1
|
Bassetti M, Andreoni M, Santus P, Scaglione F. NSAIDs for early management of acute respiratory infections. Curr Opin Infect Dis 2024; 37:304-311. [PMID: 38779903 PMCID: PMC11213495 DOI: 10.1097/qco.0000000000001024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
PURPOSE OF REVIEW To review the rationale for and the potential clinical benefits of an early approach to viral acute respiratory infections with NSAIDs to switch off the inflammatory cascade before the inflammatory process becomes complicated. RECENT FINDINGS It has been shown that in COVID-19 as in other viral respiratory infections proinflammatory cytokines are produced, which are responsible of respiratory and systemic symptoms. There have been concerns that NSAIDs could increase susceptibility to SARS-CoV-2 infection or aggravate COVID-19. However, recent articles reviewing experimental research, observational clinical studies, randomized clinical trials, and meta-analyses conclude that there is no basis to limit the use of NSAIDs, which may instead represent effective self-care measures to control symptoms. SUMMARY The inflammatory response plays a pivotal role in the early phase of acute respiratory tract infections (ARTIs); a correct diagnosis of the cause and a prompt therapeutic approach with NSAIDs may have the potential to control the pathophysiological mechanisms that can complicate the condition, while reducing symptoms to the benefit of the patient. A timely treatment with NSAIDs may limit the inappropriate use of other categories of drugs, such as antibiotics, which are useless when viral cause is confirmed and whose inappropriate use is responsible for the development of resistance.
Collapse
Affiliation(s)
- Matteo Bassetti
- Division of Infectious Diseases, Department of Health Sciences (DISSAL), University of Genova
- IRCCS Ospedale Policlinico San Martino, Genova
| | - Massimo Andreoni
- Infectious Disease Clinic, Policlinico Tor Vergata University Hospital
- Department of System Medicine Tor Vergata, University of Rome, Rome, Italy
| | - Pierachille Santus
- Division of Respiratory Diseases, Ospedale Luigi Sacco, Polo Universitario, ASST Fatebenefratelli-Sacco
- Department of Biomedical and Clinical Sciences (DIBIC), Università Degli Studi di Milano
| | - Francesco Scaglione
- Department of Oncology and Hemato-Oncology, Postgraduate School of Clinical Pharmacology and Toxicology, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
2
|
Koch-Heier J, Vogel AB, Füll Y, Ebensperger M, Schönsiegel A, Zinser RS, Planz O. MEK-inhibitor treatment reduces the induction of regulatory T cells in mice after influenza A virus infection. Front Immunol 2024; 15:1360698. [PMID: 38979428 PMCID: PMC11228811 DOI: 10.3389/fimmu.2024.1360698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 06/10/2024] [Indexed: 07/10/2024] Open
Abstract
Regulatory T cells (Tregs) play a crucial and complex role in balancing the immune response to viral infection. Primarily, they serve to regulate the immune response by limiting the expression of proinflammatory cytokines, reducing inflammation in infected tissue, and limiting virus-specific T cell responses. But excessive activity of Tregs can also be detrimental and hinder the ability to effectively clear viral infection, leading to prolonged disease and potential worsening of disease severity. Not much is known about the impact of Tregs during severe influenza. In the present study, we show that CD4+/CD25+FoxP3+ Tregs are strongly involved in disease progression during influenza A virus (IAV) infection in mice. By comparing sublethal with lethal dose infection in vivo, we found that not the viral load but an increased number of CD4+/CD25+FoxP3+ Tregs may impair the immune response by suppressing virus specific CD8+ T cells and favors disease progression. Moreover, the transfer of induced Tregs into mice with mild disease symptoms had a negative and prolonged effect on disease outcome, emphasizing their importance for pathogenesis. Furthermore, treatment with MEK-inhibitors resulted in a significant reduction of induced Tregs in vitro and in vivo and positively influenced the progression of the disease. Our results demonstrate that CD4+/CD25+FoxP3+ Tregs are involved in the pathogenesis of severe influenza and indicate the potential of the MEK-inhibitor zapnometinib to modulate CD4+/CD25+FoxP3+ Tregs. Thus, making MEK-inhibitors even more promising for the treatment of severe influenza virus infections.
Collapse
Affiliation(s)
- Julia Koch-Heier
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University, Tübingen, Germany
- Atriva Therapeutics GmbH, Tübingen, Germany
| | | | | | | | - Annika Schönsiegel
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University, Tübingen, Germany
- Atriva Therapeutics GmbH, Tübingen, Germany
| | - Raphael S. Zinser
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University, Tübingen, Germany
| | - Oliver Planz
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University, Tübingen, Germany
| |
Collapse
|
3
|
Flynn J, Ahmadi MM, McFarland CT, Kubal MD, Taylor MA, Cheng Z, Torchia EC, Edwards MG. Crowdsourcing temporal transcriptomic coronavirus host infection data: Resources, guide, and novel insights. Biol Methods Protoc 2023; 8:bpad033. [PMID: 38107402 PMCID: PMC10723038 DOI: 10.1093/biomethods/bpad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/07/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023] Open
Abstract
The emergence of severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) reawakened the need to rapidly understand the molecular etiologies, pandemic potential, and prospective treatments of infectious agents. The lack of existing data on SARS-CoV-2 hampered early attempts to treat severe forms of coronavirus disease-2019 (COVID-19) during the pandemic. This study coupled existing transcriptomic data from severe acute respiratory syndrome-related coronavirus 1 (SARS-CoV-1) lung infection animal studies with crowdsourcing statistical approaches to derive temporal meta-signatures of host responses during early viral accumulation and subsequent clearance stages. Unsupervised and supervised machine learning approaches identified top dysregulated genes and potential biomarkers (e.g. CXCL10, BEX2, and ADM). Temporal meta-signatures revealed distinct gene expression programs with biological implications to a series of host responses underlying sustained Cxcl10 expression and Stat signaling. Cell cycle switched from G1/G0 phase genes, early in infection, to a G2/M gene signature during late infection that correlated with the enrichment of DNA damage response and repair genes. The SARS-CoV-1 meta-signatures were shown to closely emulate human SARS-CoV-2 host responses from emerging RNAseq, single cell, and proteomics data with early monocyte-macrophage activation followed by lymphocyte proliferation. The circulatory hormone adrenomedullin was observed as maximally elevated in elderly patients who died from COVID-19. Stage-specific correlations to compounds with potential to treat COVID-19 and future coronavirus infections were in part validated by a subset of twenty-four that are in clinical trials to treat COVID-19. This study represents a roadmap to leverage existing data in the public domain to derive novel molecular and biological insights and potential treatments to emerging human pathogens.
Collapse
Affiliation(s)
- James Flynn
- Illumina Corporation, San Diego, CA 92122, United States
| | - Mehdi M Ahmadi
- Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | | | | | - Mark A Taylor
- Bioinfo Solutions LLC, Parker, CO 80134, United States
| | - Zhang Cheng
- Illumina Corporation, San Diego, CA 92122, United States
| | - Enrique C Torchia
- Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | | |
Collapse
|
4
|
Le CTT, Ahn SY, Ho TL, Lee J, Lee DH, Hwang HS, Kang SM, Ko EJ. Adjuvant effects of combination monophosphoryl lipid A and poly I:C on antigen-specific immune responses and protective efficacy of influenza vaccines. Sci Rep 2023; 13:12231. [PMID: 37507413 PMCID: PMC10382554 DOI: 10.1038/s41598-023-39210-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Toll-like receptor (TLR) agonists improve vaccine immunogenicity and efficacy, but they are currently unlicensed as adjuvants in influenza vaccines. This study aimed to investigate whether a combination of monophosphoryl lipid A (MPL, a TLR4 agonist) and polyriboinosinic polyribocytidylic acid (poly I:C, a TLR3 agonist) can enhance the protective efficacy of an inactivated A/Puerto Rico/8/1934 (A/PR8) H1N1 influenza vaccine against homologous influenza infection and minimize illness outcomes. Results showed that combination MPL and poly I:C adjuvanted influenza vaccination increased the production of antigen-specific antibodies, decreased the levels of cytokines and cellular infiltrates at the infection sites, and induced significant memory T and B cell responses in mice. The results of this study suggest that the combination of MPL and poly I:C can be developed into a possible adjuvant for enhancing the efficacy of influenza vaccines.
Collapse
Affiliation(s)
- Chau Thuy Tien Le
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - So Yeon Ahn
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, Republic of Korea
| | - Thi Len Ho
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea
| | - Jueun Lee
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, Republic of Korea
| | - Dong-Ha Lee
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, Republic of Korea
| | - Hye Suk Hwang
- Department of Biomedical Science, College of Life Science and Industry, Sunchon National University, Suncheon, 57922, Republic of Korea.
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
| | - Eun-Ju Ko
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea.
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, Republic of Korea.
| |
Collapse
|
5
|
Leiva-Rebollo R, Gémez-Mata J, Castro D, Borrego JJ, Labella AM. Immune response of DNA vaccinated-gilthead seabream ( Sparus aurata) against LCDV-Sa infection: relevance of the inflammatory process. Front Immunol 2023; 14:1209926. [PMID: 37346045 PMCID: PMC10279854 DOI: 10.3389/fimmu.2023.1209926] [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: 04/21/2023] [Accepted: 05/22/2023] [Indexed: 06/23/2023] Open
Abstract
Lymphocystis disease is one of the main viral pathologies affecting cultured gilthead seabream (Sparus aurata) in the Mediterranean region. Recently, we have developed a DNA vaccine based on the major capsid protein (MCP) of the Lymphocystis disease virus 3 (LCDV-Sa). The immune response triggered by either LCDV-Sa infection or vaccination have been previously studied and seem to be highly related to the modulation of the inflammatory and the IFN response. However, a comprehensive evaluation of immune-related gene expression in vaccinated fish after viral infection to identify immunogenes involved in vaccine-induced protection have not been carried out to date. The present study aimed to fulfill this objective by analyzing samples of head-kidney, spleen, intestine, and caudal fin from fish using an OpenArray® platform containing targets related to the immune response of gilthead seabream. The results obtained showed an increase of deregulated genes in the hematopoietic organs between vaccinated and non-vaccinated fish. However, in the intestine and fin, the results showed the opposite trend. The global effect of fish vaccination was a significant decrease (p<0.05) of viral replication in groups of fish previously vaccinated, and the expression of the following immune genes related to viral recognition (tlr9), humoral and cellular response (rag1 and cd48), inflammation (csf1r, elam, il1β, and il6), antiviral response (isg15, mx1, mx2, mx3), cell-mediated cytotoxicity (nccrp1), and apoptosis (prf1). The exclusive modulation of the immune response provoked by the vaccination seems to control the progression of the infection in the experimentally challenged gilthead seabream.
Collapse
Affiliation(s)
| | | | | | | | - Alejandro M. Labella
- Department of Microbiology, Faculty of Sciences, University of Malaga, Malaga, Spain
| |
Collapse
|
6
|
Chaudhary R, Meher A, Krishnamoorthy P, Kumar H. Interplay of host and viral factors in inflammatory pathway mediated cytokine storm during RNA virus infection. CURRENT RESEARCH IN IMMUNOLOGY 2023; 4:100062. [PMID: 37273890 PMCID: PMC10238879 DOI: 10.1016/j.crimmu.2023.100062] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 06/06/2023] Open
Abstract
RNA viruses always have been a serious concern for human health by causing several outbreaks, often pandemics. The excessive mortality and deaths associated with the outbreaks caused by these viruses were due to the excessive induction of pro-inflammatory cytokines leading to cytokine storm. Cytokines are important for cell-to-cell communication to maintain cell homeostasis. Disturbances of this homeostasis can lead to intricate chain reactions resulting in a massive release of cytokines. This could lead to a severe self-reinforcement of several feedback processes, which could eventually cause systemic harm, multiple organ failure, or death. Multiple inflammation-associated pathways were involved in the cytokine production and its regulation. Different RNA viruses induce these pathways through the interplay with their viral factors and host proteins and miRNAs regulating these pathways. This review will discuss the interplay of host proteins and miRNAs that can play an important role in the regulation of cytokine storm and the possible therapeutic potential of these molecules for the treatment and the challenges associated with the clinical translation.
Collapse
Affiliation(s)
- Riya Chaudhary
- Department of Biological Sciences, Laboratory of Immunology and Infectious Disease Biology, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, 462066, MP, India
| | - Aparna Meher
- Department of Biological Sciences, Laboratory of Immunology and Infectious Disease Biology, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, 462066, MP, India
| | - Pandikannan Krishnamoorthy
- Department of Biological Sciences, Laboratory of Immunology and Infectious Disease Biology, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, 462066, MP, India
| | - Himanshu Kumar
- Department of Biological Sciences, Laboratory of Immunology and Infectious Disease Biology, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, 462066, MP, India
- Laboratory of Host Defense, WPI Immunology, Frontier Research Centre, Osaka University, Osaka, 5650871, Japan
| |
Collapse
|
7
|
Sorrentino L, Toscanelli W, Fracella M, De Angelis M, Frasca F, Scagnolari C, Petrarca L, Nenna R, Midulla F, Palamara AT, Nencioni L, Pierangeli A. NRF2 Antioxidant Response and Interferon-Stimulated Genes Are Differentially Expressed in Respiratory-Syncytial-Virus- and Rhinovirus-Infected Hospitalized Children. Pathogens 2023; 12:pathogens12040577. [PMID: 37111463 PMCID: PMC10144743 DOI: 10.3390/pathogens12040577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 04/29/2023] Open
Abstract
Respiratory diseases caused by respiratory syncytial virus (RSV) and human rhinovirus (HRV) are frequent causes of the hospitalization of children; nonetheless, RSV is responsible for the most severe and life-threatening illnesses. Viral infection triggers an inflammatory response, activating interferon (IFN)-mediated responses, including IFN-stimulated genes (ISG) expression with antiviral and immunomodulatory activities. In parallel, the reactive oxygen species (ROS) production activates nuclear factor erythroid 2-related factor 2 (NRF2), whose antioxidant activity can reduce inflammation by interacting with the NF-kB pathway and the IFN response. To clarify how the interplay of IFN and NRF2 may impact on clinical severity, we enrolled children hospitalized for bronchiolitis and pneumonia, and measured gene expression of type-I and III IFNs, of several ISGs, of NRF2 and antioxidant-related genes, i.e., glucose-6-phosphate dehydrogenase (G6PD), heme oxygenase 1 (HO1), and NAD(P)H dehydrogenase [Quinone] 1 (NQO1) in RSV- (RSV-A N = 33 and RSV-B N = 30) and HRV (N = 22)-positive respiratory samples. NRF2 and HO1 expression is significantly elevated in children with HRV infection compared to RSV (p = 0.012 and p = 0.007, respectively), whereas ISG15 and ISG56 expression is higher in RSV-infected children (p = 0.016 and p = 0.049, respectively). Children admitted to a pediatric intensive care unit (PICU) had reduced NRF2 expression (p = 0.002). These data suggest, for the first time, that lower activation of the NRF2 antioxidant response in RSV-infected infants may contribute to bronchiolitis severity.
Collapse
Affiliation(s)
- Leonardo Sorrentino
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, 00185 Rome, Italy
| | - Walter Toscanelli
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Matteo Fracella
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, 00185 Rome, Italy
| | - Marta De Angelis
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Federica Frasca
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, 00185 Rome, Italy
| | - Carolina Scagnolari
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, 00185 Rome, Italy
| | - Laura Petrarca
- Department of Maternal Infantile and Urological Sciences, Sapienza University, 00185 Rome, Italy
| | - Raffaella Nenna
- Department of Maternal Infantile and Urological Sciences, Sapienza University, 00185 Rome, Italy
| | - Fabio Midulla
- Department of Maternal Infantile and Urological Sciences, Sapienza University, 00185 Rome, Italy
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Alessandra Pierangeli
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, 00185 Rome, Italy
| |
Collapse
|
8
|
Hulme KD, Noye EC, Short KR, Labzin LI. Dysregulated Inflammation During Obesity: Driving Disease Severity in Influenza Virus and SARS-CoV-2 Infections. Front Immunol 2021; 12:770066. [PMID: 34777390 PMCID: PMC8581451 DOI: 10.3389/fimmu.2021.770066] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
Acute inflammation is a critical host defense response during viral infection. When dysregulated, inflammation drives immunopathology and tissue damage. Excessive, damaging inflammation is a hallmark of both pandemic influenza A virus (IAV) infections and Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) infections. Chronic, low-grade inflammation is also a feature of obesity. In recent years, obesity has been recognized as a growing pandemic with significant mortality and associated costs. Obesity is also an independent risk factor for increased disease severity and death during both IAV and SARS-CoV-2 infection. This review focuses on the effect of obesity on the inflammatory response in the context of viral respiratory infections and how this leads to increased viral pathology. Here, we will review the fundamentals of inflammation, how it is initiated in IAV and SARS-CoV-2 infection and its link to disease severity. We will examine how obesity drives chronic inflammation and trained immunity and how these impact the immune response to IAV and SARS-CoV-2. Finally, we review both medical and non-medical interventions for obesity, how they impact on the inflammatory response and how they could be used to prevent disease severity in obese patients. As projections of global obesity numbers show no sign of slowing down, future pandemic preparedness will require us to consider the metabolic health of the population. Furthermore, if weight-loss alone is insufficient to reduce the risk of increased respiratory virus-related mortality, closer attention must be paid to a patient’s history of health, and new therapeutic options identified.
Collapse
Affiliation(s)
- Katina D Hulme
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Ellesandra C Noye
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Larisa I Labzin
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
9
|
Bender EC, Kraynak CA, Huang W, Suggs LJ. Cell-Inspired Biomaterials for Modulating Inflammation. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:279-294. [PMID: 33528306 DOI: 10.1089/ten.teb.2020.0276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Inflammation is a crucial part of wound healing and pathogen clearance. However, it can also play a role in exacerbating chronic diseases and cancer progression when not regulated properly. A subset of current innate immune engineering research is focused on how molecules such as lipids, proteins, and nucleic acids native to a healthy inflammatory response can be harnessed in the context of biomaterial design to promote healing, decrease disease severity, and prolong survival. The engineered biomaterials in this review inhibit inflammation by releasing anti-inflammatory cytokines, sequestering proinflammatory cytokines, and promoting phenotype switching of macrophages in chronic inflammatory disease models. Conversely, other biomaterials discussed here promote inflammation by mimicking pathogen invasion to inhibit tumor growth in cancer models. The form that these biomaterials take spans a spectrum from nanoparticles to large-scale hydrogels to surface coatings on medical devices. Cell-inspired molecules have been incorporated in a variety of creative ways, including loaded into or onto the surface of biomaterials or used as the biomaterials themselves.
Collapse
Affiliation(s)
- Elizabeth C Bender
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA
| | - Chelsea A Kraynak
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA
| | - Wenbai Huang
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA.,Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas, USA
| | - Laura J Suggs
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
10
|
Clementi N, Ghosh S, De Santis M, Castelli M, Criscuolo E, Zanoni I, Clementi M, Mancini N. Viral Respiratory Pathogens and Lung Injury. Clin Microbiol Rev 2021; 34:e00103-20. [PMID: 33789928 PMCID: PMC8142519 DOI: 10.1128/cmr.00103-20] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Several viruses target the human respiratory tract, causing different clinical manifestations spanning from mild upper airway involvement to life-threatening acute respiratory distress syndrome (ARDS). As dramatically evident in the ongoing SARS-CoV-2 pandemic, the clinical picture is not always easily predictable due to the combined effect of direct viral and indirect patient-specific immune-mediated damage. In this review, we discuss the main RNA (orthomyxoviruses, paramyxoviruses, and coronaviruses) and DNA (adenoviruses, herpesviruses, and bocaviruses) viruses with respiratory tropism and their mechanisms of direct and indirect cell damage. We analyze the thin line existing between a protective immune response, capable of limiting viral replication, and an unbalanced, dysregulated immune activation often leading to the most severe complication. Our comprehension of the molecular mechanisms involved is increasing and this should pave the way for the development and clinical use of new tailored immune-based antiviral strategies.
Collapse
Affiliation(s)
- Nicola Clementi
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
- Laboratory of Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sreya Ghosh
- Harvard Medical School, Boston Children's Hospital, Division of Immunology, Boston, Massachusetts, USA
| | - Maria De Santis
- Department of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| | - Matteo Castelli
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
| | - Elena Criscuolo
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
| | - Ivan Zanoni
- Harvard Medical School, Boston Children's Hospital, Division of Immunology, Boston, Massachusetts, USA
- Harvard Medical School, Boston Children's Hospital, Division of Gastroenterology, Boston, Massachusetts, USA
| | - Massimo Clementi
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
- Laboratory of Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicasio Mancini
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
- Laboratory of Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
11
|
Louboutin L, Cabon J, Vigouroux E, Morin T, Danion M. Comparative analysis of the course of infection and the immune response in rainbow trout (Oncorhynchus mykiss) infected with the 5 genotypes of infectious hematopoietic necrosis virus. Virology 2021; 552:20-31. [DOI: 10.1016/j.virol.2020.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 12/29/2022]
|
12
|
Veenstra KA, Hodneland K, Fischer S, Takehana K, Belmonte R, Fischer U. Cellular Immune Responses in Rainbow Trout ( Onchorhynchus mykiss) Following Vaccination and Challenge Against Salmonid Alphavirus (SAV). Vaccines (Basel) 2020; 8:vaccines8040725. [PMID: 33276596 PMCID: PMC7761581 DOI: 10.3390/vaccines8040725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 01/25/2023] Open
Abstract
Viral disease outbreaks remain a significant limiting factor for aquaculture. The majority of licensed vaccines used in the industry are administered as oil-adjuvanted formulations carrying inactivated whole pathogens. Cell-mediated immune responses, in particular those based on virus-specific cytotoxic T-cells (CTLs) to conventional inactivated oil-based vaccines, are largely unexplored. As vaccines cannot be optimized against viral pathogens if knowledge of host cellular immune mechanisms remains unknown, in this study we examined fundamental cell-mediated immune responses after vaccination of rainbow trout with an oil-adjuvanted inactivated vaccine against salmonid alphavirus (SAV) and after infection with SAV. A unique in vitro model system was developed to examine MHC class I restricted CTL responses in a clonal line of rainbow trout. The levels of cell-mediated cytotoxicity were compared to pathology, virus load, specific antibody response, changes in immune cell populations, and mRNA expression. Our results hint that different protective mechanisms are being triggered by infection compared to vaccination. While vaccination itself did not cause a strong cytotoxic or humoral response, subsequent challenge of vaccinated fish resulted in significantly stronger and faster specific cytotoxicity, alongside reduced viral titers and pathology. Hence, testing a vaccine on the capacity to induce cell-mediated cytotoxicity will still require a challenge test. Examination of cellular markers additionally indicates that the initial innate response induced by the vaccine could play an important role in steering adaptive mechanisms.
Collapse
Affiliation(s)
- Kimberly A. Veenstra
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Infectology, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (K.A.V.); (S.F.)
| | - Kjartan Hodneland
- MSD Animal Health Innovation, Thormøhlens Gate 55, 5006 Bergen, Norway; (K.H.); (R.B.)
| | - Susanne Fischer
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Infectology, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (K.A.V.); (S.F.)
| | - Kota Takehana
- Nagano Prefectural Fisheries Experimental Station, 2871 Oaza-Nakagawate, Akashina, Azumino-shi, Nagano 399-7102, Japan;
| | - Rodrigo Belmonte
- MSD Animal Health Innovation, Thormøhlens Gate 55, 5006 Bergen, Norway; (K.H.); (R.B.)
| | - Uwe Fischer
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Infectology, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (K.A.V.); (S.F.)
- Correspondence: ; Tel.: +49-38351-71175
| |
Collapse
|
13
|
Kraynak CA, Yan DJ, Suggs LJ. Modulating inflammatory macrophages with an apoptotic body-inspired nanoparticle. Acta Biomater 2020; 108:250-260. [PMID: 32251779 DOI: 10.1016/j.actbio.2020.03.041] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/20/2020] [Accepted: 03/27/2020] [Indexed: 01/09/2023]
Abstract
Macrophages play a critical role in the initiation, maintenance, and resolution of inflammation because of their diverse and plastic phenotypic responses to extracellular stimuli. Inflammatory stimuli drive the recruitment and activation of inflammatory (M1) macrophages, capable of significant cytokine production that potentiates inflammation. Local environmental signals including apoptotic cell efferocytosis drive a phenotypic transition toward pro-reparative (M2) macrophages to facilitate the resolution of inflammation. However, prolonged or dysregulated inflammatory macrophage response contributes to many disease states and tissue damage. We have developed a nanoparticle to help resolve macrophage-mediated inflammation by mimicking the anti-inflammatory effect of apoptotic cell engulfment. The nanoparticle, comprised of a poly(lactide-co-glycolide) core, is coated in phosphatidylserine (PS)-supplemented cell plasma membrane to emulate key characteristics of the apoptotic cell surface. These apoptotic body-inspired PS/membrane-coated nanoparticles (PS-MNPs) reduce inflammatory cytokine expression to promote an anti-inflammatory, phenotypic shift in macrophages in vitro, without the use of small molecule inhibitors or other drugs. Specifically, PS-MNP treatment before lipopolysaccharide (LPS)-induced inflammatory challenge resulted in a 2.5-fold reduction in secreted tumor necrosis factor α (TNFα) at 24 h, with co-treatment of PS-MNPs and LPS demonstrating a 5-fold TNFα reduction compared to LPS alone. Reduced TNFα production, as well as gene expression of several pro-inflammatory cytokines, correlated with a reduction in NFκB activation from PS-MNP treatment. The development of a nanoparticle to reduce the production of multiple inflammatory cytokines and transition away from an inflammatory macrophage phenotype, through the use of a physiologic anti-inflammatory pathway, illustrates a new potential strategy in creating anti-inflammatory therapeutics. STATEMENT OF SIGNIFICANCE: Macrophages propagate inflammation as the major source of cytokine production in the body. In inflammatory diseases, pro-inflammatory macrophages persist in the site of inflammation and exacerbate tissue destruction. Current anti-inflammatory drugs have significant drawbacks, including variable response rates and off-target effects. Here, we have developed an apoptotic-body inspired nanoparticle to modulate inflammatory macrophage phenotype. This polymeric nanoparticle is coated with phosphatidylserine-supplemented cell plasma membrane to mimic the anti-inflammatory effect of apoptotic cell engulfment. Nanoparticle delivery reduces inflammatory cytokine production and promotes an anti-inflammatory phenotypic macrophage shift. The capacity of these nanoparticles to help resolve macrophage-mediated inflammation may be a useful tool to study macrophage-apoptotic cell interactions, the role of macrophages in inflammatory diseases, and in the design of anti-inflammatory therapeutics.
Collapse
|
14
|
Leiva-Rebollo R, Labella AM, Borrego JJ, Castro D. Immune gene expression in gilthead seabream (Sparus aurata) after Lymphocystis disease virus (LCDV-Sa) challenge resulting in asymptomatic infection. J Appl Microbiol 2019; 128:41-53. [PMID: 31529740 DOI: 10.1111/jam.14454] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/04/2019] [Accepted: 09/04/2019] [Indexed: 12/27/2022]
Abstract
AIMS To determine the immune gene expression response of gilthead seabream (Sparus aurata) that is experimentally infected with the lymphocystivirus LCDV-Sa. METHODS AND RESULTS Viral DNA and transcripts were detected by qPCR in all samples from fish injected with LCDV-Sa, demonstrating that the virus establish a systemic and asymptomatic infection. The expression of 23 immune-related genes was also analysed by RT-qPCR in the head kidney (HK) and intestine at several times post-infection (dpi). In HK, the expression of five type I interferon (IFN)-related genes (ifn, irf3, mx2, mx3 and isg15), il10 and ck10 was upregulated at 1-3 dpi, while genes related to the inflammation process (tnfα, il1ß, il6, casp1) were not differentially expressed or even downregulated. The expression profile in the intestine was different regarding type I INF-related genes. An upregulated c3 and ighm expression was observed in both HK and intestine at 3-8 dpi. Finally, the transcription of nccrp1 and mhcIIα was induced in HK, whereas tcrβ expression was downregulated in both organs. CONCLUSIONS LCDV-Sa seems to trigger an immune response in gilthead seabream characterized by a partial activation of type I IFN system and a lack of systemic inflammatory response which may be related to viral persistence. SIGNIFICANCE AND IMPACT OF THE STUDY The immune response observed in gilthead seabream infected by LCDV-Sa could be implicated in the establishment of an asymptomatic persistent infection.
Collapse
Affiliation(s)
- R Leiva-Rebollo
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain
| | - A M Labella
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain
| | - J J Borrego
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain
| | - D Castro
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain
| |
Collapse
|
15
|
Dupuy C, Cabon J, Louboutin L, Le Floch S, Morin T, Danion M. Cellular, humoral and molecular responses in rainbow trout (Oncorhynchus mykiss) exposed to a herbicide and subsequently infected with infectious hematopoietic necrosis virus. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 215:105282. [PMID: 31509759 DOI: 10.1016/j.aquatox.2019.105282] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/19/2019] [Accepted: 08/19/2019] [Indexed: 06/10/2023]
Abstract
Aquatic ecosystems are now chronically polluted by a cocktail of many chemical substances. There is now clear evidence of associations between exposure to pollutants and greater susceptibility to pathogens. The aim of the present study was to characterize the defense capacities of rainbow trout (Oncorhynchus mykiss), chronically exposed to pendimethalin (PD), to subsequent experimental challenge with the infectious hematopoietic necrosis virus (IHNV). Immunological responses were examined at different organizational levels, from individuals to gene expression. No negative effects of PD were noted on the Fulton index nor on the liver or spleen somatic indices (LSI; SSI) before viral infection, but the infectious stress seems to generate a weak but significant decrease in Fulton and LSI values, which could be associated with consumption of energy reserves. During the viral challenges, the distribution of cumulative mortality was slightly different between infected groups. The impact of the virus on fish previously contaminated by PD started earlier and lasted longer than controls. The proportion of seropositive fish was lower in the fish group exposed to PD than in the control group, with similar quantities of anti-IHNV antibodies secreted in positive fish, regardless of the treatment. While no significant differences in C3-1 expression levels were detected throughout the experiment, TNF1&2, TLR3, Il-1β and IFN expression levels were increased in all infected fish, but the difference was more significant in fish groups previously exposed to herbicide. On the other hand, β-def expression was decreased in the pendimethalin-IHNV group compared to that in fish only infected by the virus (control-IHNV group).
Collapse
Affiliation(s)
- Célie Dupuy
- French Agency for Food, Environmental and Occupational Health & Safety (ANSES), Ploufragan-Plouzané-Niort Laboratory, Fish Viral Pathology Unit, Technopôle Brest-Iroise, 29280, Plouzané, France; European University of Brittany, France
| | - Joëlle Cabon
- French Agency for Food, Environmental and Occupational Health & Safety (ANSES), Ploufragan-Plouzané-Niort Laboratory, Fish Viral Pathology Unit, Technopôle Brest-Iroise, 29280, Plouzané, France; European University of Brittany, France
| | - Lénaïg Louboutin
- French Agency for Food, Environmental and Occupational Health & Safety (ANSES), Ploufragan-Plouzané-Niort Laboratory, Fish Viral Pathology Unit, Technopôle Brest-Iroise, 29280, Plouzané, France; European University of Brittany, France
| | - Stéphane Le Floch
- Centre of Documentation, Research and Experimentation on Accidental Water Pollution (CEDRE), 715 Rue Alain Colas, 29200, Brest, France
| | - Thierry Morin
- French Agency for Food, Environmental and Occupational Health & Safety (ANSES), Ploufragan-Plouzané-Niort Laboratory, Fish Viral Pathology Unit, Technopôle Brest-Iroise, 29280, Plouzané, France; European University of Brittany, France
| | - Morgane Danion
- French Agency for Food, Environmental and Occupational Health & Safety (ANSES), Ploufragan-Plouzané-Niort Laboratory, Fish Viral Pathology Unit, Technopôle Brest-Iroise, 29280, Plouzané, France; European University of Brittany, France.
| |
Collapse
|
16
|
Abstract
Ebola virus (EBOV) disease (EVD) results from an exacerbated immunological response that is highlighted by a burst in the production of inflammatory mediators known as a "cytokine storm." Previous reports have suggested that nonspecific activation of T lymphocytes may play a central role in this phenomenon. T-cell immunoglobulin and mucin domain-containing protein 1 (Tim-1) has recently been shown to interact with virion-associated phosphatidylserine to promote infection. Here, we demonstrate the central role of Tim-1 in EBOV pathogenesis, as Tim-1-/- mice exhibited increased survival rates and reduced disease severity; surprisingly, only a limited decrease in viremia was detected. Tim-1-/- mice exhibited a modified inflammatory response as evidenced by changes in serum cytokines and activation of T helper subsets. A series of in vitro assays based on the Tim-1 expression profile on T cells demonstrated that despite the apparent absence of detectable viral replication in T lymphocytes, EBOV directly binds to isolated T lymphocytes in a phosphatidylserine-Tim-1-dependent manner. Exposure to EBOV resulted in the rapid development of a CD4Hi CD3Low population, non-antigen-specific activation, and cytokine production. Transcriptome and Western blot analysis of EBOV-stimulated CD4+ T cells confirmed the induction of the Tim-1 signaling pathway. Furthermore, comparative analysis of transcriptome data and cytokine/chemokine analysis of supernatants highlight the similarities associated with EBOV-stimulated T cells and the onset of a cytokine storm. Flow cytometry revealed virtually exclusive binding and activation of central memory CD4+ T cells. These findings provide evidence for the role of Tim-1 in the induction of a cytokine storm phenomenon and the pathogenesis of EVD.IMPORTANCE Ebola virus infection is characterized by a massive release of inflammatory mediators, which has come to be known as a cytokine storm. The severity of the cytokine storm is consistently linked with fatal disease outcome. Previous findings have demonstrated that specific T-cell subsets are key contributors to the onset of a cytokine storm. In this study, we investigated the role of Tim-1, a T-cell-receptor-independent trigger of T-cell activation. We first demonstrated that Tim-1-knockout (KO) mice survive lethal Ebola virus challenge. We then used a series of in vitro assays to demonstrate that Ebola virus directly binds primary T cells in a Tim-1-phosphatidylserine-dependent manner. We noted that binding induces a cytokine storm-like phenomenon and that blocking Tim-1-phosphatidylserine interactions reduces viral binding, T-cell activation, and cytokine production. These findings highlight a previously unknown role of Tim-1 in the development of a cytokine storm and "immune paralysis."
Collapse
|
17
|
Younan P, Iampietro M, Nishida A, Ramanathan P, Santos RI, Dutta M, Lubaki NM, Koup RA, Katze MG, Bukreyev A. Ebola Virus Binding to Tim-1 on T Lymphocytes Induces a Cytokine Storm. mBio 2017. [PMID: 28951472 DOI: 10.1128/mbio.00845-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ebola virus (EBOV) disease (EVD) results from an exacerbated immunological response that is highlighted by a burst in the production of inflammatory mediators known as a "cytokine storm." Previous reports have suggested that nonspecific activation of T lymphocytes may play a central role in this phenomenon. T-cell immunoglobulin and mucin domain-containing protein 1 (Tim-1) has recently been shown to interact with virion-associated phosphatidylserine to promote infection. Here, we demonstrate the central role of Tim-1 in EBOV pathogenesis, as Tim-1-/- mice exhibited increased survival rates and reduced disease severity; surprisingly, only a limited decrease in viremia was detected. Tim-1-/- mice exhibited a modified inflammatory response as evidenced by changes in serum cytokines and activation of T helper subsets. A series of in vitro assays based on the Tim-1 expression profile on T cells demonstrated that despite the apparent absence of detectable viral replication in T lymphocytes, EBOV directly binds to isolated T lymphocytes in a phosphatidylserine-Tim-1-dependent manner. Exposure to EBOV resulted in the rapid development of a CD4Hi CD3Low population, non-antigen-specific activation, and cytokine production. Transcriptome and Western blot analysis of EBOV-stimulated CD4+ T cells confirmed the induction of the Tim-1 signaling pathway. Furthermore, comparative analysis of transcriptome data and cytokine/chemokine analysis of supernatants highlight the similarities associated with EBOV-stimulated T cells and the onset of a cytokine storm. Flow cytometry revealed virtually exclusive binding and activation of central memory CD4+ T cells. These findings provide evidence for the role of Tim-1 in the induction of a cytokine storm phenomenon and the pathogenesis of EVD.IMPORTANCE Ebola virus infection is characterized by a massive release of inflammatory mediators, which has come to be known as a cytokine storm. The severity of the cytokine storm is consistently linked with fatal disease outcome. Previous findings have demonstrated that specific T-cell subsets are key contributors to the onset of a cytokine storm. In this study, we investigated the role of Tim-1, a T-cell-receptor-independent trigger of T-cell activation. We first demonstrated that Tim-1-knockout (KO) mice survive lethal Ebola virus challenge. We then used a series of in vitro assays to demonstrate that Ebola virus directly binds primary T cells in a Tim-1-phosphatidylserine-dependent manner. We noted that binding induces a cytokine storm-like phenomenon and that blocking Tim-1-phosphatidylserine interactions reduces viral binding, T-cell activation, and cytokine production. These findings highlight a previously unknown role of Tim-1 in the development of a cytokine storm and "immune paralysis."
Collapse
Affiliation(s)
- Patrick Younan
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA.,Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA.,The University of Texas Medical Branch, Galveston, Texas, USA
| | - Mathieu Iampietro
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA.,Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA.,The University of Texas Medical Branch, Galveston, Texas, USA
| | - Andrew Nishida
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Palaniappan Ramanathan
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA.,Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA.,The University of Texas Medical Branch, Galveston, Texas, USA
| | - Rodrigo I Santos
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA.,Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA.,The University of Texas Medical Branch, Galveston, Texas, USA
| | - Mukta Dutta
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Ndongala Michel Lubaki
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA.,Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA.,The University of Texas Medical Branch, Galveston, Texas, USA
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael G Katze
- Department of Microbiology, University of Washington, Seattle, Washington, USA.,Washington National Primate Research Center, Seattle, Washington, USA
| | - Alexander Bukreyev
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA .,Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, USA.,Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA.,The University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
18
|
Rösler B, Herold S. Lung epithelial GM-CSF improves host defense function and epithelial repair in influenza virus pneumonia-a new therapeutic strategy? Mol Cell Pediatr 2016; 3:29. [PMID: 27480877 PMCID: PMC4969252 DOI: 10.1186/s40348-016-0055-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/15/2016] [Indexed: 01/05/2023] Open
Abstract
Influenza viruses (IVs) circulate seasonally and are a common cause of respiratory infections in pediatric and adult patients. Additionally, recurrent pandemics cause massive morbidity and mortality worldwide. Infection may result in rapid progressive viral pneumonia with fatal outcome. Since accurate treatment strategies are still missing, research refocuses attention to lung pathology and cellular crosstalk to develop new therapeutic options. Alveolar epithelial cells (AECs) play an important role in orchestrating the pulmonary antiviral host response. After IV infection they release a cascade of immune mediators, one of which is granulocyte and macrophage colony-stimulating factor (GM-CSF). GM-CSF is known to promote differentiation, activation and mobilization of myeloid cells. In the lung, GM-CSF drives immune functions of alveolar macrophages and dendritic cells (DCs) and also improves epithelial repair processes through direct interaction with AECs. During IV infection, AEC-derived GM-CSF shows a lung-protective effect that is also present after local GM-CSF application. This mini-review provides an overview on GM-CSF-modulated immune responses to IV pneumonia and its therapeutic potential in severe IV pneumonia.
Collapse
Affiliation(s)
- Barbara Rösler
- Dr von Hauner Children's Hospital, Ludwig-Maximilians-University, Lindwurmstrasse 4, 80337, Munich, Germany. .,Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany.
| | - Susanne Herold
- Department of Internal Medicine II, Section for Infectious Diseases, University Hospital Giessen and Marburg, Klinikstr. 33, Giessen, 35392, Germany.,Universities Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.,Department of Internal Medicine II, Section for Infectious Diseases, Universities Giessen and Marburg Lung Center (UGMLC), Klinikstr. 33, 35392, Giessen, Germany
| |
Collapse
|
19
|
Robledo D, Taggart JB, Ireland JH, McAndrew BJ, Starkey WG, Haley CS, Hamilton A, Guy DR, Mota-Velasco JC, Gheyas AA, Tinch AE, Verner-Jeffreys DW, Paley RK, Rimmer GSE, Tew IJ, Bishop SC, Bron JE, Houston RD. Gene expression comparison of resistant and susceptible Atlantic salmon fry challenged with Infectious Pancreatic Necrosis virus reveals a marked contrast in immune response. BMC Genomics 2016; 17:279. [PMID: 27066778 PMCID: PMC4827185 DOI: 10.1186/s12864-016-2600-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/22/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Infectious Pancreatic Necrosis (IPN) is a highly contagious birnavirus disease of farmed salmonid fish, which often causes high levels of morbidity and mortality. A large host genetic component to resistance has been previously described for Atlantic salmon (Salmo salar L.), which mediates high mortality rates in some families and zero mortality in others. However, the molecular and immunological basis for this resistance is not yet fully known. This manuscript describes a global comparison of the gene expression profiles of resistant and susceptible Atlantic salmon fry following challenge with the IPN virus. RESULTS Salmon fry from two IPNV-resistant and two IPNV-susceptible full sibling families were challenged with the virus and sampled at 1 day, 7 days and 20 days post-challenge. Significant viral titre was observed in both resistant and susceptible fish at all timepoints, although generally at higher levels in susceptible fish. Gene expression profiles combined with gene ontology and pathway analyses demonstrated that while a clear immune response was observed in both resistant and susceptible fish, there were striking differences between the two phenotypes. The susceptible fish showed marked up-regulation of genes related to cytokine activity and inflammatory response that evidently failed to protect against the virus. In contrast, the resistant fish demonstrated a less pronounced immune response including up-regulation of genes relating to the M2 macrophage system. CONCLUSIONS While only the susceptible phenotype shows appreciable mortality levels, both resistant and susceptible fish can become infected with IPNV. Susceptible fish are characterized by a much larger, yet ineffective, immune response, largely related to cytokine and inflammatory systems. Resistant fish demonstrate a more moderate, putative macrophage-mediated inflammatory response, which may contribute to their survival.
Collapse
Affiliation(s)
- Diego Robledo
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK.,Departamento de Genética, Facultad de Biología, Universidad de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - John B Taggart
- Institute of Aquaculture, School of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK
| | - Jacqueline H Ireland
- Institute of Aquaculture, School of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK
| | - Brendan J McAndrew
- Institute of Aquaculture, School of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK
| | - William G Starkey
- Institute of Aquaculture, School of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK
| | - Chris S Haley
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK
| | - Alastair Hamilton
- Landcatch Natural Selection Ltd., 15 Beta Centre, Stirling University Innovation Park, Stirling, FK9 4NF, UK
| | - Derrick R Guy
- Landcatch Natural Selection Ltd., 15 Beta Centre, Stirling University Innovation Park, Stirling, FK9 4NF, UK
| | - Jose C Mota-Velasco
- Landcatch Natural Selection Ltd., 15 Beta Centre, Stirling University Innovation Park, Stirling, FK9 4NF, UK
| | - Almas A Gheyas
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK.,Landcatch Natural Selection Ltd., 15 Beta Centre, Stirling University Innovation Park, Stirling, FK9 4NF, UK
| | - Alan E Tinch
- Landcatch Natural Selection Ltd., 15 Beta Centre, Stirling University Innovation Park, Stirling, FK9 4NF, UK
| | | | - Richard K Paley
- Centre for Environment, Fisheries and Aquaculture Science (Cefas), Weymouth, DT4 8UB, UK
| | - Georgina S E Rimmer
- Centre for Environment, Fisheries and Aquaculture Science (Cefas), Weymouth, DT4 8UB, UK
| | - Ian J Tew
- Centre for Environment, Fisheries and Aquaculture Science (Cefas), Weymouth, DT4 8UB, UK
| | - Stephen C Bishop
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK
| | - James E Bron
- Institute of Aquaculture, School of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK
| | - Ross D Houston
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK.
| |
Collapse
|
20
|
Schmidt ME, Meynköhn A, Habermann N, Wiskemann J, Oelmann J, Hof H, Wessels S, Klassen O, Debus J, Potthoff K, Steindorf K, Ulrich CM. Resistance Exercise and Inflammation in Breast Cancer Patients Undergoing Adjuvant Radiation Therapy: Mediation Analysis From a Randomized, Controlled Intervention Trial. Int J Radiat Oncol Biol Phys 2016; 94:329-37. [DOI: 10.1016/j.ijrobp.2015.10.058] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/08/2015] [Accepted: 10/26/2015] [Indexed: 12/20/2022]
|
21
|
Glennie S, Gritzfeld JF, Pennington SH, Garner-Jones M, Coombes N, Hopkins MJ, Vadesilho CF, Miyaji EN, Wang D, Wright AD, Collins AM, Gordon SB, Ferreira DM. Modulation of nasopharyngeal innate defenses by viral coinfection predisposes individuals to experimental pneumococcal carriage. Mucosal Immunol 2016; 9:56-67. [PMID: 25921341 PMCID: PMC4703943 DOI: 10.1038/mi.2015.35] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 03/13/2015] [Indexed: 02/04/2023]
Abstract
Increased nasopharyngeal colonization density has been associated with pneumonia. We used experimental human pneumococcal carriage to investigate whether upper respiratory tract viral infection predisposes individuals to carriage. A total of 101 healthy subjects were screened for respiratory virus before pneumococcal intranasal challenge. Virus was associated with increased odds of colonization (75% virus positive became colonized vs. 46% virus-negative subjects; P=0.02). Nasal Factor H (FH) levels were increased in virus-positive subjects and were associated with increased colonization density. Using an in vitro epithelial model we explored the impact of increased mucosal FH in the context of coinfection. Epithelial inflammation and FH binding resulted in increased pneumococcal adherence to the epithelium. Binding was partially blocked by antibodies targeting the FH-binding protein Pneumococcal surface protein C (PspC). PspC epitope mapping revealed individuals lacked antibodies against the FH binding region. We propose that FH binding to PspC in vivo masks this binding site, enabling FH to facilitate pneumococcal/epithelial attachment during viral infection despite the presence of anti-PspC antibodies. We propose that a PspC-based vaccine lacking binding to FH could reduce pneumococcal colonization, and may have enhanced protection in those with underlying viral infection.
Collapse
Affiliation(s)
- S Glennie
- grid.48004.380000 0004 1936 9764Respiratory Infection Group, Liverpool School of Tropical Medicine, Liverpool, UK ,grid.5337.20000 0004 1936 7603Present Address: 7Present address: School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK., ,
| | - J F Gritzfeld
- grid.48004.380000 0004 1936 9764Respiratory Infection Group, Liverpool School of Tropical Medicine, Liverpool, UK
| | - S H Pennington
- grid.48004.380000 0004 1936 9764Respiratory Infection Group, Liverpool School of Tropical Medicine, Liverpool, UK
| | - M Garner-Jones
- grid.48004.380000 0004 1936 9764Respiratory Infection Group, Liverpool School of Tropical Medicine, Liverpool, UK
| | - N Coombes
- grid.48004.380000 0004 1936 9764Respiratory Infection Group, Liverpool School of Tropical Medicine, Liverpool, UK
| | - M J Hopkins
- grid.269741.f0000 0004 0421 1585Liverpool Specialist Virology Centre, Royal Liverpool and Broadgreen University Hospital, Liverpool, UK
| | - C F Vadesilho
- grid.418514.d0000 0001 1702 8585Centro de Biotecnologia, Instituto Butantan, Sao Paulo, Brazil
| | - E N Miyaji
- grid.418514.d0000 0001 1702 8585Centro de Biotecnologia, Instituto Butantan, Sao Paulo, Brazil
| | - D Wang
- grid.48004.380000 0004 1936 9764Tropical Clinical Trial Unit, Liverpool School of Tropical Medicine, Liverpool, UK
| | - A D Wright
- grid.48004.380000 0004 1936 9764Respiratory Infection Group, Liverpool School of Tropical Medicine, Liverpool, UK ,grid.269741.f0000 0004 0421 1585NIHR Royal Liverpool and Broadgreen University Hospital NHS Trust, Liverpool, UK
| | - A M Collins
- grid.48004.380000 0004 1936 9764Respiratory Infection Group, Liverpool School of Tropical Medicine, Liverpool, UK ,grid.269741.f0000 0004 0421 1585NIHR Royal Liverpool and Broadgreen University Hospital NHS Trust, Liverpool, UK
| | - S B Gordon
- grid.48004.380000 0004 1936 9764Respiratory Infection Group, Liverpool School of Tropical Medicine, Liverpool, UK
| | - D M Ferreira
- grid.48004.380000 0004 1936 9764Respiratory Infection Group, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
22
|
Liu Q, Zhou YH, Yang ZQ. The cytokine storm of severe influenza and development of immunomodulatory therapy. Cell Mol Immunol 2016; 13:3-10. [PMID: 26189369 PMCID: PMC4711683 DOI: 10.1038/cmi.2015.74] [Citation(s) in RCA: 496] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Revised: 06/28/2015] [Accepted: 06/28/2015] [Indexed: 12/25/2022] Open
Abstract
Severe influenza remains unusual in its virulence for humans. Complications or ultimately death arising from these infections are often associated with hyperinduction of proinflammatory cytokine production, which is also known as 'cytokine storm'. For this disease, it has been proposed that immunomodulatory therapy may improve the outcome, with or without the combination of antiviral agents. Here, we review the current literature on how various effectors of the immune system initiate the cytokine storm and exacerbate pathological damage in hosts. We also review some of the current immunomodulatory strategies for the treatment of cytokine storms in severe influenza, including corticosteroids, peroxisome proliferator-activated receptor agonists, sphingosine-1-phosphate receptor 1 agonists, cyclooxygenase-2 inhibitors, antioxidants, anti-tumour-necrosis factor therapy, intravenous immunoglobulin therapy, statins, arbidol, herbs, and other potential therapeutic strategies.
Collapse
Affiliation(s)
- Qiang Liu
- The First College of Clinical Medical Science, China Three Gorges University/Yichang Central People's Hospital, Yichang 443000, China
| | - Yuan-hong Zhou
- The First College of Clinical Medical Science, China Three Gorges University/Yichang Central People's Hospital, Yichang 443000, China
| | - Zhan-qiu Yang
- State Key Laboratory of Virology/Institute of Medical Virology, School of Medicine, Wuhan University, Wuhan 430071, China
| |
Collapse
|
23
|
Rehder KJ, Wilson EA, Zimmerman KO, Cunningham CK, Turner DA. Detection of Multiple Respiratory Viruses Associated With Mortality and Severity of Illness in Children. Pediatr Crit Care Med 2015; 16:e201-6. [PMID: 26121097 PMCID: PMC4560664 DOI: 10.1097/pcc.0000000000000492] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Respiratory viral infection is a common source of morbidity and mortality in children. Coinfection with multiple viruses occurs frequently; however, the clinical significance of concomitant viral pathogens is unclear. We hypothesized that presence of more than one respiratory virus is associated with increased morbidity and mortality when compared with children with a single respiratory virus. DESIGN Retrospective cohort study. SETTING A tertiary care hospital. PATIENTS All children at Duke Children's Hospital over a 2-year period with isolation of a virus on an extended viral respiratory panel result. Demographic data, comorbidities, and details of hospital encounter were recorded. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Two hundred thirty-five hospital encounters demonstrated positive extended viral respiratory panels. Immunocompromised status (37%) and respiratory comorbidities (23%) were common. Twenty-eight patients (12%) tested positive for multiple viruses, with adenovirus (23/28) and respiratory syncytial virus (15/28) most prevalent in patients with multiple viruses. Viral codetection was associated with increased use of noninvasive ventilation (p = 0.02), extracorporeal membrane oxygenation (p = 0.02), increased likelihood of moderate or severe illness (p = 0.005), and increased mortality (p = 0.01). Subgroup analysis demonstrated that this mortality association persisted for children with normal immune function (p = 0.003) and children with no comorbidities (p = 0.007). CONCLUSIONS Children with multiple respiratory viruses may be at increased risk of moderate or severe illness and mortality, with previously healthy children potentially being at greatest risk. Further studies are indicated to determine the significance and generalizability of this finding and to better understand the pathophysiology of viral coinfection.
Collapse
Affiliation(s)
- Kyle J Rehder
- 1Division of Pediatric Critical Care Medicine, Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center, Durham, NC. 2Division of Critical Care, Children's National Medical Center, Washington, DC. 3Division of Infectious Diseases, Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center, Durham, NC
| | | | | | | | | |
Collapse
|
24
|
Meng H, Lee Y, Ba Z, Fleming JA, Furumoto EJ, Roberts RF, Kris-Etherton PM, Rogers CJ. In vitro Production of IL-6 and IFN-γ is Influenced by Dietary Variables and Predicts Upper Respiratory Tract Infection Incidence and Severity Respectively in Young Adults. Front Immunol 2015; 6:94. [PMID: 25788896 PMCID: PMC4349184 DOI: 10.3389/fimmu.2015.00094] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/18/2015] [Indexed: 12/11/2022] Open
Abstract
Assessment of immune responses in healthy adults following dietary or lifestyle interventions is challenging due to significant inter-individual variability. Thus, gaining a better understanding of host factors that contribute to the heterogeneity in immunity is necessary. To address this question, healthy adults [n = 36, 18-40 years old, body mass index (BMI) 20-35 kg/m(2)] were recruited. Dietary intake was obtained via 3-day dietary recall records, physical activity level was evaluated using the International Physical Activity Questionnaire, and peripheral blood mononuclear cells were isolated from peripheral blood. Expression of activation markers on unstimulated immune subsets was assessed by flow cytometry. T-cell proliferation and cytokine secretion was assessed following in vitro stimulation with anti-CD3 or lipopolysaccharide. Furthermore, the incidence and severity of cold or flu symptoms were obtained from self-reported upper respiratory tract infection (URTI) questionnaires. The relationship between activation marker expression on T cells and T-cell effector functions; and in vitro cytokine secretion and URTI was determined by linear or logistic regression. CD69 and CD25 expression on unstimulated T cells was significantly associated with T-cell proliferation and interleukin-2 secretion. Incidence and severity of cold or flu symptoms was significantly associated with in vitro interleukin-6 and interferon-gamma secretion, respectively. Furthermore, host factors (e.g., age, BMI, physical activity, and diet) contributed significantly to the relationship between activation marker expression and T-cell effector function, and cytokine secretion and cold and flu status. In conclusion, these results suggest that lifestyle and dietary factors are important variables that contribute to immune responses and should be included in human clinical trials that assess immune endpoints.
Collapse
Affiliation(s)
- Huicui Meng
- Department of Nutritional Sciences, Pennsylvania State University , University Park, PA , USA
| | - Yujin Lee
- Department of Nutritional Sciences, Pennsylvania State University , University Park, PA , USA
| | - Zhaoyong Ba
- Department of Food Science, Pennsylvania State University , University Park, PA , USA
| | - Jennifer A Fleming
- Department of Nutritional Sciences, Pennsylvania State University , University Park, PA , USA
| | - Emily J Furumoto
- Department of Food Science, Pennsylvania State University , University Park, PA , USA
| | - Robert F Roberts
- Department of Food Science, Pennsylvania State University , University Park, PA , USA
| | - Penny M Kris-Etherton
- Department of Nutritional Sciences, Pennsylvania State University , University Park, PA , USA
| | - Connie J Rogers
- Department of Nutritional Sciences, Pennsylvania State University , University Park, PA , USA
| |
Collapse
|
25
|
Richert LE, Harmsen AL, Rynda-Apple A, Wiley JA, Servid AE, Douglas T, Harmsen AG. Inducible bronchus-associated lymphoid tissue (iBALT) synergizes with local lymph nodes during antiviral CD4+ T cell responses. Lymphat Res Biol 2014; 11:196-202. [PMID: 24364842 DOI: 10.1089/lrb.2013.0015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Exposure of the lungs to an antigen or pathogen elicits the formation of lymphoid satellite islands termed inducible bronchus-associated lymphoid tissue (iBALT). However, little is known about how the presence of iBALT, induced by a stimulus unrelated to the subsequent challenge agent, influences systemic immunity in distal locations, whether it be independently, antagonistically, or synergistically. Here, we determined the kinetics of the influenza-specific responses in the iBALT, tracheobronchial lymph node (TBLN), and spleen of mice with and without pre-formed iBALT. METHODS AND RESULTS Mice with VLP-induced iBALT or no pre-formed iBALT were challenged with influenza. We found that, as we have previously described, those mice whose lungs contained pre-formed iBALT were protected from morbidity, and furthermore, that these mice had increased dendritic cell, and alveolar macrophage accumulation in both the iBALT and TBLNs. This translated to similarly accelerated kinetics and intensified influenza-specific CD4(+), but not CD8(+) T cell responses in the iBALT, TBLN, and spleen. This expansion was then followed by a more rapid T cell contraction in all lymphoid tissues in the mice with pre-formed iBALT. CONCLUSIONS Thus, iBALT itself may not be responsible for the accelerated primary immune response we observe in mice with pre-formed iBALT, but may contribute to an overall accelerated local and systemic primary CD4(+), but not CD8(+) T cell response. Furthermore, less damaging immune responses observed in mice with pre-formed iBALT may be due to a quicker contraction of CD4(+) T cell responses in both local and systemic secondary lymphoid tissue.
Collapse
Affiliation(s)
- Laura E Richert
- 1 Department of Immunology and Infectious Diseases, Montana State University , Bozeman, Montana
| | | | | | | | | | | | | |
Collapse
|
26
|
Collet B. Innate immune responses of salmonid fish to viral infections. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 43:160-73. [PMID: 23981327 DOI: 10.1016/j.dci.2013.08.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 08/19/2013] [Accepted: 08/19/2013] [Indexed: 05/07/2023]
Abstract
Viruses are the most serious pathogenic threat to the production of the main aquacultured salmonid species the rainbow trout Oncorhynchus mykiss and the Atlantic salmon Salmo salar. The viral diseases Infectious Pancreatic Necrosis (IPN), Pancreatic Disease (PD), Infectious Haemorrhagic Necrosis (IHN), Viral Haemorrhagic Septicaemia (VHS), and Infectious Salmon Anaemia (ISA) cause massive economic losses to the global salmonid aquaculture industry every year. To date, no solution exists to treat livestock affected by a viral disease and only a small number of efficient vaccines are available to prevent infection. As a consequence, understanding the host immune response against viruses in these fish species is critical to develop prophylactic and preventive control measures. The innate immune response represents an important part of the host defence mechanism preventing viral replication after infection. It is a fast acting response designed to inhibit virus propagation immediately within the host, allowing for the adaptive specific immunity to develop. It has cellular and humoral components which act in synergy. This review will cover inflammation responses, the cell types involved, apoptosis, antimicrobial peptides. Particular attention will be given to the type I interferon system as the major player in the innate antiviral defence mechanism of salmonids. Viral evasion strategies will also be discussed.
Collapse
|
27
|
Richert LE, Rynda-Apple A, Harmsen AL, Han S, Wiley JA, Douglas T, Larson K, Morton RV, Harmsen AG. CD11c⁺ cells primed with unrelated antigens facilitate an accelerated immune response to influenza virus in mice. Eur J Immunol 2014; 44:397-408. [PMID: 24222381 PMCID: PMC3926668 DOI: 10.1002/eji.201343587] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 09/17/2013] [Accepted: 11/06/2013] [Indexed: 12/24/2022]
Abstract
Recent evidence suggests that an individual's unique history and sequence of exposures to pathogens and antigens may dictate downstream immune responses to disparate antigens. We show that the i.n. delivery of nonreplicative virus-like particles (VLPs), which bear structural but no antigenic similarities to respiratory pathogens, acts to prime the lungs of both C56BL/6 and BALB/c mice, facilitating heightened and accelerated primary immune responses to high-dose influenza challenge, thus providing a nonpathogenic model of innate imprinting. These responses correspond closely to those observed following natural infection with the opportunistic fungus, Pneumocystis murina, and are characterized by accelerated antigen processing by DCs and alveolar macrophages, an enhanced influx of cells to the local tracheobronchial lymph node, and early upregulation of T-cell co-stimulatory/adhesion molecules. CD11c⁺ cells, which have been directly exposed to VLPs or Pneumocystis are necessary in facilitating enhanced clearance of influenza virus, and the repopulation of the lung by Ly-6C⁺ precursors relies on CCR2 expression. Thus, immune imprinting 72 h after VLP-priming, or 2 weeks after Pneumocystis-priming is CCR2-mediated and results from the enhanced antigen processing, maturation, and trafficking abilities of DCs and alveolar macrophages, which cause accelerated influenza-specific primary immune responses and result in superior viral clearance.
Collapse
Affiliation(s)
- Laura E. Richert
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, USA
| | - Agnieszka Rynda-Apple
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, USA
| | - Ann L. Harmsen
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, USA
| | - Soo Han
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, USA
| | - James A. Wiley
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, USA
| | - Trevor Douglas
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, USA
| | - Kyle Larson
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, USA
| | - Rachelle V. Morton
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, USA
| | - Allen G. Harmsen
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
28
|
Pothlichet J, Meunier I, Davis BK, Ting JPY, Skamene E, von Messling V, Vidal SM. Type I IFN triggers RIG-I/TLR3/NLRP3-dependent inflammasome activation in influenza A virus infected cells. PLoS Pathog 2013; 9:e1003256. [PMID: 23592984 PMCID: PMC3623797 DOI: 10.1371/journal.ppat.1003256] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 02/04/2013] [Indexed: 12/25/2022] Open
Abstract
Influenza A virus (IAV) triggers a contagious and potentially lethal respiratory disease. A protective IL-1β response is mediated by innate receptors in macrophages and lung epithelial cells. NLRP3 is crucial in macrophages; however, which sensors elicit IL-1β secretion in lung epithelial cells remains undetermined. Here, we describe for the first time the relative roles of the host innate receptors RIG-I (DDX58), TLR3, and NLRP3 in the IL-1β response to IAV in primary lung epithelial cells. To activate IL-1β secretion, these cells employ partially redundant recognition mechanisms that differ from those described in macrophages. RIG-I had the strongest effect through a MAVS/TRIM25/Riplet-dependent type I IFN signaling pathway upstream of TLR3 and NLRP3. Notably, RIG-I also activated the inflammasome through interaction with caspase 1 and ASC in primary lung epithelial cells. Thus, NS1, an influenza virulence factor that inhibits the RIG-I/type I IFN pathway, strongly modulated the IL-1β response in lung epithelial cells and in ferrets. The NS1 protein derived from a highly pathogenic strain resulted in increased interaction with RIG-I and inhibited type I IFN and IL-1β responses compared to the least pathogenic virus strains. These findings demonstrate that in IAV-infected lung epithelial cells RIG-I activates the inflammasome both directly and through a type I IFN positive feedback loop.
Collapse
Affiliation(s)
- Julien Pothlichet
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- McGill Centre for the Study of Host Resistance, McGill University, Montreal, Quebec, Canada
- Institut Pasteur, Centre François Jacob, Paris, France
| | | | - Beckley K. Davis
- Department of Microbiology and Immunology School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jenny P-Y. Ting
- Department of Microbiology and Immunology School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Emil Skamene
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- McGill Centre for the Study of Host Resistance, McGill University, Montreal, Quebec, Canada
| | | | - Silvia M. Vidal
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- McGill Centre for the Study of Host Resistance, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
29
|
Abstract
The substantial increase in the worldwide prevalence of asthma and atopy has been attributed to lifestyle changes that reduce exposure to bacteria. A recent insight is that the largely bacterial microbiome maintains a state of basal immune homoeostasis, which modulates immune responses to microbial pathogens. However, some respiratory viral infections cause bronchiolitis of infancy and childhood wheeze, and can exacerbate established asthma; whereas allergens can partly mimic infectious agents. New insights into the host’s innate sensing systems, combined with recently developed methods that characterise commensal and pathogenic microbial exposure, now allow a unified theory for how microbes cause mucosal inflammation in asthma. The respiratory mucosa provides a key microbial interface where epithelial and dendritic cells interact with a range of functionally distinct lymphocytes. Lymphoid cells then control a range of pathways, both innate and specific, which organise the host mucosal immune response. Fundamental to innate immune responses to microbes are the interactions between pathogen-associated molecular patterns and pattern recognition receptors, which are associated with production of type I interferons, proinflammatory cytokines, and the T-helper-2 cell pathway in predisposed people. These coordinated, dynamic immune responses underlie the differing asthma phenotypes, which we delineate in terms of Seven Ages of Asthma. An understanding of the role of microbes in the atopic march towards asthma, and in causing exacerbations of established asthma, provides the rationale for new specific treatments that can be assessed in clinical trials. On the basis of these new ideas, specific host biomarkers might then allow personalised treatment to become a reality for patients with asthma.
Collapse
Affiliation(s)
- Trevor T Hansel
- National Heart and Lung Institute, Centre for Respiratory Infection, MRC, London, UK.
| | | | | |
Collapse
|
30
|
Unkel B, Hoegner K, Clausen BE, Lewe-Schlosser P, Bodner J, Gattenloehner S, Janßen H, Seeger W, Lohmeyer J, Herold S. Alveolar epithelial cells orchestrate DC function in murine viral pneumonia. J Clin Invest 2012; 122:3652-64. [PMID: 22996662 DOI: 10.1172/jci62139] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 07/19/2012] [Indexed: 12/23/2022] Open
Abstract
Influenza viruses (IVs) cause pneumonia in humans with progression to lung failure. Pulmonary DCs are key players in the antiviral immune response, which is crucial to restore alveolar barrier function. The mechanisms of expansion and activation of pulmonary DC populations in lung infection remain widely elusive. Using mouse BM chimeric and cell-specific depletion approaches, we demonstrated that alveolar epithelial cell (AEC) GM-CSF mediates recovery from IV-induced injury by affecting lung DC function. Epithelial GM-CSF induced the recruitment of CD11b+ and monocyte-derived DCs. GM-CSF was also required for the presence of CD103+ DCs in the lung parenchyma at baseline and for their sufficient activation and migration to the draining mediastinal lymph nodes (MLNs) during IV infection. These activated CD103+ DCs were indispensable for sufficient clearance of IVs by CD8+ T cells and for recovery from IV-induced lung injury. Moreover, GM-CSF applied intratracheally activated CD103+ DCs, inducing increased migration to MLNs, enhanced viral clearance, and attenuated lung injury. Together, our data reveal that GM-CSF-dependent cross-talk between IV-infected AECs and CD103+ DCs is crucial for effective viral clearance and recovery from injury, which has potential implications for GM-CSF treatment in severe IV pneumonia.
Collapse
Affiliation(s)
- Barbara Unkel
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center, Giessen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
The cytokine storm has captured the attention of the public and the scientific community alike, and while the general notion of an excessive or uncontrolled release of proinflammatory cytokines is well known, the concept of a cytokine storm and the biological consequences of cytokine overproduction are not clearly defined. Cytokine storms are associated with a wide variety of infectious and noninfectious diseases. The term was popularized largely in the context of avian H5N1 influenza virus infection, bringing the term into popular media. In this review, we focus on the cytokine storm in the context of virus infection, and we highlight how high-throughput genomic methods are revealing the importance of the kinetics of cytokine gene expression and the remarkable degree of redundancy and overlap in cytokine signaling. We also address evidence for and against the role of the cytokine storm in the pathology of clinical and infectious disease and discuss why it has been so difficult to use knowledge of the cytokine storm and immunomodulatory therapies to improve the clinical outcomes for patients with severe acute infections.
Collapse
|
32
|
Hussell T, Godlee A, Salek-Ardakani S, Snelgrove RJ. Respiratory viral infections: knowledge based therapeutics. Curr Opin Immunol 2012; 24:438-43. [PMID: 22770666 DOI: 10.1016/j.coi.2012.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 05/15/2012] [Accepted: 06/06/2012] [Indexed: 12/13/2022]
Abstract
Lung viral infections continue to kill millions of people worldwide. Virus-specific properties, replication kinetics and longevity affect the subsequent vigour of innate and adaptive immunity, which contribute to clinical manifestations. The point at which lung innate immunity activates is different between individuals and is determined by age, genetics, underlying conditions and infection history. On resolution of virus-induced lung disease an 'altered state of homeostasis' exists that in turn affects the next antigenic challenge. The last two years has produced a plethora of studies on the resolution of inflammatory lung disease; highlighting potential for immune modulation. In the future a more precise etiological diagnosis, combined with a knowledge of co-morbidities and an immune signature will lead to the development of more specifically targeted therapeutics.
Collapse
Affiliation(s)
- Tracy Hussell
- Imperial College London, Leukocyte Biology Section, National Heart and Lung Institute, London SW7 2AZ, UK.
| | | | | | | |
Collapse
|
33
|
Plasticity and virus specificity of the airway epithelial cell immune response during respiratory virus infection. J Virol 2012; 86:5422-36. [PMID: 22398282 DOI: 10.1128/jvi.06757-11] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Airway epithelial cells (AECs) provide the first line of defense in the respiratory tract and are the main target of respiratory viruses. Here, using oligonucleotide and protein arrays, we analyze the infection of primary polarized human AEC cultures with influenza virus and respiratory syncytial virus (RSV), and we show that the immune response of AECs is quantitatively and qualitatively virus specific. Differentially expressed genes (DEGs) specifically induced by influenza virus and not by RSV included those encoding interferon B1 (IFN-B1), type III interferons (interleukin 28A [IL-28A], IL-28B, and IL-29), interleukins (IL-6, IL-1A, IL-1B, IL-23A, IL-17C, and IL-32), and chemokines (CCL2, CCL8, and CXCL5). Lack of type I interferon or STAT1 signaling decreased the expression and secretion of cytokines and chemokines by the airway epithelium. We also observed strong basolateral polarization of the secretion of cytokines and chemokines by human and murine AECs during infection. Importantly, the antiviral response of human AECs to influenza virus or to RSV correlated with the infection signature obtained from peripheral blood mononuclear cells (PBMCs) isolated from patients with acute influenza or RSV bronchiolitis, respectively. IFI27 (also known as ISG12) was identified as a biomarker of respiratory virus infection in both AECs and PBMCs. In addition, the extent of the transcriptional perturbation in PBMCs correlated with the clinical disease severity. Our results demonstrate that the human airway epithelium mounts virus-specific immune responses that are likely to determine the subsequent systemic immune responses and suggest that the absence of epithelial immune mediators after RSV infection may contribute to explaining the inadequacy of systemic immunity to the virus.
Collapse
|
34
|
Gardner EM, Beli E, Clinthorne JF, Duriancik DM. Energy intake and response to infection with influenza. Annu Rev Nutr 2011; 31:353-67. [PMID: 21548773 DOI: 10.1146/annurev-nutr-081810-160812] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Influenza is a worldwide public health concern, particularly with emerging new strains of influenza to which vaccines are ineffective, limited, or unavailable. In addition, the relationship between adequate nutrition and immune function has been repeatedly demonstrated. Mouse models provide strong evidence that energy extremes, including energy restriction (ER) and diet-induced obesity (DIO), have deleterious effects on the immune response to influenza infection. Both ER and DIO mice demonstrate increased susceptibility and mortality to influenza infection. The effects of ER are more pronounced during innate responses to influenza infection, whereas the effects of DIO are evidenced during innate and adaptive responses to both primary and secondary infection. There are striking similarities between ER and DIO during influenza infection, including impaired natural killer cell function and altered inflammation. Future studies must develop effective nutritional paradigms to offset the effects of these energy extremes on the immune response to an acute infection.
Collapse
Affiliation(s)
- Elizabeth M Gardner
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48823, USA.
| | | | | | | |
Collapse
|
35
|
Cormet-Boyaka E, Jolivette K, Bonnegarde-Bernard A, Rennolds J, Hassan F, Mehta P, Tridandapani S, Webster-Marketon J, Boyaka PN. An NF-κB-independent and Erk1/2-dependent mechanism controls CXCL8/IL-8 responses of airway epithelial cells to cadmium. Toxicol Sci 2011; 125:418-29. [PMID: 22094458 DOI: 10.1093/toxsci/kfr310] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Airway epithelial cells in the lung are the first line of defense against pathogens and environmental pollutants. Inhalation of the environmental pollutant cadmium has been linked to the development of lung cancer and chronic obstructive pulmonary disease, which are diseases characterized by chronic inflammation. To address the role of airway epithelial cells in cadmium-induced lung inflammation, we investigated how cadmium regulates secretion of interleukin 8 (IL-8) by airway epithelial cells. We show that exposure of human airway epithelial cells to subtoxic doses of cadmium in vitro promotes a characteristic inflammatory cytokine response consisting of IL-8, but not IL-1β or tumor necrosis factor-alpha. We also found that intranasal delivery of cadmium increases lung levels of the murine IL-8 homologs macrophage inflammatory protein-2 and keracinocyte-derived chemokine and results in an influx of Gr1+ cells into the lung. We determined that inhibition of the nuclear factor-κB (NF-κB) pathway had no effect on cadmium-induced IL-8 secretion by human airway epithelial cells, suggesting that IL-8 production was mediated through an NF-κB-independent pathway. Mitogen-activated protein kinases (MAPKs) are often involved in proinflammatory signaling. Cadmium could activate the main MAPKs (i.e., p38, JNK, and Erk1/2) in human airway epithelial cells. However, only pharmacological inhibition of Erk1/2 pathway or knockdown of the expression of Erk1 and Erk2 using small interfering RNAs suppressed secretion of IL-8 induced by cadmium. Our findings identify cadmium as a potent activator of the proinflammatory cytokine IL-8 in lung epithelial cells and reveal for the first time the role of an NF-κB-independent but Erk1/2-dependent pathway in cadmium-induced lung inflammation.
Collapse
Affiliation(s)
- Estelle Cormet-Boyaka
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Alleva LM, Gualano RC, Clark IA. Current work and future possibilities for the management of severe influenza: using immunomodulatory agents that target the host response. Future Virol 2011. [DOI: 10.2217/fvl.11.51] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this article, we argue the case that the excessive inflammatory response seen in severe influenza contributes to severe illness and death by disabling oxidative phosphorylation in mitochondria, leading to reduced cellular levels of ATP. When the mitochondrial permeability transition is induced, cells cannot die by apoptosis in the face of reduced ATP levels, because apoptosis depends upon ATP availability, and so cells undergo necrosis. Cellular necrosis causes release of proinflammatory molecules such as high mobility group box 1 protein and mitochondrial DNA, and these could contribute to the prolongation of inflammation during severe influenza. With these concepts in mind, we discuss how immunomodulatory agents that prevent cellular necrosis (by restoring mitochondrial function) and limit inflammation are promising influenza treatments.
Collapse
Affiliation(s)
| | - Rosa C Gualano
- Department of Pharmacology, The University of Melbourne, Parkville VIC 3010, Australia
| | - Ian A Clark
- Division of Biomedical Science & Biochemistry, Research School of Biology, The Australian National University, Canberra ACT 0200, Australia
| |
Collapse
|
37
|
Snelgrove RJ, Godlee A, Hussell T. Airway immune homeostasis and implications for influenza-induced inflammation. Trends Immunol 2011; 32:328-34. [PMID: 21612981 DOI: 10.1016/j.it.2011.04.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 04/15/2011] [Accepted: 04/18/2011] [Indexed: 10/18/2022]
Abstract
The lung is exposed to a myriad of innocuous antigens on a daily basis and must maintain a state of immune ignorance or tolerance to these harmless stimuli to retain pulmonary homeostasis and to prevent potentially fatal immunopathology. Here, we examine how, in the lower airways, resident cell populations contribute to the immune regulatory strategies that restrain inflammation. During influenza infection, these suppressive signals must be overcome to elicit a protective immune response that eliminates the virus. We also discuss how, after resolution of infection, the lung does not return to the original homeostatic state, and how the induced altered state can persist for long periods, which leaves the lung more susceptible to other infectious insults.
Collapse
Affiliation(s)
- Robert J Snelgrove
- Imperial College London, Leukocyte Biology Section, National Heart and Lung Institute, London, SW7 2AZ, UK
| | | | | |
Collapse
|
38
|
Viral replication and innate host responses in primary human alveolar epithelial cells and alveolar macrophages infected with influenza H5N1 and H1N1 viruses. J Virol 2011; 85:6844-55. [PMID: 21543489 DOI: 10.1128/jvi.02200-10] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Highly pathogenic influenza H5N1 virus continues to pose a threat to public health. Although the mechanisms underlying the pathogenesis of the H5N1 virus have not been fully defined, it has been suggested that cytokine dysregulation plays an important role. As the human respiratory epithelium is the primary target cell for influenza viruses, elucidating the viral tropism and innate immune responses of influenza H5N1 virus in the alveolar epithelium may help us to understand the pathogenesis of the severe pneumonia associated with H5N1 disease. Here we used primary cultures of differentiated human alveolar type II cells, alveolar type I-like cells, and alveolar macrophages isolated from the same individual to investigate viral replication competence and host innate immune responses to influenza H5N1 (A/HK/483/97) and H1N1 (A/HK/54/98) virus infection. The viral replication kinetics and cytokine and chemokine responses were compared by quantitative PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA). We demonstrated that influenza H1N1 and H5N1 viruses replicated productively in type II cells and type I-like cells although with different kinetics. The H5N1 virus replicated productively in alveolar macrophages, whereas the H1N1 virus led to an abortive infection. The H5N1 virus was a more potent inducer of proinflammatory cytokines and chemokines than the H1N1 virus in all cell types. However, higher levels of cytokine expression were observed for peripheral blood monocyte-derived macrophages than for alveolar macrophages in response to H5N1 virus infection. Our findings provide important insights into the viral tropisms and host responses of different cell types found in the lung and are relevant to an understanding of the pathogenesis of severe human influenza disease.
Collapse
|
39
|
Fitch PM, Henderson P, Schwarze J. Respiratory and gastrointestinal epithelial modulation of the immune response during viral infection. Innate Immun 2011; 18:179-89. [PMID: 21239454 DOI: 10.1177/1753425910391826] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Respiratory and enteric viral infections cause significant morbidity and mortality world-wide and represent a major socio-economic burden. Many of these viruses have received unprecedented public and media interest in recent years. A popular public misconception is that viruses are a threat to which the human body has only limited defences. However, the majority of primary and secondary exposures to virus are asymptomatic or induce only minor symptoms. The mucosal epithelial surfaces are the main portal of entry for viral pathogens and are centrally involved in the initiation, maintenance and polarisation of the innate and adaptive immune response to infection. This review describes the defences employed by the epithelium of the respiratory and gastrointestinal tracts during viral infections with focus on epithelial modulation of the immune response at the innate/adaptive interface.
Collapse
Affiliation(s)
- Paul M Fitch
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, UK
| | - Paul Henderson
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, UK
- Department of Child Life and Health, University of Edinburgh, UK
| | - Jürgen Schwarze
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, UK
- Department of Child Life and Health, University of Edinburgh, UK
| |
Collapse
|