accompanies virus-induced acute respiratory diseases (VARV), is primarily responsible for the accompanying high morbidity and mortality, and can be controlled therapeutically in influenza virus infection of mice and ferrets by administration of sphingosine-1-phosphate 1 receptor (S1P1R) agonists. Here, two novel findings are recorded. First, in contrast to influenza infection, where the cytokine storm is initiated early by the innate immune system, for pneumonia virus of mice (PVM), a model of RSV, the cytokine storm is initiated late in infection by the adaptive immune response: specifically, by virus-specific CD8 T cells via their release of IFN-γ and TNF-α. Blockading these cytokines with neutralizing antibodies blunts the cytokine storm and protects the host. Second, PVM infection is controlled by administration of an S1P1R agonist.
Collapse
|
28
|
Suryadevara M, Bonville CA, Rosenberg HF, Domachowske JB. Local production of CCL3, CCL11, and IFN-γ correlates with disease severity in murine parainfluenza virus infection. Virol J 2013; 10:357. [PMID: 24359540 PMCID: PMC3878101 DOI: 10.1186/1743-422x-10-357] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 12/10/2013] [Indexed: 01/07/2023] Open
Abstract
Background Using a murine model of parainfluenza virus infection (mPIV1 or Sendai virus; SeV), we compared the inflammatory responses to lethal and sub-lethal infections in inbred DBA/2 mice. Methods Mice were intranasally inoculated with either 1.6×103 or 1.6×105 infectious units (IU) of SeV or diluent control. Clinical data including daily weights, oxygen saturation, and lung function via whole body plethysmography were collected on days 0, 3–7, and 9–14. Clarified whole lung homogenates were evaluated for inflammatory markers by enzyme-linked immunoassay (ELISA). Data were analyzed using ANOVA or Student t-tests, as appropriate. Results Mice inoculated with 1.6×105 IU of SeV developed a lethal infection with 100% mortality by day 7, while mice inoculated with 1.6×103 IU developed a clinically significant infection, with universal weight loss but only 32% mortality. Interestingly, peak virus recovery from the lungs of mice inoculated with 1.6×105 IU of SeV did not differ substantially from that detected in mice that received the 100-fold lower inoculum. In contrast, concentrations of CCL5 (RANTES), CCL11 (eotaxin), interferon-γ, CXCL10 (IP-10), and CCL3 (MIP-1α) were significantly higher in lung tissue homogenates from mice inoculated with 1.6×105 IU (p < 0.05). In the lethal infection, levels of CCL11, interferon- γ and CCL3 all correlated strongly with disease severity. Conclusion We observed that severity of SeV-infection in DBA/2 mice was not associated with virus recovery but rather with the levels of proinflammatory cytokines, specifically CCL11, interferon- γ and CCL3, detected in lung tissue in response to SeV infection.
Collapse
Affiliation(s)
- Manika Suryadevara
- Department of Pediatrics, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA.
| | | | | | | |
Collapse
|
29
|
Bachelerie F, Ben-Baruch A, Burkhardt AM, Combadiere C, Farber JM, Graham GJ, Horuk R, Sparre-Ulrich AH, Locati M, Luster AD, Mantovani A, Matsushima K, Murphy PM, Nibbs R, Nomiyama H, Power CA, Proudfoot AEI, Rosenkilde MM, Rot A, Sozzani S, Thelen M, Yoshie O, Zlotnik A. International Union of Basic and Clinical Pharmacology. [corrected]. LXXXIX. Update on the extended family of chemokine receptors and introducing a new nomenclature for atypical chemokine receptors. Pharmacol Rev 2013; 66:1-79. [PMID: 24218476 DOI: 10.1124/pr.113.007724] [Citation(s) in RCA: 668] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Sixteen years ago, the Nomenclature Committee of the International Union of Pharmacology approved a system for naming human seven-transmembrane (7TM) G protein-coupled chemokine receptors, the large family of leukocyte chemoattractant receptors that regulates immune system development and function, in large part by mediating leukocyte trafficking. This was announced in Pharmacological Reviews in a major overview of the first decade of research in this field [Murphy PM, Baggiolini M, Charo IF, Hébert CA, Horuk R, Matsushima K, Miller LH, Oppenheim JJ, and Power CA (2000) Pharmacol Rev 52:145-176]. Since then, several new receptors have been discovered, and major advances have been made for the others in many areas, including structural biology, signal transduction mechanisms, biology, and pharmacology. New and diverse roles have been identified in infection, immunity, inflammation, development, cancer, and other areas. The first two drugs acting at chemokine receptors have been approved by the U.S. Food and Drug Administration (FDA), maraviroc targeting CCR5 in human immunodeficiency virus (HIV)/AIDS, and plerixafor targeting CXCR4 for stem cell mobilization for transplantation in cancer, and other candidates are now undergoing pivotal clinical trials for diverse disease indications. In addition, a subfamily of atypical chemokine receptors has emerged that may signal through arrestins instead of G proteins to act as chemokine scavengers, and many microbial and invertebrate G protein-coupled chemokine receptors and soluble chemokine-binding proteins have been described. Here, we review this extended family of chemokine receptors and chemokine-binding proteins at the basic, translational, and clinical levels, including an update on drug development. We also introduce a new nomenclature for atypical chemokine receptors with the stem ACKR (atypical chemokine receptor) approved by the Nomenclature Committee of the International Union of Pharmacology and the Human Genome Nomenclature Committee.
Collapse
Affiliation(s)
- Francoise Bachelerie
- Chair, Subcommittee on Chemokine Receptors, Nomenclature Committee-International Union of Pharmacology, Bldg. 10, Room 11N113, NIH, Bethesda, MD 20892.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
The paramyxovirus pneumonia virus of mice (PVM) is a rodent model of human respiratory syncytial virus (hRSV) pathogenesis. Here we characterized the PVM-specific CD8(+) T-cell repertoire in susceptible C57BL/6 mice. In total, 15 PVM-specific CD8(+) T-cell epitopes restricted by H-2D(b) and/or H-2K(b) were identified. These data open the door for using widely profiled, genetically manipulated C57BL/6 mice to study the contribution of epitope-specific CD8(+) T cells to PVM pathogenesis.
Collapse
|
31
|
Abstract
OBJECTIVE To determine biventricular cardiac function in pneumovirus-induced acute lung injury in spontaneously breathing mice. DESIGN Experimental animal study. SETTING Animal laboratory. SUBJECTS C57Bl/6 mice. INTERVENTION Mice were inoculated with the rodent pneumovirus, pneumonia virus of mice. MEASUREMENTS AND MAIN RESULTS Pneumonia virus of mice-infected mice were studied for right and left ventricular function variables by high-field strength (7 Tesla) cardiac MRI at specific time points during the course of disease compared with baseline. One day before and at peak disease severity, pneumonia virus of mice-infected mice showed significant right and left ventricular systolic and diastolic volume changes, with a progressive decrease in stroke volume and ejection fraction. No evidence for viral myocarditis or viral presence in heart tissue was found. CONCLUSIONS These findings show adverse pulmonary-cardiac interaction in pneumovirus-induced acute lung injury, unrelated to direct virus-mediated effects on the heart.
Collapse
|
32
|
LV WEI, DUAN QIANGLIN, WANG LEMIN, GONG ZHU, YANG FAN, SONG YANLI. Gene expression levels of cytokines in peripheral blood mononuclear cells from patients with pulmonary embolism. Mol Med Rep 2013; 7:1245-50. [DOI: 10.3892/mmr.2013.1344] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 02/18/2013] [Indexed: 11/06/2022] Open
|
33
|
Chandrasekar B, Deobagkar-Lele M, Victor ES, Nandi D. Regulation of Chemokines, CCL3 and CCL4, by Interferon γ and Nitric Oxide Synthase 2 in Mouse Macrophages and During Salmonella enterica Serovar Typhimurium Infection. J Infect Dis 2013; 207:1556-68. [DOI: 10.1093/infdis/jit067] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
34
|
Innate and adaptive immune response to pneumonia virus of mice in a resistant and a susceptible mouse strain. Viruses 2013; 5:295-320. [PMID: 23337382 PMCID: PMC3564122 DOI: 10.3390/v5010295] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 01/13/2013] [Accepted: 01/15/2013] [Indexed: 12/25/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of infant bronchiolitis. The closely related pneumonia virus of mice (PVM) causes a similar immune-mediated disease in mice, which allows an analysis of host factors that lead to severe illness. This project was designed to compare the immune responses to lethal and sublethal doses of PVM strain 15 in Balb/c and C57Bl/6 mice. Balb/c mice responded to PVM infection with an earlier and stronger innate response that failed to control viral replication. Production of inflammatory cyto- and chemokines, as well as infiltration of neutrophils and IFN-γ secreting natural killer cells into the lungs, was more predominant in Balb/c mice. In contrast, C57Bl/6 mice were capable of suppressing both viral replication and innate inflammatory responses. After a sublethal infection, PVM-induced IFN-γ production by splenocytes was stronger early during infection and weaker at late time points in C57Bl/6 mice when compared to Balb/c mice. Furthermore, although the IgG levels were similar and the mucosal IgA titres lower, the virus neutralizing antibody titres were higher in C57Bl/6 mice than in Balb/c mice. Overall, the difference in susceptibility of these two strains appeared to be related not to an inherent T helper bias, but to the capacity of the C57Bl/6 mice to control both viral replication and the immune response elicited by PVM.
Collapse
|
35
|
Peng BH, Borisevich V, Popov VL, Zacks MA, Estes DM, Campbell GA, Paessler S. Production of IL-8, IL-17, IFN-gamma and IP-10 in human astrocytes correlates with alphavirus attenuation. Vet Microbiol 2013; 163:223-34. [PMID: 23428380 PMCID: PMC7117234 DOI: 10.1016/j.vetmic.2012.11.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 11/06/2012] [Accepted: 11/22/2012] [Indexed: 01/30/2023]
Abstract
Venezuelan equine encephalitis virus (VEEV) is an important, naturally emerging zoonotic pathogen. Recent outbreaks in Venezuela and Colombia in 1995 indicate that VEEV still poses a serious public health threat. Astrocytes may be target cells in human and mouse infection and they play an important role in repair through gliosis. In this study, we report that virulent VEEV efficiently infects cultured normal human astrocytes, three different murine astrocyte cell lines and astrocytes in the mouse brain. The attenuation of virus replication positively correlates with the increased levels of production of IL-8, IL-17, IFN-gamma and IP-10. In addition, VEEV infection induces release of basic fibroblast growth factor and production of potent chemokines such as RANTES and MIP-1-beta from cultured human astrocytes. This growth factor and cytokine profile modeled by astrocytes in vitro may contribute to both neuroprotection and repair and may play a role in leukocyte recruitment in vivo.
Collapse
Affiliation(s)
- Bi-Hung Peng
- Department of Pathology/Institute for Human Infections and Immunity, Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555-0609, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Dyer KD, Garcia-Crespo KE, Glineur S, Domachowske JB, Rosenberg HF. The Pneumonia Virus of Mice (PVM) model of acute respiratory infection. Viruses 2012; 4:3494-510. [PMID: 23342367 PMCID: PMC3528276 DOI: 10.3390/v4123494] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 11/28/2012] [Accepted: 11/28/2012] [Indexed: 01/16/2023] Open
Abstract
Pneumonia Virus of Mice (PVM) is related to the human and bovine respiratory syncytial virus (RSV) pathogens, and has been used to study respiratory virus replication and the ensuing inflammatory response as a component of a natural host—pathogen relationship. As such, PVM infection in mice reproduces many of the clinical and pathologic features of the more severe forms of RSV infection in human infants. Here we review some of the most recent findings on the basic biology of PVM infection and its use as a model of disease, most notably for explorations of virus infection and allergic airways disease, for vaccine evaluation, and for the development of immunomodulatory strategies for acute respiratory virus infection.
Collapse
Affiliation(s)
- Kimberly D. Dyer
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; E-Mails: (K.E.G.-C.); (S.G.); (H.F.R.)
| | - Katia E. Garcia-Crespo
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; E-Mails: (K.E.G.-C.); (S.G.); (H.F.R.)
| | - Stephanie Glineur
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; E-Mails: (K.E.G.-C.); (S.G.); (H.F.R.)
| | - Joseph B. Domachowske
- Department of Pediatrics, SUNY Upstate Medical University, Syracuse, NY 13210, USA; E-Mail:
| | - Helene F. Rosenberg
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; E-Mails: (K.E.G.-C.); (S.G.); (H.F.R.)
| |
Collapse
|
37
|
Lionakis MS, Fischer BG, Lim JK, Swamydas M, Wan W, Richard Lee CC, Cohen JI, Scheinberg P, Gao JL, Murphy PM. Chemokine receptor Ccr1 drives neutrophil-mediated kidney immunopathology and mortality in invasive candidiasis. PLoS Pathog 2012; 8:e1002865. [PMID: 22916017 PMCID: PMC3420964 DOI: 10.1371/journal.ppat.1002865] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 07/04/2012] [Indexed: 11/23/2022] Open
Abstract
Invasive candidiasis is the 4th leading cause of nosocomial bloodstream infection in the US with mortality that exceeds 40% despite administration of antifungal therapy; neutropenia is a major risk factor for poor outcome after invasive candidiasis. In a fatal mouse model of invasive candidiasis that mimics human bloodstream-derived invasive candidiasis, the most highly infected organ is the kidney and neutrophils are the major cellular mediators of host defense; however, factors regulating neutrophil recruitment have not been previously defined. Here we show that mice lacking chemokine receptor Ccr1, which is widely expressed on leukocytes, had selectively impaired accumulation of neutrophils in the kidney limited to the late phase of the time course of the model; surprisingly, this was associated with improved renal function and survival without affecting tissue fungal burden. Consistent with this, neutrophils from wild-type mice in blood and kidney switched from Ccr1lo to Ccr1high at late time-points post-infection, when Ccr1 ligands were produced at high levels in the kidney and were chemotactic for kidney neutrophils ex vivo. Further, when a 1∶1 mixture of Ccr1+/+ and Ccr1−/− donor neutrophils was adoptively transferred intravenously into Candida-infected Ccr1+/+ recipient mice, neutrophil trafficking into the kidney was significantly skewed toward Ccr1+/+ cells. Thus, neutrophil Ccr1 amplifies late renal immunopathology and increases mortality in invasive candidiasis by mediating excessive recruitment of neutrophils from the blood to the target organ. Invasive infection by the yeast Candida represents a significant cause of morbidity and mortality in patients in the intensive care unit. Neutrophils, which are recruited to sites of Candida infection by chemokines and their receptors, are important immune cells in host defense against invasive candidiasis. Consistent with that, lack of neutrophils is a well-established risk factor for adverse outcome after infection. In this study, we performed a broad survey of the chemokine system in a mouse model of invasive candidiasis with an aim to determine factors that regulate neutrophil trafficking to sites of infection. We used that survey to identify Ccr1 as a mediator of mortality in the model via excessive recruitment of neutrophils from the blood to the kidney that results in kidney tissue injury. Strikingly, the effect of Ccr1 on neutrophil accumulation in the kidney was not seen until the late phase of the infection, when the receptor was up-regulated on the neutrophil surface. Together these data demonstrate that neutrophils, besides their recognized protective roles in antifungal host defense, may also exert detrimental effects by causing uncontrolled tissue damage, and identify Ccr1 as a mediator of neutrophil tissue injury in a mouse model of invasive candidiasis.
Collapse
Affiliation(s)
- Michail S Lionakis
- Clinical Mycology Unit, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Spolski R, Wang L, Wan CK, Bonville CA, Domachowske JB, Kim HP, Yu Z, Leonard WJ. IL-21 promotes the pathologic immune response to pneumovirus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 188:1924-32. [PMID: 22238461 PMCID: PMC3277853 DOI: 10.4049/jimmunol.1100767] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
IL-21 is a cytokine with pleiotropic actions, promoting terminal differentiation of B cells, increased Ig production, and the development of Th17 and T follicular helper cells. IL-21 is also implicated in the development of autoimmune disease and has antitumor activity. In this study, we investigated the role of IL-21 in host defense to pneumonia virus of mice (PVM), which initiates an infection in mice resembling that of respiratory syncytial virus disease in humans. We found that PVM-infected mice expressed IL-21 in lung CD4(+) T cells. Following infection, Il21r(-/-) mice exhibited less lung infiltration by neutrophils than did wild-type (WT) mice and correspondingly had lower levels of the chemokine CXCL1 in bronchoalveolar lavage fluid and lung parenchyma. CD8(+), CD4(+), and γδ T cell numbers were also lower in the lungs of PVM-infected Il21r(-/-) mice than in infected WT mice, with normal Th17 cytokines but diminished IL-6 production in PVM-infected Il21r(-/-) mice. Strikingly, Il21r(-/-) mice had enhanced survival following PVM infection, and moreover, treatment of WT mice with soluble IL-21R-Fc fusion protein enhanced their survival. These data reveal that IL-21 promotes the pathogenic inflammatory effect of PVM and indicate that manipulating IL-21 signaling may represent an immunomodulatory strategy for controlling PVM and potentially other respiratory virus infections.
Collapse
Affiliation(s)
- Rosanne Spolski
- Laboratory of Molecular Immunology National Heart, Lung, and Blood Institute, NIH Bethesda, MD 20892
| | - Lu Wang
- Laboratory of Molecular Immunology National Heart, Lung, and Blood Institute, NIH Bethesda, MD 20892
| | - Chi-Keung Wan
- Laboratory of Molecular Immunology National Heart, Lung, and Blood Institute, NIH Bethesda, MD 20892
| | - Cynthia A. Bonville
- Department of Pediatrics State University of New York Upstate Medical University Syracuse, NY 13210
| | - Joseph B. Domachowske
- Department of Pediatrics State University of New York Upstate Medical University Syracuse, NY 13210
| | - Hyoung-Pyo Kim
- Laboratory of Molecular Immunology National Heart, Lung, and Blood Institute, NIH Bethesda, MD 20892
- Department of Environmental Medical Biology Institute of Tropical Medicine and Brain Korea 21 Project for Medical Science Yonsei University College of Medicine Seoul, Korea
| | - Zuxi Yu
- Pathology Core Facility National Heart, Lung, and Blood Institute, NIH Bethesda, MD 20892
| | - Warren J. Leonard
- Laboratory of Molecular Immunology National Heart, Lung, and Blood Institute, NIH Bethesda, MD 20892
| |
Collapse
|
39
|
Rigaux P, Killoran KE, Qiu Z, Rosenberg HF. Depletion of alveolar macrophages prolongs survival in response to acute pneumovirus infection. Virology 2012; 422:338-45. [PMID: 22129848 PMCID: PMC3256929 DOI: 10.1016/j.virol.2011.10.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 10/24/2011] [Accepted: 10/31/2011] [Indexed: 12/30/2022]
Abstract
Alveolar macrophages are immunoregulatory effector cells that interact directly with respiratory virus pathogens in vivo. We examined the role of alveolar macrophages in acute infection with pneumonia virus of mice (PVM), a rodent pneumovirus that replicates the clinical sequelae of severe human respiratory syncytial virus disease. We show that PVM replicates in primary mouse macrophage culture, releasing infectious virions and proinflammatory cytokines. Alveolar macrophages isolated from PVM-infected mice express activation markers Clec43 and CD86, cytokines TNFα, IL-1, IL-6, and numerous CC and CXC chemokines. Alveolar macrophage depletion prior to PVM infection results in small but statistically significant increases in virus recovery but paradoxically prolonged survival. In parallel, macrophage depleted PVM-infected mice exhibit enhanced NK cell recruitment and increased production of IFNγ by NK, CD4(+) and CD8(+) T cells. These results suggest a protective, immunomodulatory role for IFNγ, as overproduction secondary to macrophage depletion may promote survival despite increased virus recovery.
Collapse
Affiliation(s)
| | | | | | - Helene F. Rosenberg
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
40
|
Bem RA, Domachowske JB, Rosenberg HF. Animal models of human respiratory syncytial virus disease. Am J Physiol Lung Cell Mol Physiol 2011; 301:L148-56. [PMID: 21571908 PMCID: PMC3154630 DOI: 10.1152/ajplung.00065.2011] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 05/05/2011] [Indexed: 11/22/2022] Open
Abstract
Infection with the human pneumovirus pathogen, respiratory syncytial virus (hRSV), causes a wide spectrum of respiratory disease, notably among infants and the elderly. Laboratory animal studies permit detailed experimental modeling of hRSV disease and are therefore indispensable in the search for novel therapies and preventative strategies. Present animal models include several target species for hRSV, including chimpanzees, cattle, sheep, cotton rats, and mice, as well as alternative animal pneumovirus models, such as bovine RSV and pneumonia virus of mice. These diverse animal models reproduce different features of hRSV disease, and their utilization should therefore be based on the scientific hypothesis under investigation. The purpose of this review is to summarize the strengths and limitations of each of these animal models. Our intent is to provide a resource for investigators and an impetus for future research.
Collapse
Affiliation(s)
- Reinout A Bem
- Pediatric Intensive Care Unit, Emma Children’s Hospital, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
41
|
Percopo CM, Dubovi EJ, Renshaw RW, Dyer KD, Domachowske JB, Rosenberg HF. Canine pneumovirus replicates in mouse lung tissue and elicits inflammatory pathology. Virology 2011; 416:26-31. [PMID: 21600624 PMCID: PMC3112256 DOI: 10.1016/j.virol.2011.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 01/30/2011] [Accepted: 04/22/2011] [Indexed: 11/22/2022]
Abstract
Canine pneumovirus (CnPnV) was recently isolated from the respiratory tracts of shelter dogs and shares sequence similarity with the rodent pathogen, pneumonia virus of mice (PVM). We show here that CnPnV replicates in and can elicit local proinflammatory cytokine production and neutrophil recruitment to lung tissue and the airways. In contrast to PVM J3666 infection, fatal CnPnV infections are observed only in response to high titer intranasal inocula (>67 TCID(50) units). Sera from mice that recover from CnPnV infection contain antibodies that cross-react with PVM antigens; these mice are protected against lethal PVM infection. Given these findings, it will be intriguing to determine the relative role(s) of CnPnV and PVM in eliciting respiratory symptoms in susceptible canine species.
Collapse
Affiliation(s)
| | - Edward J. Dubovi
- Animal Health Diagnostic Center, College of Veterinary Medicine at Cornell, Ithaca, NY 14853
| | - Randall W. Renshaw
- Animal Health Diagnostic Center, College of Veterinary Medicine at Cornell, Ithaca, NY 14853
| | - Kimberly D. Dyer
- Laboratory of Allergic Diseases, NIAID, NIH, Bethesda, MD, 20892
| | | | | |
Collapse
|
42
|
van den Berg E, van Woensel JBM, Bos AP, Bem RA, Altemeier WA, Gill SE, Martin TR, Matute-Bello G. Role of the Fas/FasL system in a model of RSV infection in mechanically ventilated mice. Am J Physiol Lung Cell Mol Physiol 2011; 301:L451-60. [PMID: 21743025 DOI: 10.1152/ajplung.00368.2010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Infection with respiratory syncytial virus (RSV) in children can progress to respiratory distress and acute lung injury necessitating mechanical ventilation (MV). MV enhances apoptosis and inflammation in mice infected with pneumonia virus of mice (PVM), a mouse pneumovirus that has been used as a model for severe RSV infection in mice. We hypothesized that the Fas/Fas ligand (FasL) system, a dual proapoptotic/proinflammatory system involved in other forms of lung injury, is required for enhanced lung injury in mechanically ventilated mice infected with PVM. C57BL/6 mice and Fas-deficient ("lpr") mice were inoculated intratracheally with PVM. Seven or eight days after PVM inoculation, the mice were subjected to 4 h of MV (tidal volume 10 ml/kg, fraction of inspired O(2) = 0.21, and positive end-expiratory pressure = 3 cm H(2)O). Seven days after PVM inoculation, exposure to MV resulted in less severe injury in lpr mice than in C57BL/6 mice, as evidenced by decreased numbers of polymorphonuclear neutrophils in the bronchoalveolar lavage (BAL), and lower concentrations of the proinflammatory chemokines KC, macrophage inflammatory protein (MIP)-1α, and MIP-2 in the lungs. However, when PVM infection was allowed to progress one additional day, all of the lpr mice (7/7) died unexpectedly between 0.5 and 3.5 h after the onset of ventilation compared with three of the seven ventilated C57BL/6 mice. Parameters of lung injury were similar in nonventilated mice, as was the viral content in the lungs and other organs. Thus, the Fas/FasL system was partly required for the lung inflammatory response in ventilated mice infected with PVM, but attenuation of lung inflammation did not prevent subsequent mortality.
Collapse
Affiliation(s)
- Elske van den Berg
- Pediatric Intensive Care Unit, Emma Children’s Hospital, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Davidson S, Kaiko G, Loh Z, Lalwani A, Zhang V, Spann K, Foo SY, Hansbro N, Uematsu S, Akira S, Matthaei KI, Rosenberg HF, Foster PS, Phipps S. Plasmacytoid dendritic cells promote host defense against acute pneumovirus infection via the TLR7-MyD88-dependent signaling pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:5938-48. [PMID: 21482736 PMCID: PMC3404606 DOI: 10.4049/jimmunol.1002635] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection in infants. In human infants, plasmacytoid dendritic cells (pDC) are recruited to the nasal compartment during infection and initiate host defense through the secretion of type I IFN, IL-12, and IL-6. However, RSV-infected pDC are refractory to TLR7-mediated activation. In this study, we used the rodent-specific pathogen, pneumonia virus of mice (PVM), to determine the contribution of pDC and TLR7 signaling to the development of the innate inflammatory and early adaptive immune response. In wild-type, but not TLR7- or MyD88-deficient mice, PVM inoculation led to a marked infiltration of pDC and increased expression of type I, II, and III IFNs. The delayed induction of IFNs in the absence of TLR7 or MyD88 was associated with a diminished innate inflammatory response and augmented virus recovery from lung tissue. In the absence of TLR7, PVM-specific CD8(+) T cell cytokine production was abrogated. The adoptive transfer of TLR7-sufficient, but not TLR7-deficient pDC to TLR7 gene-deleted mice recapitulated the antiviral responses observed in wild-type mice and promoted virus clearance. In summary, TLR7-mediated signaling by pDC is required for appropriate innate responses to acute pneumovirus infection. It is conceivable that as-yet-unidentified defects in the TLR7 signaling pathway may be associated with elevated levels of RSV-associated morbidity and mortality among otherwise healthy human infants.
Collapse
Affiliation(s)
- Sophia Davidson
- Centre for Asthma and Respiratory Diseases and Hunter Medical Research Institute, University of Newcastle, NSW, Australia
| | - Gerard Kaiko
- Centre for Asthma and Respiratory Diseases and Hunter Medical Research Institute, University of Newcastle, NSW, Australia
| | - Zhixuan Loh
- School of Biomedical Sciences, The University of Queensland, QLD, Australia
| | - Amit Lalwani
- School of Biomedical Sciences, The University of Queensland, QLD, Australia
| | - Vivian Zhang
- School of Biomedical Sciences, The University of Queensland, QLD, Australia
| | - Kirsten Spann
- Sir Albert Sakzewski Virus Research Centre, The University of Queensland, QLD, Australia
| | - Shen Yun Foo
- Centre for Asthma and Respiratory Diseases and Hunter Medical Research Institute, University of Newcastle, NSW, Australia
| | - Nicole Hansbro
- Centre for Asthma and Respiratory Diseases and Hunter Medical Research Institute, University of Newcastle, NSW, Australia
| | - Satoshi Uematsu
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Klaus I. Matthaei
- Stem Cell and Gene Targeting Laboratory, John Curtin School of Medical Research, The Australian National University, ACT, Australia
- Stem Cell Unit, Department of Anatomy, College of Medicine and King Khalid University Hospital, King Saud University, Riyadh 11461, Kingdom of Saudi Arabia
| | - Helene F. Rosenberg
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Paul S. Foster
- Centre for Asthma and Respiratory Diseases and Hunter Medical Research Institute, University of Newcastle, NSW, Australia
| | - Simon Phipps
- School of Biomedical Sciences, The University of Queensland, QLD, Australia
| |
Collapse
|
44
|
|
45
|
Gabryszewski SJ, Bachar O, Dyer KD, Percopo CM, Killoran KE, Domachowske JB, Rosenberg HF. Lactobacillus-mediated priming of the respiratory mucosa protects against lethal pneumovirus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:1151-61. [PMID: 21169550 PMCID: PMC3404433 DOI: 10.4049/jimmunol.1001751] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The inflammatory response to respiratory virus infection can be complex and refractory to standard therapy. Lactobacilli, when targeted to the respiratory epithelium, are highly effective at suppressing virus-induced inflammation and protecting against lethal disease. Specifically, wild-type mice primed via intranasal inoculation with live or heat-inactivated Lactobacillus plantarum or Lactobacillus reuteri were completely protected against lethal infection with the virulent rodent pathogen, pneumonia virus of mice; significant protection (60% survival) persisted for at least 13 wk. Protection was not unique to Lactobacillus species, and it was also observed in response to priming with nonpathogenic Gram-positive Listeria innocua. Priming with live lactobacilli resulted in diminished granulocyte recruitment, diminished expression of multiple proinflammatory cytokines (CXCL10, CXCL1, CCL2, and TNF), and reduced virus recovery, although we have demonstrated clearly that absolute virus titer does not predict clinical outcome. Lactobacillus priming also resulted in prolonged survival and protection against the lethal sequelae of pneumonia virus of mice infection in MyD88 gene-deleted (MyD88(-/-)) mice, suggesting that the protective mechanisms may be TLR-independent. Most intriguing, virus recovery and cytokine expression patterns in Lactobacillus-primed MyD88(-/-) mice were indistinguishable from those observed in control-primed MyD88(-/-) counterparts. In summary, we have identified and characterized an effective Lactobacillus-mediated innate immune shield, which may ultimately serve as critical and long-term protection against infection in the absence of specific antiviral vaccines.
Collapse
Affiliation(s)
| | - Ofir Bachar
- Eosinophil Biology Section, National Institutes of Health, Bethesda, Maryland, USA
| | - Kimberly D. Dyer
- Eosinophil Biology Section, National Institutes of Health, Bethesda, Maryland, USA
| | - Caroline M. Percopo
- Eosinophil Biology Section, National Institutes of Health, Bethesda, Maryland, USA
| | - Kristin E. Killoran
- Lymphocyte Biology Unit, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Helene F. Rosenberg
- Eosinophil Biology Section, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
46
|
Shamri R, Xenakis JJ, Spencer LA. Eosinophils in innate immunity: an evolving story. Cell Tissue Res 2010; 343:57-83. [PMID: 21042920 DOI: 10.1007/s00441-010-1049-6] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 09/01/2010] [Indexed: 12/27/2022]
Abstract
Eosinophils are innate immune leukocytes found in relatively low numbers within the blood. Terminal effector functions of eosinophils, deriving from their capacity to release their content of tissue-destructive cationic proteins, have historically been considered primary effector mechanisms against specific parasites, and are likewise implicated in tissue damage accompanying allergic responses such as asthma. However, the past decade has seen dramatic advancements in the field of eosinophil immunobiology, revealing eosinophils to also be key participants in many other facets of innate immunity, from bridging innate and adaptive immune responses to orchestrating tissue remodeling events. Here, we review the multifaceted functions of eosinophils in innate immunity that are currently known, and discuss new avenues in this evolving story.
Collapse
Affiliation(s)
- Revital Shamri
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | |
Collapse
|
47
|
Murine lung eosinophil activation and chemokine production in allergic airway inflammation. Cell Mol Immunol 2010; 7:361-74. [PMID: 20622891 DOI: 10.1038/cmi.2010.31] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Eosinophils play important roles in asthma and lung infections. Murine models are widely used for assessing the functional significance and mechanistic basis for eosinophil involvements in these diseases. However, little is known about tissue eosinophils in homeostasis. In addition, little data on eosinophil chemokine production during allergic airway inflammation are available. In this study, the properties and functions of homeostatic and activated eosinophils were compared. Eosinophils from normal tissues expressed costimulation and adhesion molecules B7-1, B7-2 and ICAM-1 for Ag presentation but little major histocompatibility complex (MHC) class II, and were found to be poor stimulators of T-cell proliferation. However, these eosinophils expressed high levels of chemokine mRNA including C10, macrophage inflammatory protein (MIP)-1alpha, MIP-1gamma, MIP-2, eotaxin and monocyte chemoattractant protein-5 (MCP-5), and produced chemokine proteins. Eosinophil intracellular chemokines decreased rapidly with concomitant surface marker downregulation upon in vitro culturing consistent with piecemeal degranulation. Lung eosinophils from mice with induced allergic airway inflammation exhibited increased chemokines mRNA expression and chemokines protein production and upregulated MHC class II and CD11c expression. They were also found to be the predominant producers of the CCR1 ligands CCL6/C10 and CCL9/MIP-1gamma in inflamed lungs. Eosinophil production of C10 and MIP-1gamma correlated with the marked influx of CD11b(high) lung dendritic cells during allergic airway inflammation and the high expression of CCR1 on these dendritic cells (DCs). The study provided baseline information on tissue eosinophils, documented the upregulation of activation markers and chemokine production in activated eosinophils, and indicated that eosinophils were a key chemokine-producing cell type in allergic lung inflammation.
Collapse
|
48
|
Georgiev VS. Pneumonia Virus of Mice (PVM): Exploring Novel Therapeutic Options In a Severe Respiratory Disease Model. NATIONAL INSTITUTE OF ALLERGY AND INFECTIOUS DISEASES, NIH 2010. [PMCID: PMC7176177 DOI: 10.1007/978-1-60761-512-5_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Respiratory syncytial virus (RSV) is the most important respiratory pathogen among infants and toddlers, with infections prevalent and nearly universal in this age group. Severe infections are more common among premature infants, those with cardiac and pulmonary anomalies, and the immunosupressed. Effective prophylactic monoclonal antibody treatment is available for high-risk infants, but there is no effective vaccine. Mouse challenge models have been used for the study of the human RSV pathogen, but the most severe forms of RSV disease are not replicated by this approach. Pneumonia virus of mice (PVM; family Paramyxoviridae, genus Pneumovirus) is a mouse pathogen of the same family as human respiratory syncytial virus. PVM replicates efficiently in mouse-lung epithelial cells in vivo in response to a minimal virus inoculum, and replication is accompanied by local production of proinflammatory cytokines (MIP-1α, MIP-2, MCP-1, and IFN-γ) and granulocyte recruitment to the lung. PVM infection and the ensuing inflammatory response can lead to pulmonary edema and respiratory compromise. Our laboratories have pioneered the use of the PVM model for the study of human clinical disease, which has provided important insights into the role of the inflammatory response in the pathogenesis of severe respiratory virus infection. As part of this work, we have presented several immunomodulatory strategies that clearly reduce morbidity and mortality when administered to PVM infected, symptomatic mice, and thus hold promise as realistic therapeutic strategies for severe RSV infection in human subjects.
Collapse
Affiliation(s)
- Vassil St. Georgiev
- Dept. Health & Human Services, National Institute of Health, Rockledge Drive 6610, Bethesda, 20892 Maryland USA
| |
Collapse
|
49
|
Bem RA, van Woensel JBM, Lutter R, Domachowske JB, Medema JP, Rosenberg HF, Bos AP. Granzyme A- and B-cluster deficiency delays acute lung injury in pneumovirus-infected mice. THE JOURNAL OF IMMUNOLOGY 2009; 184:931-8. [PMID: 20018616 DOI: 10.4049/jimmunol.0903029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Lower respiratory tract infection by the human pneumovirus respiratory syncytial virus is a frequent cause of acute lung injury in children. Severe pneumovirus disease in humans is associated with activation of the granzyme pathway by effector lymphocytes, which may promote pathology by exaggerating proapoptotic caspase activity and proinflammatory activity. The main goal of this study was to determine whether granzymes contribute to the development of acute lung injury in pneumovirus-infected mice. Granzyme-expressing mice and granzyme A- and B-cluster single- and double-knockout mice were inoculated with the rodent pneumovirus pneumonia virus of mice strain J3666, and were studied for markers of lung inflammation and injury. Expression of granzyme A and B is detected in effector lymphocytes in mouse lungs in response to pneumovirus infection. Mice deficient for granzyme A and the granzyme B cluster have unchanged virus titers in the lungs but show a significantly delayed clinical response to fatal pneumovirus infection, a feature that is associated with delayed neutrophil recruitment, diminished activation of caspase-3, and reduced lung permeability. We conclude that granzyme A- and B-cluster deficiency delays the acute progression of pneumovirus disease by reducing alveolar injury.
Collapse
Affiliation(s)
- Reinout A Bem
- Pediatric Intensive Care Unit, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
50
|
Percopo CM, Qiu Z, Phipps S, Foster PS, Domachowske JB, Rosenberg HF. Pulmonary eosinophils and their role in immunopathologic responses to formalin-inactivated pneumonia virus of mice. THE JOURNAL OF IMMUNOLOGY 2009; 183:604-12. [PMID: 19542471 DOI: 10.4049/jimmunol.0802270] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Enhanced disease is the term used to describe the aberrant Th2-skewed responses to naturally acquired human respiratory syncytial virus (hRSV) infection observed in individuals vaccinated with formalin-inactivated viral Ags. Here we explore this paradigm with pneumonia virus of mice (PVM), a pathogen that faithfully reproduces features of severe hRSV infection in a rodent host. We demonstrate that PVM infection in mice vaccinated with formalin-inactivated Ags from PVM-infected cells (PVM Ags) yields Th2-skewed hypersensitivity, analogous to that observed in response to hRSV. Specifically, we detect elevated levels of IL-4, IL-5, IL-13, and eosinophils in bronchoalveolar lavage fluid of PVM-infected mice that were vaccinated with PVM Ags, but not among mice vaccinated with formalin-inactivated Ags from uninfected cells (control Ags). Interestingly, infection in PVM Ag-vaccinated mice was associated with a approximately 10-fold reduction in lung virus titer and protection against weight loss when compared with infected mice vaccinated with control Ags, despite the absence of serum-neutralizing Abs. Given recent findings documenting a role for eosinophils in promoting clearance of hRSV in vivo, we explored the role of eosinophils in altering the pathogenesis of disease with eosinophil-deficient mice. We found that eosinophil deficiency had no impact on virus titer in PVM Ag-vaccinated mice, nor on weight loss or levels of CCL11 (eotaxin-1), IFN-gamma, IL-5, or IL-13 in bronchoalveolar lavage fluid. However, levels of both IL-4 and CCL3 (macrophage inflammatory protein-1alpha) in bronchoalveolar lavage fluid were markedly diminished in PVM Ag-vaccinated, PVM-infected eosinophil-deficient mice when compared with wild-type controls.
Collapse
Affiliation(s)
- Caroline M Percopo
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|