1
|
Farazuddin M, Acker G, Zourob J, O’Konek JJ, Wong PT, Morris S, Rasky AJ, Kim CH, Lukacs NW, Baker JR. Inhibiting retinoic acid signaling in dendritic cells suppresses respiratory syncytial virus infection through enhanced antiviral immunity. iScience 2024; 27:110103. [PMID: 39045100 PMCID: PMC11263793 DOI: 10.1016/j.isci.2024.110103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/21/2023] [Accepted: 05/21/2024] [Indexed: 07/25/2024] Open
Abstract
Retinoic acid (RA), controls the immunoregulatory functions of many immune cells, including dendritic cells (DCs), and is important for mucosal immunity. In DCs, RA regulates the expression of pattern recognition receptors and stimulates interferon production. Here, we investigated the role of RA in DCs in mounting immunity to respiratory syncytial virus (RSV). To abolish RA signaling in DCs, we used mice expressing a dominant negative form of retinoic acid receptor-α (RARα) under the CD11c promoter (CD11c-dnRARα). Paradoxically, upon RSV challenge, these animals had lower viral burden, reduced pathology, and greater Th1 polarized immunity than wild-type (WT) mice. Moreover, CD11c-dnRARα DCs infected with RSV showed enhancement in innate and adaptive immunity genes, while genes associated with viral replication were downregulated. These findings suggest that the absence of RA signaling in DCs enhances innate immunity against RSV infection leading to decreased viral load and reduced pathogenicity.
Collapse
Affiliation(s)
- Mohammad Farazuddin
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Grant Acker
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Joseph Zourob
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jessica J. O’Konek
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Pamela T. Wong
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Susan Morris
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Andrew J. Rasky
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Chang H. Kim
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Nicholas W. Lukacs
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - James R. Baker
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
2
|
Harnessing Cellular Immunity for Vaccination against Respiratory Viruses. Vaccines (Basel) 2020. [DOI: 10.3390/vaccines8040783
expr 839529059 + 832255227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Severe respiratory viral infections, such as influenza, metapneumovirus (HMPV), respiratory syncytial virus (RSV), rhinovirus (RV), and coronaviruses, including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), cause significant mortality and morbidity worldwide. These viruses have been identified as important causative agents of acute respiratory disease in infants, the elderly, and immunocompromised individuals. Clinical signs of infection range from mild upper respiratory illness to more serious lower respiratory illness, including bronchiolitis and pneumonia. Additionally, these illnesses can have long-lasting impact on patient health well beyond resolution of the viral infection. Aside from influenza, there are currently no licensed vaccines against these viruses. However, several research groups have tested various vaccine candidates, including those that utilize attenuated virus, virus-like particles (VLPs), protein subunits, and nanoparticles, as well as recent RNA vaccines, with several of these approaches showing promise. Historically, vaccine candidates have advanced, dependent upon the ability to activate the humoral immune response, specifically leading to strong B cell responses and neutralizing antibody production. More recently, it has been recognized that the cellular immune response is also critical in proper resolution of viral infection and protection against detrimental immunopathology associated with severe disease and therefore, must also be considered when analyzing the efficacy and safety of vaccine candidates. These candidates would ideally result in robust CD4+ and CD8+ T cell responses as well as high-affinity neutralizing antibody. This review will aim to summarize established and new approaches that are being examined to harness the cellular immune response during respiratory viral vaccination.
Collapse
|
3
|
Lukacs NW, Malinczak CA. Harnessing Cellular Immunity for Vaccination against Respiratory Viruses. Vaccines (Basel) 2020; 8:783. [PMID: 33371275 PMCID: PMC7766447 DOI: 10.3390/vaccines8040783] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Severe respiratory viral infections, such as influenza, metapneumovirus (HMPV), respiratory syncytial virus (RSV), rhinovirus (RV), and coronaviruses, including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), cause significant mortality and morbidity worldwide. These viruses have been identified as important causative agents of acute respiratory disease in infants, the elderly, and immunocompromised individuals. Clinical signs of infection range from mild upper respiratory illness to more serious lower respiratory illness, including bronchiolitis and pneumonia. Additionally, these illnesses can have long-lasting impact on patient health well beyond resolution of the viral infection. Aside from influenza, there are currently no licensed vaccines against these viruses. However, several research groups have tested various vaccine candidates, including those that utilize attenuated virus, virus-like particles (VLPs), protein subunits, and nanoparticles, as well as recent RNA vaccines, with several of these approaches showing promise. Historically, vaccine candidates have advanced, dependent upon the ability to activate the humoral immune response, specifically leading to strong B cell responses and neutralizing antibody production. More recently, it has been recognized that the cellular immune response is also critical in proper resolution of viral infection and protection against detrimental immunopathology associated with severe disease and therefore, must also be considered when analyzing the efficacy and safety of vaccine candidates. These candidates would ideally result in robust CD4+ and CD8+ T cell responses as well as high-affinity neutralizing antibody. This review will aim to summarize established and new approaches that are being examined to harness the cellular immune response during respiratory viral vaccination.
Collapse
Affiliation(s)
- Nicholas W. Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA;
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
4
|
Choi EJ, Wu W, Chen Y, Yan W, Li L, Choudhury A, Bao X. The role of M2-2 PDZ-binding motifs in pulmonary innate immune responses to human metapneumovirus. J Med Virol 2020; 92:2946-2954. [PMID: 32073159 PMCID: PMC8357536 DOI: 10.1002/jmv.25713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/11/2020] [Indexed: 12/14/2022]
Abstract
Human metapneumovirus (HMPV) is a leading cause of lower respiratory tract infection (LRTI) in pediatric and geriatric populations. We recently found that two PDZ-binding motifs of the M2-2 protein, 29-DEMI-32 and 39-KEALSDGI-46, play a significant role in mediating HMPV immune evasion in airway epithelial cells (AECs). However, their role in the overall pulmonary responses to HMPV infection has not been investigated. In this study, we found that two recombinant HMPVs (rHMPV) lacking the individual M2-2 PDZ-binding motif are attenuated in mouse lungs. Mice infected with mutants produce more cytokines/chemokines in bronchoalveolar lavage (BAL) fluid compared to mice infected with wild-type rHMPV. In addition, both mutants are able to enhance the pulmonary recruitment of dendritic cells (DCs) and T cells and induce effective protections against the HMPV challenge. The DC maturation is also significantly improved by the motif mutation. Taken together, our data provide proof-of-principle for two live-attenuated M2-2 mutants to be promising HMPV vaccine candidates that are effective in inducing higher pulmonary innate immunity and generating protection against HMPV infection.
Collapse
Affiliation(s)
- Eun-Jin Choi
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
| | - Wenzhe Wu
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
| | - Yu Chen
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
| | - Weiyu Yan
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
- Honeybee Research Institute, Jiangxi Agriculture University, Nanchang, Jiangxi, China
| | - Liqing Li
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
- Department of Microbiology, The University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Atanu Choudhury
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
- The University of Texas at Austin, Austin, TX78712, USA
| | - Xiaoyong Bao
- Department of Pediatrics, The University of Texas Medical Branch at Galveston (UTMB), Galveston
- Sealy Center for Molecular Medicine, UTMB, Galveston, TX 77555, USA
- The Institute of Translational Sciences, UTMB, Galveston, TX 77555, USA
- The Institute for Human Infections and Immunity, UTMB, Galveston, TX 77555, USA
| |
Collapse
|
5
|
Einwächter H, Heiseke A, Schlitzer A, Gasteiger G, Adler H, Voehringer D, Manz MG, Ruzsics Z, Dölken L, Koszinowski UH, Sparwasser T, Reindl W, Jordan S. The Innate Immune Response to Infection Induces Erythropoietin-Dependent Replenishment of the Dendritic Cell Compartment. Front Immunol 2020; 11:1627. [PMID: 32849551 PMCID: PMC7411349 DOI: 10.3389/fimmu.2020.01627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/17/2020] [Indexed: 12/29/2022] Open
Abstract
Dendritic cells (DC) play a key role in the adaptive immune response due to their ability to present antigens and stimulate naïve T cells. Many bacteria and viruses can efficiently target DC, resulting in impairment of their immunostimulatory function or elimination. Hence, the DC compartment requires replenishment following infection to ensure continued operational readiness of the adaptive immune system. Here, we investigated the molecular and cellular mechanisms of inflammation-induced DC generation. We found that infection with viral and bacterial pathogens as well as Toll-like receptor 9 (TLR9) ligation with CpG-oligodeoxynucleotide (CpG-ODN) expanded an erythropoietin (EPO)-dependent TER119+CD11a+ cell population in the spleen that had the capacity to differentiate into TER119+CD11chigh and TER119-CD11chigh cells both in vitro and in vivo. TER119+CD11chigh cells contributed to the conventional DC pool in the spleen and specifically increased in lymph nodes draining the site of local inflammation. Our results reveal a so far undescribed inflammatory EPO-dependent pathway of DC differentiation and establish a mechanistic link between innate immune recognition of potential immunosuppressive pathogens and the maintenance of the DC pool during and after infection.
Collapse
Affiliation(s)
- Henrik Einwächter
- II. Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Alexander Heiseke
- II. Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | | | - Georg Gasteiger
- Institute of Systems Immunology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Heiko Adler
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Munich, Germany.,German Center of Lung Research (DZL), Giessen, Germany
| | - David Voehringer
- Department of Infection Biology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Nürnberg, Erlangen, Germany
| | - Markus G Manz
- Division of Hematology, Department of Internal Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Zsolt Ruzsics
- Institute of Virology, University Medical Center, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Lars Dölken
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Ulrich H Koszinowski
- Max von Pettenkofer-Institute, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medicine Mainz, Johannes-Gutenberg-University Mainz, Mainz, Germany
| | - Wolfgang Reindl
- II. Medizinische Klinik, Medizinische Fakultät Mannheim, Universität Heidelberg, Mannheim, Germany
| | - Stefan Jordan
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
| |
Collapse
|
6
|
Diab M, Schmiedel D, Seidel E, Bacharach E, Mandelboim O. Human Metapneumovirus Escapes NK Cell Recognition through the Downregulation of Stress-Induced Ligands for NKG2D. Viruses 2020; 12:v12070781. [PMID: 32698530 PMCID: PMC7412239 DOI: 10.3390/v12070781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 11/28/2022] Open
Abstract
The Pneumoviridae family includes human metapneumovirus (HMPV) and human orthopneumovirus, which is also known as a respiratory syncytial virus (HRSV). These are large enveloped, negative single-strand RNA viruses. HMPV and HRSV are the human members, which commonly infect children. HMPV, which was discovered in 2001, infects most children until the age of five, which causes an influenza-like illness. The interaction of this virus with immune cells is poorly understood. In this study, we show that HMPV evades natural killer (NK) cell attack by downregulating stress-induced ligands for the activating receptor NKG2D including: Major histocompatibility complex (MHC) class I polypeptide-related sequences A and B (MICA, MICB), UL16 binding proteins ULBP2, and ULBP3, but not ULBP1. Mechanistically, we show that the viral protein G is involved in the downregulation of ULBP2 and that the viral protein M2.2 is required for MICA and MICB downregulation. These findings emphasize the importance of NK cells, in general, and NKG2D, in particular, in controlling HMPV infection, which opens new avenues for treating HMPV.
Collapse
Affiliation(s)
- Mohammad Diab
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University Hadassah Medical School, Jerusalem 91120, Israel; (M.D.); (D.S.); (E.S.)
| | - Dominik Schmiedel
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University Hadassah Medical School, Jerusalem 91120, Israel; (M.D.); (D.S.); (E.S.)
| | - Einat Seidel
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University Hadassah Medical School, Jerusalem 91120, Israel; (M.D.); (D.S.); (E.S.)
| | - Eran Bacharach
- Department of Cell Research and Immunology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel;
| | - Ofer Mandelboim
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University Hadassah Medical School, Jerusalem 91120, Israel; (M.D.); (D.S.); (E.S.)
- Correspondence: ; Tel.: +972-2675-7515
| |
Collapse
|
7
|
Prospects of and Barriers to the Development of Epitope-Based Vaccines against Human Metapneumovirus. Pathogens 2020; 9:pathogens9060481. [PMID: 32570728 PMCID: PMC7350342 DOI: 10.3390/pathogens9060481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/10/2020] [Accepted: 06/16/2020] [Indexed: 12/21/2022] Open
Abstract
Human metapneumovirus (HMPV) is a major cause of respiratory illnesses in children, the elderly and immunocompromised patients. Although this pathogen was only discovered in 2001, an enormous amount of research has been conducted in order to develop safe and effective vaccines to prevent people from contracting the disease. In this review, we summarize current knowledge about the most promising experimental B- and T-cell epitopes of human metapneumovirus for the rational design of HMPV vaccines using vector delivery systems, paying special attention to the conservation of these epitopes among different lineages/genotypes of HMPV. The prospects of the successful development of an epitope-based HMPV vaccine are discussed in the context of recent findings regarding HMPV’s ability to modulate host immunity. In particular, we discuss the lack of data on experimental human CD4 T-cell epitopes for HMPV despite the role of CD4 lymphocytes in both the induction of higher neutralizing antibody titers and the establishment of CD8 memory T-cell responses. We conclude that current research should be focused on searching for human CD4 T-cell epitopes of HMPV that can help us to design a safe and cross-protective epitope-based HMPV vaccine.
Collapse
|
8
|
Andrade CA, Pacheco GA, Gálvez NMS, Soto JA, Bueno SM, Kalergis AM. Innate Immune Components that Regulate the Pathogenesis and Resolution of hRSV and hMPV Infections. Viruses 2020; 12:E637. [PMID: 32545470 PMCID: PMC7354512 DOI: 10.3390/v12060637] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023] Open
Abstract
The human respiratory syncytial virus (hRSV) and human Metapneumovirus (hMPV) are two of the leading etiological agents of acute lower respiratory tract infections, which constitute the main cause of mortality in infants. However, there are currently approved vaccines for neither hRSV nor hMPV. Moreover, despite the similarity between the pathology caused by both viruses, the immune response elicited by the host is different in each case. In this review, we discuss how dendritic cells, alveolar macrophages, neutrophils, eosinophils, natural killer cells, innate lymphoid cells, and the complement system regulate both pathogenesis and the resolution of hRSV and hMPV infections. The roles that these cells play during infections by either of these viruses will help us to better understand the illnesses they cause. We also discuss several controversial findings, relative to some of these innate immune components. To better understand the inflammation in the lungs, the role of the respiratory epithelium in the recruitment of innate immune cells is briefly discussed. Finally, we review the main prophylactic strategies and current vaccine candidates against both hRSV and hMPV.
Collapse
Affiliation(s)
- Catalina A. Andrade
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Gaspar A. Pacheco
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Nicolas M. S. Gálvez
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Jorge A. Soto
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Susan M. Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Alexis M. Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| |
Collapse
|
9
|
Ballegeer M, Saelens X. Cell-Mediated Responses to Human Metapneumovirus Infection. Viruses 2020; 12:v12050542. [PMID: 32423043 PMCID: PMC7290942 DOI: 10.3390/v12050542] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 12/29/2022] Open
Abstract
Viruses are the most common cause of acute respiratory tract infections (ARTI). Human metapneumovirus (hMPV) frequently causes viral pneumonia which can become life-threatening if the virus spreads to the lungs. Even though hMPV was only isolated in 2001, this negative-stranded RNA virus has probably been circulating in the human population for many decades. Interestingly, almost all adults have serologic evidence of hMPV infection. A well-established host immune response is evoked when hMPV infection occurs. However, the virus has evolved to circumvent and even exploit the host immune response. Further, infection with hMPV induces a weak memory response, and re-infections during life are common. In this review, we provide a comprehensive overview of the different cell types involved in the immune response in order to better understand the immunopathology induced by hMPV. Such knowledge may contribute to the development of vaccines and therapeutics directed against hMPV.
Collapse
Affiliation(s)
- Marlies Ballegeer
- VIB-UGent Center for Medical Biotechnology, VIB, B-9052 Ghent, Belgium;
- Department of Biochemistry and Microbiology, Ghent University, B-9000 Ghent, Belgium
| | - Xavier Saelens
- VIB-UGent Center for Medical Biotechnology, VIB, B-9052 Ghent, Belgium;
- Department of Biochemistry and Microbiology, Ghent University, B-9000 Ghent, Belgium
- Correspondence:
| |
Collapse
|
10
|
Cell-Mediated Responses to Human Metapneumovirus Infection. Viruses 2020; 12:542. [PMID: 32423043 PMCID: PMC7290942 DOI: 10.3390/v12050542&set/a 882111696+808152660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Viruses are the most common cause of acute respiratory tract infections (ARTI). Human metapneumovirus (hMPV) frequently causes viral pneumonia which can become life-threatening if the virus spreads to the lungs. Even though hMPV was only isolated in 2001, this negative-stranded RNA virus has probably been circulating in the human population for many decades. Interestingly, almost all adults have serologic evidence of hMPV infection. A well-established host immune response is evoked when hMPV infection occurs. However, the virus has evolved to circumvent and even exploit the host immune response. Further, infection with hMPV induces a weak memory response, and re-infections during life are common. In this review, we provide a comprehensive overview of the different cell types involved in the immune response in order to better understand the immunopathology induced by hMPV. Such knowledge may contribute to the development of vaccines and therapeutics directed against hMPV.
Collapse
|
11
|
Cell-Mediated Responses to Human Metapneumovirus Infection. Viruses 2020. [DOI: 10.3390/v12050542
expr 836379838 + 819716165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Viruses are the most common cause of acute respiratory tract infections (ARTI). Human metapneumovirus (hMPV) frequently causes viral pneumonia which can become life-threatening if the virus spreads to the lungs. Even though hMPV was only isolated in 2001, this negative-stranded RNA virus has probably been circulating in the human population for many decades. Interestingly, almost all adults have serologic evidence of hMPV infection. A well-established host immune response is evoked when hMPV infection occurs. However, the virus has evolved to circumvent and even exploit the host immune response. Further, infection with hMPV induces a weak memory response, and re-infections during life are common. In this review, we provide a comprehensive overview of the different cell types involved in the immune response in order to better understand the immunopathology induced by hMPV. Such knowledge may contribute to the development of vaccines and therapeutics directed against hMPV.
Collapse
|
12
|
González-Parra G, Dobrovolny HM. The rate of viral transfer between upper and lower respiratory tracts determines RSV illness duration. J Math Biol 2019; 79:467-483. [PMID: 31011792 DOI: 10.1007/s00285-019-01364-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/11/2019] [Indexed: 12/26/2022]
Abstract
Respiratory syncytial virus can lead to serious lower respiratory infection (LRI), particularly in children and the elderly. LRI can cause longer infections, lingering respiratory problems, and higher incidence of hospitalization. In this paper, we use a simplified ordinary differential equation model of viral dynamics to study the role of transport mechanisms in the occurrence of LRI. Our model uses two compartments to simulate the upper respiratory tract and the lower respiratory tract (LRT) and assumes two distinct types of viral transfer between the two compartments: diffusion and advection. We find that a range of diffusion and advection values lead to long-lasting infections in the LRT, elucidating a possible mechanism for the severe LRI infections observed in humans.
Collapse
|
13
|
Tognarelli EI, Bueno SM, González PA. Immune-Modulation by the Human Respiratory Syncytial Virus: Focus on Dendritic Cells. Front Immunol 2019; 10:810. [PMID: 31057543 PMCID: PMC6478035 DOI: 10.3389/fimmu.2019.00810] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/26/2019] [Indexed: 12/23/2022] Open
Abstract
The human respiratory syncytial virus (hRSV) is the leading cause of pneumonia in infants and produces a significant burden in the elderly. It can also infect and produce disease in otherwise healthy adults and recurrently infect those previously exposed to the virus. Importantly, recurrent infections are not necessarily a consequence of antigenic variability, as described for other respiratory viruses, but most likely due to the capacity of this virus to interfere with the host's immune response and the establishment of a protective and long-lasting immunity. Although some genes encoded by hRSV are known to have a direct participation in immune evasion, it seems that repeated infection is mainly given by its capacity to modulate immune components in such a way to promote non-optimal antiviral responses in the host. Importantly, hRSV is known to interfere with dendritic cell (DC) function, which are key cells involved in establishing and regulating protective virus-specific immunity. Notably, hRSV infects DCs, alters their maturation, migration to lymph nodes and their capacity to activate virus-specific T cells, which likely impacts the host antiviral response against this virus. Here, we review and discuss the most important and recent findings related to DC modulation by hRSV, which might be at the basis of recurrent infections in previously infected individuals and hRSV-induced disease. A focus on the interaction between DCs and hRSV will likely contribute to the development of effective prophylactic and antiviral strategies against this virus.
Collapse
Affiliation(s)
- Eduardo I Tognarelli
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
14
|
Soto JA, Gálvez NMS, Benavente FM, Pizarro-Ortega MS, Lay MK, Riedel C, Bueno SM, Gonzalez PA, Kalergis AM. Human Metapneumovirus: Mechanisms and Molecular Targets Used by the Virus to Avoid the Immune System. Front Immunol 2018; 9:2466. [PMID: 30405642 PMCID: PMC6207598 DOI: 10.3389/fimmu.2018.02466] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 10/05/2018] [Indexed: 12/27/2022] Open
Abstract
Human metapneumovirus (hMPV) is a respiratory virus, first reported the year 2001. Since then, it has been described as one of the main etiological agents that causes acute lower respiratory tract infections (ALRTIs), which is characterized by symptoms such as bronchiolitis, wheezing and coughing. Susceptible population to hMPV-infection includes newborn, children, elderly and immunocompromised individuals. This viral agent is a negative-sense, single-stranded RNA enveloped virus, that belongs to the Pneumoviridae family and Metapneumovirus genus. Early reports—previous to 2001—state several cases of respiratory illness without clear identification of the responsible pathogen, which could be related to hMPV. Despite the similarities of hMPV with several other viruses, such as the human respiratory syncytial virus or influenza virus, mechanisms used by hMPV to avoid the host immune system are still unclear. In fact, evidence indicates that hMPV induces a poor innate immune response, thereby affecting the adaptive immunity. Among these mechanisms, is the promotion of an anergic state in T cells, instead of an effective polarization or activation, which could be induced by low levels of cytokine secretion. Further, the evidences support the notion that hMPV interferes with several pattern recognition receptors (PRRs) and cell signaling pathways triggered by interferon-associated genes. However, these mechanisms reported in hMPV are not like the ones reported for hRSV, as the latter has two non-structural proteins that are able to inhibit these pathways. Several reports suggest that viral glycoproteins, such as G and SH, could play immune-modulator roles during infection. In this work, we discuss the state of the art regarding the mechanisms that underlie the poor immunity elicited by hMPV. Importantly, these mechanisms will be compared with those elicited by other common respiratory viruses.
Collapse
Affiliation(s)
- Jorge A Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás M S Gálvez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe M Benavente
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Magdalena S Pizarro-Ortega
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K Lay
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Claudia Riedel
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A Gonzalez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
15
|
Uche IK, Guerrero-Plata A. Interferon-Mediated Response to Human Metapneumovirus Infection. Viruses 2018; 10:v10090505. [PMID: 30231515 PMCID: PMC6163993 DOI: 10.3390/v10090505] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/12/2018] [Accepted: 09/15/2018] [Indexed: 12/13/2022] Open
Abstract
Human metapneumovirus (HMPV) is one of the leading causes of respiratory diseases in infants and children worldwide. Although this pathogen infects mainly young children, elderly and immunocompromised people can be also seriously affected. To date, there is no commercial vaccine available against it. Upon HMPV infection, the host innate arm of defense produces interferons (IFNs), which are critical for limiting HMPV replication. In this review, we offer an updated landscape of the HMPV mediated-IFN response in different models as well as some of the defense tactics employed by the virus to circumvent IFN response.
Collapse
Affiliation(s)
- Ifeanyi K Uche
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Antonieta Guerrero-Plata
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA.
- Center for Experimental Infectious Disease Research, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
16
|
Schmidt ME, Varga SM. Cytokines and CD8 T cell immunity during respiratory syncytial virus infection. Cytokine 2018; 133:154481. [PMID: 30031680 PMCID: PMC6551303 DOI: 10.1016/j.cyto.2018.07.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 07/07/2018] [Indexed: 01/10/2023]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection and hospitalization in infants. In spite of the enormous clinical burden caused by RSV infections, there remains no efficacious RSV vaccine. CD8 T cells mediate viral clearance as well as provide protection against a secondary RSV infection. However, RSV-specific CD8 T cells may also induce immunopathology leading to exacerbated morbidity and mortality. Many of the crucial functions performed by CD8 T cells are mediated by the cytokines they produce. IFN-γ and TNF are produced by CD8 T cells following RSV infection and contribute to both the acceleration of viral clearance and the induction of immunopathology. To prevent immunopathology, regulatory mechanisms are in place within the immune system to inhibit CD8 T cell effector functions after the infection has been cleared. The actions of a variety of cytokines, including IL-10 and IL-4, play a critical role in the regulation of CD8 T cell effector activity. Herein, we review the current literature on CD8 T cell responses and the functions of the cytokines they produce following RSV infection. Additionally, we discuss the regulation of CD8 T cell activation and effector functions through the actions of various cytokines.
Collapse
Affiliation(s)
- Megan E Schmidt
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA; Department of Pathology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
17
|
Baños-Lara MDR, Zabaleta J, Garai J, Baddoo M, Guerrero-Plata A. Comparative analysis of miRNA profile in human dendritic cells infected with respiratory syncytial virus and human metapneumovirus. BMC Res Notes 2018; 11:432. [PMID: 29970194 PMCID: PMC6029031 DOI: 10.1186/s13104-018-3541-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/26/2018] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Human metapneumovirus (HMPV) and respiratory syncytial virus (RSV) are responsible for respiratory diseases, mostly in children. Despite the clinical and epidemiological similarities between these two pneumoviruses, they elicit different immune responses. This work aims to further our understanding of the differential immune response induced by these respiratory viruses by determining the changes of small non-coding RNAs (miRNAs), which regulate gene expression and are involved in numerous cellular processes including the immune system. RESULTS In the present study, we analyzed the expression of miRNA transcripts of human dendritic cells infected with RSV or HMPV by high throughput sequencing using Illumina sequencing technology. Further validation of miRNA expression by quantitative polymerase chain reaction indicated that HMPV infection up-regulated the expression of 2 miRNAs (hsa-miR-182-5p and hsa-miR-4634), while RSV infection induced significant expression of 3 miRNAs (hsa-miR-4448, hsa-miR-30a-5p and hsa-miR-4634). The predominant miRNA induced by both viruses was hsa-miR-4634.
Collapse
Affiliation(s)
- Ma Del Rocio Baños-Lara
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA.,Universidad Popular Autonoma del Estado de Puebla, UPAEP, Puebla, Mexico
| | - Jovanny Zabaleta
- Department of Pediatrics, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA.,Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Jone Garai
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Melody Baddoo
- Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Antonieta Guerrero-Plata
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA. .,Center for Experimental Infectious Disease Research, Louisiana State University, Baton Rouge, LA, 70803, USA.
| |
Collapse
|
18
|
Central Role of the NF-κB Pathway in the Scgb1a1-Expressing Epithelium in Mediating Respiratory Syncytial Virus-Induced Airway Inflammation. J Virol 2018; 92:JVI.00441-18. [PMID: 29593031 DOI: 10.1128/jvi.00441-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 12/24/2022] Open
Abstract
Lower respiratory tract infection with respiratory syncytial virus (RSV) produces profound inflammation. Despite an understanding of the role of adaptive immunity in RSV infection, the identity of the major sentinel cells initially triggering inflammation is controversial. Here we evaluate the role of nonciliated secretoglobin (Scgb1a1)-expressing bronchiolar epithelial cells in RSV infection. Mice expressing a tamoxifen (TMX)-inducible Cre recombinase-estrogen receptor fusion protein (CreERTM) knocked into the Scgb1a1 locus were crossed with mice that harbor a RelA conditional allele (RelAfl ), with loxP sites flanking exons 5 to 8 of the Rel homology domain. The Scgb1a1CreERTM/+ × RelAfl/fl mouse is a RelA conditional knockout (RelACKO) of a nonciliated epithelial cell population enriched in the small bronchioles. TMX-treated RelACKO mice have reduced pulmonary neutrophilic infiltration and impaired expression and secretion of NF-κB-dependent cytokines in response to RSV. In addition, RelACKO mice had reduced expression levels of interferon (IFN) regulatory factor 1/7 (IRF1/7) and retinoic acid-inducible gene I (RIG-I), components of the mucosal IFN positive-feedback loop. We demonstrate that RSV replication induces RelA to complex with bromodomain-containing protein 4 (BRD4), a cofactor required for RNA polymerase II (Pol II) phosphorylation, activating the atypical histone acetyltransferase (HAT) activity of BRD4 required for phospho-Ser2 Pol II formation, histone H3K122 acetylation, and cytokine secretion in vitro and in vivo TMX-treated RelACKO mice have less weight loss and reduced airway obstruction/hyperreactivity yet similar levels of IFN-γ production despite higher levels of virus production. These data indicate that the nonciliated Scgb1a1-expressing epithelium is a major innate sensor for restricting RSV infection by mediating neutrophilic inflammation and chemokine and mucosal IFN production via the RelA-BRD4 pathway.IMPORTANCE RSV infection is the most common cause of infant hospitalizations in the United States, resulting in 2.1 million children annually requiring medical attention. RSV primarily infects nasal epithelial cells, spreading distally to produce severe lower respiratory tract infections. Our study examines the role of a nonciliated respiratory epithelial cell population in RSV infection. We genetically engineered a mouse that can be selectively depleted of the NF-κB/RelA transcription factor in this subset of epithelial cells. These mice show an impaired activation of the bromodomain-containing protein 4 (BRD4) coactivator, resulting in reduced cytokine expression and neutrophilic inflammation. During the course of RSV infection, epithelial RelA-depleted mice have reduced disease scores and airway hyperreactivity yet increased levels of virus replication. We conclude that RelA-BRD4 signaling in nonciliated bronchiolar epithelial cells mediates neutrophilic airway inflammation and disease severity. This complex is an attractive target to reduce the severity of infection.
Collapse
|
19
|
Ascough S, Paterson S, Chiu C. Induction and Subversion of Human Protective Immunity: Contrasting Influenza and Respiratory Syncytial Virus. Front Immunol 2018; 9:323. [PMID: 29552008 PMCID: PMC5840263 DOI: 10.3389/fimmu.2018.00323] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/06/2018] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) and influenza are among the most important causes of severe respiratory disease worldwide. Despite the clinical need, barriers to developing reliably effective vaccines against these viruses have remained firmly in place for decades. Overcoming these hurdles requires better understanding of human immunity and the strategies by which these pathogens evade it. Although superficially similar, the virology and host response to RSV and influenza are strikingly distinct. Influenza induces robust strain-specific immunity following natural infection, although protection by current vaccines is short-lived. In contrast, even strain-specific protection is incomplete after RSV and there are currently no licensed RSV vaccines. Although animal models have been critical for developing a fundamental understanding of antiviral immunity, extrapolating to human disease has been problematic. It is only with recent translational advances (such as controlled human infection models and high-dimensional technologies) that the mechanisms responsible for differences in protection against RSV compared to influenza have begun to be elucidated in the human context. Influenza infection elicits high-affinity IgA in the respiratory tract and virus-specific IgG, which correlates with protection. Long-lived influenza-specific T cells have also been shown to ameliorate disease. This robust immunity promotes rapid emergence of antigenic variants leading to immune escape. RSV differs markedly, as reinfection with similar strains occurs despite natural infection inducing high levels of antibody against conserved antigens. The immunomodulatory mechanisms of RSV are thus highly effective in inhibiting long-term protection, with disturbance of type I interferon signaling, antigen presentation and chemokine-induced inflammation possibly all contributing. These lead to widespread effects on adaptive immunity with impaired B cell memory and reduced T cell generation and functionality. Here, we discuss the differences in clinical outcome and immune response following influenza and RSV. Specifically, we focus on differences in their recognition by innate immunity; the strategies used by each virus to evade these early immune responses; and effects across the innate-adaptive interface that may prevent long-lived memory generation. Thus, by comparing these globally important pathogens, we highlight mechanisms by which optimal antiviral immunity may be better induced and discuss the potential for these insights to inform novel vaccines.
Collapse
Affiliation(s)
- Stephanie Ascough
- Section of Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| | - Suzanna Paterson
- Section of Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| | - Christopher Chiu
- Section of Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| |
Collapse
|
20
|
Abstract
Human metapneumovirus (HMPV) is a leading cause of acute respiratory infection, particularly in children, immunocompromised patients, and the elderly. HMPV, which is closely related to avian metapneumovirus subtype C, has circulated for at least 65 years, and nearly every child will be infected with HMPV by the age of 5. However, immunity is incomplete, and re-infections occur throughout adult life. Symptoms are similar to those of other respiratory viral infections, ranging from mild (cough, rhinorrhea, and fever) to more severe (bronchiolitis and pneumonia). The preferred method for diagnosis is reverse transcription-polymerase chain reaction as HMPV is difficult to culture. Although there have been many advances made in the past 16 years since its discovery, there are still no US Food and Drug Administration-approved antivirals or vaccines available to treat HMPV. Both small animal and non-human primate models have been established for the study of HMPV. This review will focus on the epidemiology, transmission, and clinical manifestations in humans as well as the animal models of HMPV pathogenesis and host immune response.
Collapse
Affiliation(s)
- Nazly Shafagati
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John Williams
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
21
|
Diaz-Dinamarca DA, Ibañez FJ, Soto DA, Soto JA, Cespedes PF, Muena NA, Garate DS, Kalergis AM, Vasquez AE. Immunization with a Mixture of Nucleoprotein from Human Metapneumovirus and AbISCO-100 Adjuvant Reduces Viral Infection in Mice Model. Viral Immunol 2018; 31:306-314. [PMID: 29373084 DOI: 10.1089/vim.2017.0159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The human metapneumovirus (hMPV) is the second leading cause globally of acute infection of the respiratory tract in children, infecting the upper and lower airways. The hMPV may induce an inappropriate Th2-type immune response, which causes severe pulmonary inflammation, leading to the obstruction of airways. Despite its severe epidemiological relevance, no vaccines are currently available for the prevention of hMPV-induced illness. In this investigation, we demonstrated that immunization of mice with the recombinant hMPV nucleoprotein (hMPV-N) mixed with the AbISCO-100 adjuvant reduced viral replication in lungs following challenge with the virus. We found that immunized mice had reduced weight loss, decreased granulocytes in the lung, an increased level of specific nucleoprotein antibodies of IgG1 and IgG2a-isotypes, and a local profile of Th1/Th17-type cytokines. Our results suggest that immunization with the hMPV-N and the AbISCO-100 adjuvant induces a reduction of viral infection and could be considered for the development of an hMPV vaccine.
Collapse
Affiliation(s)
- Diego A Diaz-Dinamarca
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile .,2 Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy , Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco J Ibañez
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile .,2 Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy , Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel A Soto
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile
| | - Jorge A Soto
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile .,2 Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy , Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo F Cespedes
- 2 Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy , Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás A Muena
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile
| | - Diego S Garate
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile
| | - Alexis M Kalergis
- 2 Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy , Pontificia Universidad Católica de Chile, Santiago, Chile .,3 Facultad de Medicina, Departamento de Endocrinología, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Abel E Vasquez
- 1 Sección de Biotecnología, Departamento de Salud Ambiental, Instituto de Salud Pública de Chile , Santiago, Chile .,4 Universidad San Sebastián , Facultad de Ciencia, Escuela de Bioquímica, Providencia, Santiago, Chile
| |
Collapse
|
22
|
Human Metapneumovirus M2-2 Protein Acts as a Negative Regulator of Alpha Interferon Production by Plasmacytoid Dendritic Cells. J Virol 2017; 91:JVI.00579-17. [PMID: 28768858 DOI: 10.1128/jvi.00579-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/24/2017] [Indexed: 12/20/2022] Open
Abstract
Human metapneumovirus (HMPV) has the ability to inhibit Toll-like receptor 7 (TLR7)- and TLR9-dependent alpha interferon (IFN-α) production by plasmacytoid dendritic cells (pDCs). However, the inhibition mechanism remains largely unknown. To identify viral proteins responsible for this inhibition, we performed a screening of HMPV open reading frames (ORFs) for the ability to block TLR7/9-dependent signaling reconstituted in HEK293T cells by transfection with myeloid differentiation factor 88 (MyD88), tumor necrosis factor receptor-associated factor 6 (TRAF6), IKKα, and IFN regulatory factor 7 (IRF7). This screening demonstrated that the M2-2 protein was the most potent inhibitor of TLR7/9-dependent IFN-α induction. A recombinant HMPV in which the M2-2 ORF was silenced indeed induced greater IFN-α production by human pDCs than wild-type HMPV did. Immunoprecipitation experiments showed direct physical association of the M2-2 protein with the inhibitory domain (ID) of IRF7. As a natural consequence of this, transfection of IRF7 lacking the ID, a constitutively active mutant, resulted in activation of the IFN-α promoter even in the presence of M2-2. Bioluminescence resonance energy transfer assays and split Renilla luciferase complementation assays revealed that M2-2 inhibited MyD88/TRAF6/IKKα-induced homodimerization of IRF7. In contrast, expression of the M2-2 protein did not result in inhibition of IPS-1-induced homodimerization and resultant activation of IRF7. This indicates that inhibition of MyD88/TRAF6/IKKα-induced IRF7 homodimerization does not result from a steric effect of M2-2 binding. Instead, it was found that M2-2 inhibited MyD88/TRAF6/IKKα-induced phosphorylation of IRF7 on Ser477. These results suggest that M2-2 blocks TLR7/9-dependent IFN-α induction by preventing IRF7 homodimerization, possibly through its effects on the phosphorylation status of IRF7.IMPORTANCE The family Paramyxoviridae is divided into two subfamilies, the Paramyxovirinae and the Pneumovirinae Members of the subfamily Paramyxovirinae have the ability to inhibit TLR7/9-dependent IFN-α production, and the underlying inhibition mechanism has been intensively studied. In contrast, little is known about how members of the subfamily Pneumovirinae regulate IFN-α production by pDCs. We identified the M2-2 protein of HMPV, a member of the subfamily Pneumovirinae, as a negative regulator of IFN-α production by pDCs and uncovered the underlying mechanism. This study explains in part why the M2-2 knockout recombinant HMPV is attenuated and further suggests that M2-2 is a potential target for HMPV therapy.
Collapse
|
23
|
Abstract
Globally, as a leading agent of acute respiratory tract infections in children <5 years of age and the elderly, the human metapneumovirus (HMPV) has gained considerable attention. As inferred from studies comparing vaccinated and experimentally infected mice, the acquired immune response elicited by this pathogen fails to efficiently clear the virus from the airways, which leads to an exaggerated inflammatory response and lung damage. Furthermore, after disease resolution, there is a poor development of T and B cell immunological memory, which is believed to promote reinfections and viral spread in the community. In this article, we discuss the molecular mechanisms that shape the interactions of HMPV with host tissues that lead to pulmonary pathology and to the development of adaptive immunity that fails to protect against natural infections by this virus.
Collapse
|
24
|
Cheemarla NR, Baños-Lara MDR, Naidu S, Guerrero-Plata A. Neutrophils regulate the lung inflammatory response via γδ T cell infiltration in an experimental mouse model of human metapneumovirus infection. J Leukoc Biol 2017; 101:1383-1392. [PMID: 28336678 DOI: 10.1189/jlb.4a1216-519rr] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/01/2017] [Accepted: 03/03/2017] [Indexed: 12/28/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human circulation. They are the first immune cell population recruited to the sites of infection. However, the role of neutrophils to regulate host immune responses during respiratory viral infections is largely unknown. To elucidate the role of neutrophils in respiratory antiviral defense, we used an experimental mouse model of human metapneumovirus (HMPV) infection. HMPV, a member of the Paramyxoviridae family, is a leading respiratory pathogen causing severe symptoms, such as bronchiolitis and pneumonia, in young, elderly, and immunocompromised patients. We demonstrate that neutrophils are the predominant population of immune cells recruited into the lungs after HMPV infection. This led us to hypothesize that neutrophils represent a key player of the immune response during HMPV infection, thereby regulating HMPV-induced lung pathogenesis. Specific depletion of neutrophils in vivo using a mAb and simultaneous infection with HMPV exhibited higher levels of inflammatory cytokines, pulmonary inflammation, and severe clinical disease compared with HMPV-infected, competent mice. Interestingly, the lack of neutrophils altered γδ T cell accumulation in the lung. The absence of γδ T cells during HMPV infection led to reduced pulmonary inflammation. These novel findings demonstrate that neutrophils play a critical role in controlling HMPV-induced inflammatory responses by regulating γδ T cell infiltration to the site of infection.
Collapse
Affiliation(s)
- Nagarjuna R Cheemarla
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA; and
| | - Ma Del Rocío Baños-Lara
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA; and
| | - Shan Naidu
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA; and
| | - Antonieta Guerrero-Plata
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA; and .,Center for Experimental Infectious Disease Research, Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
25
|
Diab M, Glasner A, Isaacson B, Bar-On Y, Drori Y, Yamin R, Duev-Cohen A, Danziger O, Zamostiano R, Mandelboim M, Jonjic S, Bacharach E, Mandelboim O. NK-cell receptors NKp46 and NCR1 control human metapneumovirus infection. Eur J Immunol 2017; 47:692-703. [DOI: 10.1002/eji.201646756] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 12/20/2016] [Accepted: 02/08/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Mohammad Diab
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Ariella Glasner
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Batya Isaacson
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Yotam Bar-On
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Yaron Drori
- Central Virology Laboratory, Ministry of Health, Public Health Services; Chaim Sheba Medical Center, Tel-Hashomer; Ramat-Gan Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine; Tel-Aviv University; Tel-Aviv Israel
| | - Rachel Yamin
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Alexandra Duev-Cohen
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| | - Oded Danziger
- Department of Cell Research and Immunology, Faculty of Life Sciences; Tel Aviv University; Tel Aviv Israel
| | - Rachel Zamostiano
- Department of Cell Research and Immunology, Faculty of Life Sciences; Tel Aviv University; Tel Aviv Israel
| | - Michal Mandelboim
- Central Virology Laboratory, Ministry of Health, Public Health Services; Chaim Sheba Medical Center, Tel-Hashomer; Ramat-Gan Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine; Tel-Aviv University; Tel-Aviv Israel
| | - Stipan Jonjic
- Department of Histology and Embryology, Faculty of Medicine; University of Rijeka; Rijeka Croatia
- Center for Proteomics, Faculty of Medicine; University of Rijeka; Rijeka Croatia
| | - Eran Bacharach
- Department of Cell Research and Immunology, Faculty of Life Sciences; Tel Aviv University; Tel Aviv Israel
| | - Ofer Mandelboim
- The Lautenberg Center for General and Tumor Immunology, the BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC); The Hebrew University Hadassah Medical School; Jerusalem Israel
| |
Collapse
|
26
|
Céspedes PF, Rey-Jurado E, Espinoza JA, Rivera CA, Canedo-Marroquín G, Bueno SM, Kalergis AM. A single, low dose of a cGMP recombinant BCG vaccine elicits protective T cell immunity against the human respiratory syncytial virus infection and prevents lung pathology in mice. Vaccine 2017; 35:757-766. [PMID: 28065474 DOI: 10.1016/j.vaccine.2016.12.048] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 12/09/2016] [Accepted: 12/21/2016] [Indexed: 10/20/2022]
Abstract
Human respiratory syncytial virus (hRSV) is a major health burden worldwide, causing the majority of hospitalizations in children under two years old due to bronchiolitis and pneumonia. HRSV causes year-to-year outbreaks of disease, which also affects the elderly and immunocompromised adults. Furthermore, both hRSV morbidity and epidemics are explained by a consistently high rate of re-infections that take place throughout the patient life. Although significant efforts have been invested worldwide, currently there are no licensed vaccines to prevent hRSV infection. Here, we describe that a recombinant Bacillus Calmette-Guerin (BCG) vaccine expressing the nucleoprotein (N) of hRSV formulated under current good manufacture practices (cGMP rBCG-N-hRSV) confers protective immunity to the virus in mice. Our results show that a single dose of the GMP rBCG-N-hRSV vaccine retains its capacity to protect mice against a challenge with a disease-causing infection of 1×107 plaque-forming units (PFUs) of the hRSV A2 clinical strain 13018-8. Compared to unimmunized infected controls, vaccinated mice displayed reduced weight loss and less infiltration of neutrophils within the airways, as well as reduced viral loads in bronchoalveolar lavages, parameters that are characteristic of hRSV infection in mice. Also, ex vivo re-stimulation of splenic T cells at 28days post-immunization activated a repertoire of T cells secreting IFN-γ and IL-17, which further suggest that the rBCG-N-hRSV vaccine induced a mixed, CD8+ and CD4+ T cell response capable of both restraining viral spread and preventing damage of the lungs. All these features support the notion that rBCG-N-hRSV is a promising candidate vaccine to be used in humans to prevent the disease caused by hRSV in the susceptible population.
Collapse
Affiliation(s)
- Pablo F Céspedes
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Emma Rey-Jurado
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Janyra A Espinoza
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Claudia A Rivera
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Gisela Canedo-Marroquín
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile; INSERM UMR 1064, Nantes, France.
| |
Collapse
|
27
|
Bates JT, Pickens JA, Schuster JE, Johnson M, Tollefson SJ, Williams JV, Davis NL, Johnston RE, Schultz-Darken N, Slaughter JC, Smith-House F, Crowe JE. Immunogenicity and efficacy of alphavirus-derived replicon vaccines for respiratory syncytial virus and human metapneumovirus in nonhuman primates. Vaccine 2016; 34:950-6. [PMID: 26772634 DOI: 10.1016/j.vaccine.2015.12.045] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 12/15/2015] [Accepted: 12/18/2015] [Indexed: 12/14/2022]
Abstract
Human respiratory syncytial virus (hRSV) and human metapneumovirus (hMPV) are major causes of illness among children, the elderly, and the immunocompromised. No vaccine has been licensed for protection against either of these viruses. We tested the ability of two Venezuelan equine encephalitis virus-based viral replicon particle (VEE-VRP) vaccines that express the hRSV or hMPV fusion (F) protein to confer protection against hRSV or hMPV in African green monkeys. Animals immunized with VEE-VRP vaccines developed RSV or MPV F-specific antibodies and serum neutralizing activity. Compared to control animals, immunized animals were better able to control viral load in the respiratory mucosa following challenge and had lower levels of viral genome in nasopharyngeal and bronchoalveolar lavage fluids. The high level of immunogenicity and protective efficacy induced by these vaccine candidates in nonhuman primates suggest that they hold promise for further development.
Collapse
Affiliation(s)
- John T Bates
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jennifer A Pickens
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jennifer E Schuster
- The Department of Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Monika Johnson
- The Department of Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sharon J Tollefson
- The Department of Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John V Williams
- The Department of Vanderbilt University Medical Center, Nashville, TN 37232, USA; The Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nancy L Davis
- The Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Nancy Schultz-Darken
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - James C Slaughter
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; The Department of Biostatistics of Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Frances Smith-House
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James E Crowe
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; The Department of Vanderbilt University Medical Center, Nashville, TN 37232, USA; The Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
28
|
Gómez RS, Ramirez BA, Céspedes PF, Cautivo KM, Riquelme SA, Prado CE, González PA, Kalergis AM. Contribution of Fcγ receptors to human respiratory syncytial virus pathogenesis and the impairment of T-cell activation by dendritic cells. Immunology 2016; 147:55-72. [PMID: 26451966 PMCID: PMC4693880 DOI: 10.1111/imm.12541] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 09/16/2015] [Accepted: 09/29/2015] [Indexed: 12/11/2022] Open
Abstract
Human respiratory syncytial virus (hRSV) is the leading cause of infant hospitalization related to respiratory disease. Infection with hRSV produces abundant infiltration of immune cells into the airways, which combined with an exacerbated pro-inflammatory immune response can lead to significant damage to the lungs. Human RSV re-infection is extremely frequent, suggesting that this virus may have evolved molecular mechanisms that interfere with host adaptive immunity. Infection with hRSV can be reduced by administering a humanized neutralizing antibody against the virus fusion protein in high-risk infants. Although neutralizing antibodies against hRSV effectively block the infection of airway epithelial cells, here we show that both, bone marrow-derived dendritic cells (DCs) and lung DCs undergo infection with IgG-coated virus (hRSV-IC), albeit abortive. Yet, this is enough to negatively modulate DC function. We observed that such a process is mediated by Fcγ receptors (FcγRs) expressed on the surface of DCs. Remarkably, we also observed that in the absence of hRSV-specific antibodies FcγRIII knockout mice displayed significantly less cellular infiltration in the lungs after hRSV infection, compared with wild-type mice, suggesting a potentially harmful, IgG-independent role for this receptor in hRSV disease. Our findings support the notion that FcγRs can contribute significantly to the modulation of DC function by hRSV and hRSV-IC. Further, we provide evidence for an involvement of FcγRIII in the development of hRSV pathogenesis.
Collapse
MESH Headings
- Adaptive Immunity
- Animals
- Antibodies, Neutralizing/pharmacology
- Antibodies, Viral/immunology
- Antibodies, Viral/metabolism
- Antiviral Agents/pharmacology
- Cells, Cultured
- Coculture Techniques
- Cytokines/metabolism
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/virology
- Disease Models, Animal
- Immunoglobulin G/immunology
- Immunoglobulin G/metabolism
- Lung/drug effects
- Lung/immunology
- Lung/metabolism
- Lung/virology
- Lymphocyte Activation/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Palivizumab/pharmacology
- Receptors, IgG/deficiency
- Receptors, IgG/genetics
- Receptors, IgG/immunology
- Receptors, IgG/metabolism
- Respiratory Syncytial Virus Infections/drug therapy
- Respiratory Syncytial Virus Infections/genetics
- Respiratory Syncytial Virus Infections/immunology
- Respiratory Syncytial Virus Infections/metabolism
- Respiratory Syncytial Virus Infections/virology
- Respiratory Syncytial Virus, Human/drug effects
- Respiratory Syncytial Virus, Human/immunology
- Respiratory Syncytial Virus, Human/pathogenicity
- Signal Transduction
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/virology
- Viral Load
- Virus Replication
Collapse
Affiliation(s)
- Roberto S. Gómez
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Bruno A. Ramirez
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Pablo F. Céspedes
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Kelly M. Cautivo
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Sebastián A. Riquelme
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
- INSERM U1064NantesFrance
| | - Carolina E. Prado
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Pablo A. González
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
- INSERM U1064NantesFrance
- Departamento de ReumatologíaFacultad de Medicina. Pontificia Universidad Católica de ChileSantiago de ChileChile
| |
Collapse
|
29
|
Al-Afif A, Alyazidi R, Oldford SA, Huang YY, King CA, Marr N, Haidl ID, Anderson R, Marshall JS. Respiratory syncytial virus infection of primary human mast cells induces the selective production of type I interferons, CXCL10, and CCL4. J Allergy Clin Immunol 2015; 136:1346-54.e1. [DOI: 10.1016/j.jaci.2015.01.042] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 12/07/2014] [Accepted: 01/27/2015] [Indexed: 10/23/2022]
|
30
|
Rivera CA, Gómez RS, Díaz RA, Céspedes PF, Espinoza JA, González PA, Riedel CA, Bueno SM, Kalergis AM. Novel therapies and vaccines against the human respiratory syncytial virus. Expert Opin Investig Drugs 2015; 24:1613-30. [DOI: 10.1517/13543784.2015.1099626] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
31
|
Erickson JJ, Lu P, Wen S, Hastings AK, Gilchuk P, Joyce S, Shyr Y, Williams JV. Acute Viral Respiratory Infection Rapidly Induces a CD8+ T Cell Exhaustion-like Phenotype. THE JOURNAL OF IMMUNOLOGY 2015; 195:4319-30. [PMID: 26401005 DOI: 10.4049/jimmunol.1403004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 08/21/2015] [Indexed: 11/19/2022]
Abstract
Acute viral infections typically generate functional effector CD8(+) T cells (TCD8) that aid in pathogen clearance. However, during acute viral lower respiratory infection, lung TCD8 are functionally impaired and do not optimally control viral replication. T cells also become unresponsive to Ag during chronic infections and cancer via signaling by inhibitory receptors such as programmed cell death-1 (PD-1). PD-1 also contributes to TCD8 impairment during viral lower respiratory infection, but how it regulates TCD8 impairment and the connection between this state and T cell exhaustion during chronic infections are unknown. In this study, we show that PD-1 operates in a cell-intrinsic manner to impair lung TCD8. In light of this, we compared global gene expression profiles of impaired epitope-specific lung TCD8 to functional spleen TCD8 in the same human metapneumovirus-infected mice. These two populations differentially regulate hundreds of genes, including the upregulation of numerous inhibitory receptors by lung TCD8. We then compared the gene expression of TCD8 during human metapneumovirus infection to those in acute or chronic lymphocytic choriomeningitis virus infection. We find that the immunophenotype of lung TCD8 more closely resembles T cell exhaustion late into chronic infection than do functional effector T cells arising early in acute infection. Finally, we demonstrate that trafficking to the infected lung alone is insufficient for TCD8 impairment or inhibitory receptor upregulation, but that viral Ag-induced TCR signaling is also required. Our results indicate that viral Ag in infected lungs rapidly induces an exhaustion-like state in lung TCD8 characterized by progressive functional impairment and upregulation of numerous inhibitory receptors.
Collapse
Affiliation(s)
- John J Erickson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Pengcheng Lu
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Sherry Wen
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Andrew K Hastings
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Pavlo Gilchuk
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Sebastian Joyce
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232; Veterans Administration Tennessee Valley Healthcare System, Nashville, TN 37232
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - John V Williams
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232; and Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, PA 15224
| |
Collapse
|
32
|
Immune Response to Human Metapneumovirus Infection: What We Have Learned from the Mouse Model. Pathogens 2015; 4:682-96. [PMID: 26393657 PMCID: PMC4584281 DOI: 10.3390/pathogens4030682] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/09/2015] [Accepted: 09/15/2015] [Indexed: 01/17/2023] Open
Abstract
Human Metapneumovirus (hMPV) is a leading respiratory viral pathogen associated with bronchiolitis, pneumonia, and asthma exacerbation in young children, the elderly and immunocompromised individuals. The development of a potential vaccine against hMPV requires detailed understanding of the host immune system, which plays a significant role in hMPV pathogenesis, susceptibility and vaccine efficacy. As a result, animal models have been developed to better understand the mechanisms by which hMPV causes disease. Several animal models have been evaluated and established so far to study the host immune responses and pathophysiology of hMPV infection. However, inbred laboratory mouse strains have been one of the most used animal species for experimental modeling and therefore used for the studies of immunity and immunopathogenesis to hMPV. This review summarizes the contributions of the mouse model to our understanding of the immune response against hMPV infection.
Collapse
|
33
|
Ptaschinski C, Mukherjee S, Moore ML, Albert M, Helin K, Kunkel SL, Lukacs NW. RSV-Induced H3K4 Demethylase KDM5B Leads to Regulation of Dendritic Cell-Derived Innate Cytokines and Exacerbates Pathogenesis In Vivo. PLoS Pathog 2015; 11:e1004978. [PMID: 26083387 PMCID: PMC4470918 DOI: 10.1371/journal.ppat.1004978] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 05/26/2015] [Indexed: 12/22/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection can result in severe disease partially due to its ability to interfere with the initiation of Th1 responses targeting the production of type I interferons (IFN) and promoting a Th2 immune environment. Epigenetic modulation of gene transcription has been shown to be important in regulating inflammatory pathways. RSV-infected bone marrow-derived DCs (BMDCs) upregulated expression of Kdm5b/Jarid1b H3K4 demethylase. Kdm5b-specific siRNA inhibition in BMDC led to a 10-fold increase in IFN-β as well as increases in IL-6 and TNF-α compared to control-transfected cells. The generation of Kdm5bfl/fl-CD11c-Cre+ mice recapitulated the latter results during in vitro DC activation showing innate cytokine modulation. In vivo, infection of Kdm5bfl/fl-CD11c-Cre+ mice with RSV resulted in higher production of IFN-γ and reduced IL-4 and IL-5 compared to littermate controls, with significantly decreased inflammation, IL-13, and mucus production in the lungs. Sensitization with RSV-infected DCs into the airways of naïve mice led to an exacerbated response when mice were challenged with live RSV infection. When Kdm5b was blocked in DCs with siRNA or DCs from Kdm5bfl/fl-CD11c-CRE mice were used, the exacerbated response was abrogated. Importantly, human monocyte-derived DCs treated with a chemical inhibitor for KDM5B resulted in increased innate cytokine levels as well as elicited decreased Th2 cytokines when co-cultured with RSV reactivated CD4+ T cells. These results suggest that KDM5B acts to repress type I IFN and other innate cytokines to promote an altered immune response following RSV infection that contributes to development of chronic disease.
Collapse
Affiliation(s)
- Catherine Ptaschinski
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Sumanta Mukherjee
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Martin L. Moore
- Department of Pediatrics, Emory University, and Children’s Healthcare of Atlanta, Atlanta, Georgia, United States of America
| | - Mareike Albert
- Biotech Research and Innovation Centre, Centre for Epigenetics, and Danish Stem Cell Center, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Helin
- Biotech Research and Innovation Centre, Centre for Epigenetics, and Danish Stem Cell Center, University of Copenhagen, Copenhagen, Denmark
| | - Steven L. Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Nicholas W. Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
34
|
Abstract
ABSTRACT Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection and hospitalization among infants. Despite the significant healthcare burden, there is no licensed RSV vaccine currently available. This problem is further exacerbated as a natural RSV infection fails to elicit the development of long-lived immunity. It is well established that RSV-specific antibodies play a critical role in mediating protection from severe disease. The CD8 T-cell response is critical for mediating virus clearance following an acute RSV infection. However, the relative contribution of memory CD8 T cells in providing protection against secondary RSV infections remains unclear. In addition, data from animal models indicate that memory CD8 T-cell responses can be pathogenic under certain conditions. Herein, we provide an overview of the CD8 T-cell response elicited by RSV infection and how our current knowledge may impact future studies and vaccine development.
Collapse
Affiliation(s)
- Cory J Knudson
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
35
|
Deng J, Chen Y, Liu G, Ren J, Go C, Ivanciuc T, Deepthi K, Casola A, Garofalo RP, Bao X. Mitochondrial antiviral-signalling protein plays an essential role in host immunity against human metapneumovirus. J Gen Virol 2015; 96:2104-2113. [PMID: 25953917 DOI: 10.1099/vir.0.000178] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Human metapneumovirus (hMPV) is a common cause of respiratory tract infection in the paediatrics population. Recently, we and others have shown that retinoic acid-inducible gene 1 (RIG-I)-like receptors (RLRs) are essential for hMPV-induced cellular antiviral signalling. However, the contribution of those receptors to host immunity against pulmonary hMPV infection is largely unexplored. In this study, mice deficient in mitochondrial antiviral-signalling protein (MAVS), an adaptor of RLRs, were used to investigate the role(s) of these receptors in pulmonary immune responses to hMPV infection. MAVS deletion significantly impaired the induction of antiviral and pro-inflammatory cytokines and the recruitment of immune cells to the bronchoalveolar lavage fluid by hMPV. Compared with WT mice, mice lacking MAVS demonstrated decreased abilities to activate pulmonary dendritic cells (DCs) and abnormal primary T-cell responses to hMPV infection. In addition, mice deficient in MAVS had a higher peak of viral load at day 5 post-infection (p.i.) than WT mice, but were able to clear hMPV by day 7 p.i. similarly to WT mice. Taken together, our data indicate a role of MAVS-mediated pathways in the pulmonary immune responses to hMPV infection and the early control of hMPV replication.
Collapse
Affiliation(s)
- Junfang Deng
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA.,Department of Hepatobiliary Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, PR China
| | - Yu Chen
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA.,Department of Pediatrics, TongJi Hospital, Huazhong University of Science and Technology, PR China
| | - Guangliang Liu
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA.,Department of Otorhinolaryngology, Sixth Affiliated Hospital, Sun Yat-Sen University, PR China
| | - Junping Ren
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Caroline Go
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Teodora Ivanciuc
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Kolli Deepthi
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Antonella Casola
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Roberto P Garofalo
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Xiaoyong Bao
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.,Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
36
|
Gupta MR, Kolli D, Molteni C, Casola A, Garofalo RP. Paramyxovirus infection regulates T cell responses by BDCA-1+ and BDCA-3+ myeloid dendritic cells. PLoS One 2014; 9:e99227. [PMID: 24918929 PMCID: PMC4053357 DOI: 10.1371/journal.pone.0099227] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/13/2014] [Indexed: 12/24/2022] Open
Abstract
Respiratory syncytial virus (RSV) and human Metapneumovirus (hMPV), viruses belonging to the family Paramyxoviridae, are the most important causes of lower respiratory tract infection in young children. Infections with RSV and hMPV are clinically indistinguishable, and both RSV and hMPV infection have been associated with aberrant adaptive immune responses. Myeloid Dendritic cells (mDCs) play a pivotal role in shaping adaptive immune responses during infection; however, few studies have examined how interactions of RSV and hMPV with individual mDC subsets (BDCA-1+ and BDCA-3+ mDCs) affect the outcome of anti-viral responses. To determine whether RSV and hMPV induce virus-specific responses from each subset, we examined co-stimulatory molecules and cytokines expressed by BDCA-1+ and BDCA-3+ mDCs isolated from peripheral blood after infection with hMPV and RSV, and examined their ability to stimulate T cell proliferation and differentiation. Our data show that RSV and hMPV induce virus-specific and subset-specific patterns of co-stimulatory molecule and cytokine expression. RSV, but not hMPV, impaired the capacity of infected mDCs to stimulate T cell proliferation. Whereas hMPV-infected BDCA-1+ and BDCA-3+ mDCs induced expansion of Th17 cells, in response to RSV, BDCA-1+ mDCs induced expansion of Th1 cells and BDCA-3+ mDCs induced expansion of Th2 cells and Tregs. These results demonstrate a virus-specific and subset-specific effect of RSV and hMPV infection on mDC function, suggesting that these viruses may induce different adaptive immune responses.
Collapse
Affiliation(s)
- Meera R. Gupta
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| | - Deepthi Kolli
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Claudio Molteni
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonella Casola
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Roberto P. Garofalo
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
37
|
Abstract
Respiratory syncytial virus (RSV) is amongst the most important pathogenic infections of childhood and is associated with significant morbidity and mortality. Although there have been extensive studies of epidemiology, clinical manifestations, diagnostic techniques, animal models and the immunobiology of infection, there is not yet a convincing and safe vaccine available. The major histopathologic characteristics of RSV infection are acute bronchiolitis, mucosal and submucosal edema, and luminal occlusion by cellular debris of sloughed epithelial cells mixed with macrophages, strands of fibrin, and some mucin. There is a single RSV serotype with two major antigenic subgroups, A and B. Strains of both subtypes often co-circulate, but usually one subtype predominates. In temperate climates, RSV infections reflect a distinct seasonality with onset in late fall or early winter. It is believed that most children will experience at least one RSV infection by the age of 2 years. There are several key animal models of RSV. These include a model in mice and, more importantly, a bovine model; the latter reflects distinct similarity to the human disease. Importantly, the prevalence of asthma is significantly higher amongst children who are hospitalized with RSV in infancy or early childhood. However, there have been only limited investigations of candidate genes that have the potential to explain this increase in susceptibility. An atopic predisposition appears to predispose to subsequent development of asthma and it is likely that subsequent development of asthma is secondary to the pathogenic inflammatory response involving cytokines, chemokines and their cognate receptors. Numerous approaches to the development of RSV vaccines are being evaluated, as are the use of newer antiviral agents to mitigate disease. There is also significant attention being placed on the potential impact of co-infection and defining the natural history of RSV. Clearly, more research is required to define the relationships between RSV bronchiolitis, other viral induced inflammatory responses, and asthma.
Collapse
Affiliation(s)
- Andrea T. Borchers
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, 451 Health Sciences Drive, Suite 6501, Davis, CA 95616 USA
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, 451 Health Sciences Drive, Suite 6501, Davis, CA 95616 USA
| | - M. Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, 451 Health Sciences Drive, Suite 6501, Davis, CA 95616 USA
| | - Laurel J. Gershwin
- Department of Pathology, Microbiology and Immunology, University of California, Davis, School of Veterinary Medicine, Davis, CA USA
| |
Collapse
|
38
|
Paramyxovirus activation and inhibition of innate immune responses. J Mol Biol 2013; 425:4872-92. [PMID: 24056173 DOI: 10.1016/j.jmb.2013.09.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/12/2013] [Accepted: 09/12/2013] [Indexed: 12/18/2022]
Abstract
Paramyxoviruses represent a remarkably diverse family of enveloped nonsegmented negative-strand RNA viruses, some of which are the most ubiquitous disease-causing viruses of humans and animals. This review focuses on paramyxovirus activation of innate immune pathways, the mechanisms by which these RNA viruses counteract these pathways, and the innate response to paramyxovirus infection of dendritic cells (DC). Paramyxoviruses are potent activators of extracellular complement pathways, a first line of defense that viruses must face during natural infections. We discuss mechanisms by which these viruses activate and combat complement to delay neutralization. Once cells are infected, virus replication drives type I interferon (IFN) synthesis that has the potential to induce a large number of antiviral genes. Here we describe four approaches by which paramyxoviruses limit IFN induction: by limiting synthesis of IFN-inducing aberrant viral RNAs, through targeted inhibition of RNA sensors, by providing viral decoy substrates for cellular kinase complexes, and through direct blocking of the IFN promoter. In addition, paramyxoviruses have evolved diverse mechanisms to disrupt IFN signaling pathways. We describe three general mechanisms, including targeted proteolysis of signaling factors, sequestering cellular factors, and upregulation of cellular inhibitors. DC are exceptional cells with the capacity to generate adaptive immunity through the coupling of innate immune signals and T cell activation. We discuss the importance of innate responses in DC following paramyxovirus infection and their consequences for the ability to mount and maintain antiviral T cells.
Collapse
|
39
|
Céspedes PF, Gonzalez PA, Kalergis AM. Human metapneumovirus keeps dendritic cells from priming antigen-specific naive T cells. Immunology 2013; 139:366-76. [PMID: 23374037 DOI: 10.1111/imm.12083] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 12/30/2012] [Accepted: 01/28/2013] [Indexed: 12/22/2022] Open
Abstract
Human metapneumovirus (hMPV) is the second most common cause of acute lower respiratory tract infections in children, causing a significant public health burden worldwide. Given that hMPV can repeatedly infect the host without major antigenic changes, it has been suggested that hMPV may have evolved molecular mechanisms to impair host adaptive immunity and, more specifically, T-cell memory. Recent studies have shown that hMPV can interfere with superantigen-induced T-cell activation by infecting conventional dendritic cells (DCs). Here, we show that hMPV infects mouse DCs in a restricted manner and induces moderate maturation. Nonetheless, hMPV-infected DCs are rendered inefficient at activating naive antigen-specific CD4(+) T cells (OT-II), which not only display reduced proliferation, but also show a marked reduction in surface activation markers and interleukin-2 secretion. Decreased T-cell activation was not mediated by interference with DC-T-cell immunological synapse formation as recently described for the human respiratory syncytial virus (hRSV), but rather by soluble factors secreted by hMPV-infected DCs. These data suggest that although hMPV infection is restricted within DCs, it is sufficient to interfere with their capacity to activate naive T cells. Altogether, by interfering with DC function and productive priming of antigen-inexperienced T cells, hMPV could impair the generation of long-term immunity.
Collapse
Affiliation(s)
- Pablo F Céspedes
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Santiago, Chile
| | | | | |
Collapse
|
40
|
Han S, Englander EW, Gomez GA, Aronson JF, Rastellini C, Garofalo RP, Kolli D, Quertermous T, Kundu R, Greeley GH. Pancreatitis activates pancreatic apelin-APJ axis in mice. Am J Physiol Gastrointest Liver Physiol 2013; 305:G139-50. [PMID: 23681476 PMCID: PMC3725680 DOI: 10.1152/ajpgi.00370.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pancreatitis is classified into acute pancreatitis (AP) and chronic pancreatitis (CP). Apelin, a small regulatory peptide, is the endogenous ligand for the APJ receptor. Apelin and APJ are expressed in the pancreas. The aims of this study were to examine whether apelin influences the inflammatory and fibrosis responses to pancreatitis in mice and to identify mechanisms behind apelin's activities. Supramaximal cerulein induction of AP or CP caused significant (P < 0.05) elevations in pancreatic apelin and APJ expression. Levels declined during the recovery phases. In apelin gene-knockout mice with pancreatitis, pancreatic neutrophil invasion and myeloperoxidase activity were enhanced significantly, and apelin treatment suppressed both. Apelin exposure reduced CP-induced elevations of extracellular matrix-associated proteins. Apelin inhibited PDGF-simulated connective tissue growth factor production and proliferation of pancreatic stellate cells (PSCs). Serum granulocyte colony-stimulating factor and keratinocyte cytokine levels were higher in apelin gene-knockout than wild-type mice with pancreatitis. Apelin reduced AP- and CP-induced elevations in pancreatic NF-κB activation. Together, these findings imply that the pancreatic apelin-APJ system functions to curb the inflammatory and fibrosis responses during pancreatitis. Furthermore, findings suggest that apelin reduces inflammation and fibrosis by reducing neutrophil recruitment and PSC activity. Inhibition of neutrophil invasion may be mediated by reduced keratinocyte cytokine and granulocyte colony-stimulating factor secretion. Apelin-induced reductions in PSC proliferation and connective tissue growth factor production are putative mechanisms underlying apelin's inhibition of extracellular matrix production. The apelin-associated changes in NF-κB binding may be linked to apelin's regulation of pancreatic inflammatory and fibrosis responses during pancreatitis.
Collapse
Affiliation(s)
- Song Han
- 1Department of Surgery, University of Texas Medical Branch, Galveston, Texas;
| | - Ella W. Englander
- 1Department of Surgery, University of Texas Medical Branch, Galveston, Texas;
| | - Guillermo A. Gomez
- 1Department of Surgery, University of Texas Medical Branch, Galveston, Texas;
| | - Judith F. Aronson
- 4Department of Pathology, University of Texas Medical Branch, Galveston, Texas; and
| | | | - R. P. Garofalo
- 2Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas; ,3Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas;
| | - Deepthi Kolli
- 2Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas;
| | | | | | - George H. Greeley
- 1Department of Surgery, University of Texas Medical Branch, Galveston, Texas;
| |
Collapse
|
41
|
Ren J, Kolli D, Deng J, Fang R, Gong B, Xue M, Casola A, Garofalo RP, Wang T, Bao X. MyD88 controls human metapneumovirus-induced pulmonary immune responses and disease pathogenesis. Virus Res 2013; 176:241-50. [PMID: 23845303 DOI: 10.1016/j.virusres.2013.06.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 06/23/2013] [Accepted: 06/26/2013] [Indexed: 12/14/2022]
Abstract
Human metapneumovirus (hMPV) is a common cause of lung and airway infections in infants and young children. Recently, we and others have shown that hMPV infection induces Toll-like receptor (TLR)-dependent cellular signaling. However, the contribution of TLR-mediated signaling in host defenses against pulmonary hMPV infection and associated disease pathogenesis has not been elucidated. In this study, mice deficient in MyD88, a common adaptor of TLRs, was used to investigate the contribution of TLRs to in vivo pulmonary response to hMPV infection. MyD88(-/-) mice have significantly reduced pulmonary inflammation and associated disease compared with wild-type (WT) C57BL/6 mice after intranasal infection with hMPV. hMPV-induced cytokines and chemokines in bronchoalveolar lavage fluid (BALF) and isolated lung conventional dendritic cells (cDC) are also significantly impaired by MyD88 deletion. In addition, we found that MyD88 is required for the recruitment of DC, T cells, and other immune cells to the lungs, and for the functional regulation of DC and T cells in response to hMPV infection. Taken together, our data indicate that MyD88-mediated pathways are essential for the pulmonary immune and pathogenic responses to this viral pathogen.
Collapse
Affiliation(s)
- Junping Ren
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Gupta MR, Kolli D, Garofalo RP. Differential response of BDCA-1+ and BDCA-3+ myeloid dendritic cells to respiratory syncytial virus infection. Respir Res 2013; 14:71. [PMID: 23829893 PMCID: PMC3708742 DOI: 10.1186/1465-9921-14-71] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 06/28/2013] [Indexed: 12/24/2022] Open
Abstract
Background Respiratory syncytial virus (RSV) is the leading cause of respiratory infections in children, elderly, and immunocompromised individuals. Severe infection is associated with short- and long-term morbidity including pneumonia, recurrent wheezing, and abnormal pulmonary function, and several lines of evidence indicate that impaired adaptive immune responses during infection are critical in the pathophysiology of RSV-mediated disease. Myeloid Dendritic cells (mDCs) play a pivotal role in shaping antiviral immune responses in the respiratory tract; however, few studies have examined the interactions between RSV and individual mDC subsets. In this study, we examined the effect of RSV on the functional response of primary mDC subsets (BDCA-1+ and BDCA-3+) isolated from peripheral blood. Methods BDCA-1+ and BDCA-3+ mDCs were isolated from the peripheral blood of healthy adults using FACS sorting. Donor-matched BDCA-1+ and BDCA-3+ mDCs were infected with RSV at a multiplicity of infection (MOI) of 5 for 40 hours. After infection, cells were analyzed for the expression of costimulatory molecules (CD86, CD80, and PD-L1), cytokine production, and the ability to stimulate allogenic CD4+ T cell proliferation. Results Both BDCA-1+ and BDCA-3+ mDCs were susceptible to infection with RSV and demonstrated enhanced expression of CD86, and the inhibitory costimulatory molecules CD80 and PD-L1. Compared to BDCA-3+ mDCs, RSV-infected BDCA-1+ mDC produced a profile of cytokines and chemokines predominantly associated with pro-inflammatory responses (IL-1β, IL-6, IL-12, MIP-1α, and TNF-α), and both BDCA-1+ and BDCA-3+ mDCs were found to produce IL-10. Compared to uninfected mDCs, RSV-infected BDCA-1+ and BDCA-3+ mDCs demonstrated a reduced capacity to stimulate T cell proliferation. Conclusions RSV infection induces a distinct pattern of costimulatory molecule expression and cytokine production by BDCA-1+ and BDCA-3+ mDCs, and impairs their ability to stimulate T cell proliferation. The differential expression of CD86 and pro-inflammatory cytokines by highly purified mDC subsets in response to RSV provides further evidence that BDCA-1+ and BDCA-3+ mDCs have distinct roles in coordinating the host immune response during RSV infection. Findings of differential expression of PD-L1 and IL-10 by infected mDCs, suggests possible mechanisms by which RSV is able to impair adaptive immune responses.
Collapse
Affiliation(s)
- Meera R Gupta
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | | | | |
Collapse
|
43
|
Guerrero-Plata A. Dendritic cells in human Pneumovirus and Metapneumovirus infections. Viruses 2013; 5:1553-70. [PMID: 23787776 PMCID: PMC3717721 DOI: 10.3390/v5061553] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 05/24/2013] [Accepted: 06/06/2013] [Indexed: 12/27/2022] Open
Abstract
Lung dendritic cells (DC) play a fundamental role in sensing invading pathogens, as well as in the control of tolerogenic responses in the respiratory tract. Their strategic localization at the site of pathogen entry makes them particularly susceptible to initial viral invasion. Human respiratory syncytial virus (hRSV) and human metapneumovirus (hMPV) belong to the Paramyxoviridae family, within the Pneumovirus and Metapneumovirus genera, respectively. hRSV and hMPV are significant human respiratory pathogens that cause similar clinical manifestations and affect many of the same subpopulations. However, they differentially activate the host immune response, including DC, which represents a fundamental link between the innate and adaptive immune response. In this review, the role of DC in the immune response against hRSV and hMPV infections, as well as the inhibitory effects of these paramyxoviruses on the DC immunity will be discussed.
Collapse
|
44
|
Garg R, Shrivastava P, van Drunen Littel-van den Hurk S. The role of dendritic cells in innate and adaptive immunity to respiratory syncytial virus, and implications for vaccine development. Expert Rev Vaccines 2013; 11:1441-57. [PMID: 23252388 DOI: 10.1586/erv.12.117] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Respiratory syncytial virus (RSV) is a common human pathogen that causes cold-like symptoms in most healthy adults and children. However, RSV often moves into the lower respiratory tract in infants and young children predisposed to respiratory illness, making it the most common cause of pediatric broncheolitis and pneumonia. The development of an appropriate balanced immune response is critical for recovery from RSV, while an unbalanced and/or excessively vigorous response may lead to immunopathogenesis. Different dendritic cell (DC) subsets influence the magnitude and quality of the host response to RSV infection, with myeloid DCs mediating and plasmacytoid DCs modulating immunopathology. Furthermore, stimulation of DCs through Toll-like receptors is essential for induction of protective immunity to RSV. These characteristics have implications for the rational design of a RSV vaccine.
Collapse
Affiliation(s)
- Ravendra Garg
- VIDO-Intervac, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | | | | |
Collapse
|
45
|
Human parainfluenza virus type 2 V protein inhibits TRAF6-mediated ubiquitination of IRF7 to prevent TLR7- and TLR9-dependent interferon induction. J Virol 2013; 87:7966-76. [PMID: 23678181 DOI: 10.1128/jvi.03525-12] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Paramyxovirus V proteins block Toll-like receptor 7 (TLR7)- and TLR9-dependent signaling leading to alpha interferon production. Our recent study has provided evidence that interaction of the V proteins with IRF7 is important for the blockade. However, the detailed mechanisms still remain unclear. Here we reexamined the interaction of the human parainfluenza virus type 2 (HPIV2) V protein with signaling molecules involved in TLR7/9-dependent signaling. Immunoprecipitation experiments in HEK293T cells transfected with V protein and one of the signaling molecules revealed that the V protein interacted with not only IRF7 but also TRAF6, IKKα, and MyD88. Whereas overexpression of TRAF6 markedly enhanced the level of V protein associating with IRF7, IKKα, and MyD88 in HEK293T cells, the level of V protein associating with TRAF6 was little affected by overexpression of IRF7, IKKα, and MyD88. Moreover, knockdown or knockout of endogenous TRAF6 in HEK293T or mouse embryonic fibroblast cells resulted in dissociation of the V protein from IRF7, IKKα, and MyD88. These results demonstrate that binding of the V protein to IRF7, IKKα, and MyD88 is largely indirect and mediated by endogenous TRAF6. It was found that the V protein inhibited TRAF6-mediated lysine 63 (K63)-linked polyubiquitination of IRF7, which is prerequisite for IRF7 activation. Disruption of the tryptophan-rich motif of the V protein significantly affected its TRAF6-binding efficiency, which correlated well with the magnitude of inhibition of K63-linked polyubiquitination and the resultant activation of IRF7. Taken together, these results suggest that the HPIV2 V protein prevents TLR7/9-dependent interferon induction by inhibiting TRAF6-mediated K63-linked polyubiquitination of IRF7.
Collapse
|
46
|
Bao X, Kolli D, Ren J, Liu T, Garofalo RP, Casola A. Human metapneumovirus glycoprotein G disrupts mitochondrial signaling in airway epithelial cells. PLoS One 2013; 8:e62568. [PMID: 23626834 PMCID: PMC3633857 DOI: 10.1371/journal.pone.0062568] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 03/22/2013] [Indexed: 12/21/2022] Open
Abstract
Human metapneumovirus (hMPV) is a recently identified RNA virus belonging to the Paramyxoviridae family. It is a common cause of respiratory tract infections in children, adults, and immunocompromised patients, for which no specific treatment or vaccine is available. Recent investigations in our lab identified hMPV glycoprotein G as an important virulence factor, as a recombinant virus lacking the G protein (rhMPV-ΔG) exhibited enhanced production of important immune and antiviral mediators, such as cytokines, chemokines and type I interferon (IFN) in airway epithelial cells, and expression of G protein alone inhibits cellular signaling dependent on retinoic induced gene (RIG)-I, a RNA helicase with a fundamental role in initiating hMPV-induced cellular responses. In this study, we have further investigated the mechanism underlying the inhibitory role of hMPV G protein on RIG-I-dependent signaling. We found that the interaction of hMPV G with RIG-I occurs primarily through the CARD domains of RIG-I N-terminus, preventing RIG-I association with the adaptor protein MAVS (mitochondrial antiviral signaling protein), recruitment of RIG-I to mitochondria, as well as the interaction between mitochondria and mitochondria-associated membrane (MAM) component of the endoplasmic reticulum (ER), which contains STINGS, an important part of the viral-induced RIG-I/MAVS signaling pathway, leading in the end to the inhibition of cytokine, chemokine and type I IFN expression. Mutagenesis analysis showed that hMPV G protein cytoplasmic domain played a major role in the observed inhibitory activity, and recombinant viruses expressing a G protein with amino acid substitution in position 2 and 3 recapitulated most of the phenotype observed with rhMPV-ΔG mutant upon infection of airway epithelial cells.
Collapse
Affiliation(s)
- Xiaoyong Bao
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- * E-mail: (XB); (AC)
| | - Deepthi Kolli
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Junping Ren
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Tianshuang Liu
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Roberto P. Garofalo
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- Sealy Center for Vaccine Development, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Antonella Casola
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- Sealy Center for Vaccine Development, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- * E-mail: (XB); (AC)
| |
Collapse
|
47
|
Gupta M, Lo MK, Spiropoulou CF. Activation and cell death in human dendritic cells infected with Nipah virus. Virology 2013; 441:49-56. [PMID: 23587249 DOI: 10.1016/j.virol.2013.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 11/13/2012] [Accepted: 03/04/2013] [Indexed: 01/23/2023]
Abstract
Nipah virus (NiV) is a highly pathogenic paramyxovirus that causes pulmonary disease and encephalitis in humans with 40-70% fatality. Interactions between NiV and the human immune system remain poorly understood. Here, we demonstrate the effects of NiV infection on DC and T cell function. Using an in vitro system, we found that NiV infects and replicates at low levels in DCs and induces the expression of TNF-α, IL-1α, IL-1β, IL-8, and IP-10. NiV infection activates DCs, and upregulates the expression of CD40, CD80, and CD86. Also have reduced levels of bcl2 and high levels of active caspase 3, suggesting the induction of apoptosis. DCs infected by NiV are unable to efficiently prime CD4 and CD8 T cells, but instead induce apoptosis in T cells. Interestingly, DCs treated with inactivated NiV also show signs of apoptosis. These findings indicate that NiV infected DCs could play an important role in NiV pathogenesis.
Collapse
Affiliation(s)
- Manisha Gupta
- Viral Special Pathogens Branch, NCEZID, DHCPP, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA.
| | | | | |
Collapse
|
48
|
Jang S, Smit J, Kallal LE, Lukacs NW. Respiratory syncytial virus infection modifies and accelerates pulmonary disease via DC activation and migration. J Leukoc Biol 2013; 94:5-15. [PMID: 23293372 DOI: 10.1189/jlb.0412195] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In the present studies, we have established that RSV can elicit a more pathogenic environment dependent on improper DC-associated sensitization. Our initial studies demonstrated that RSV, but not influenza, infection during an allergen exposure into the airway induced a more severe allergen response. The RSV-induced exacerbation included an increased Th2 cytokine response and pathophysiology as monitored by AHR and mucus overproduction. DCs played a central role in the allergen-induced responses, as instilling RSV-infected BMDC into the airway could recapitulate a live virus challenge. With the use of CCR6-/- mice that have a primary defect in the recruitment of mDC subsets, reduced exacerbation of disease was observed when RSV was administered along with allergen. Furthermore, sensitization of mice with RSV-infected BMDC into the airway produced a more severe immune response to a live virus challenge. Subsequently, using RSV-infected BMDC from CCR7-/- mice (that do not migrate efficiently to LNs) to sensitize the exacerbated response demonstrated that the response was dependent on DC migration to the LN. Finally, the ability of RSV-infected DCs to elicit an exacerbated, allergen-induced pathogenic response could be maintained for as long as 3 weeks, suggesting that RSV-infected DCs themselves created an altered immune environment that impacts off-target mucosal responses that could have prolonged effects.
Collapse
Affiliation(s)
- Sihyug Jang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
49
|
Critical role of MDA5 in the interferon response induced by human metapneumovirus infection in dendritic cells and in vivo. J Virol 2012; 87:1242-51. [PMID: 23152520 DOI: 10.1128/jvi.01213-12] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Human metapneumovirus (hMPV) is a respiratory paramyxovirus of global clinical relevance. Despite the substantial knowledge generated during the last 10 years about hMPV infection, information regarding the activation of the immune response against this virus remains largely unknown. In this study, we demonstrated that the helicase melanoma differentiation-associated gene 5 (MDA5) is essential to induce the interferon response after hMPV infection in human and mouse dendritic cells as well as in an experimental mouse model of infection. Our findings in vitro and in vivo showed that MDA5 is required for the expression and activation of interferon (IFN) regulatory factors (IRFs). hMPV infection induces activation of IRF-3, and it regulates the expression of IRF-7. However, both IRF-3 and IRF-7 are critical for the production of type I and type III IFNs. In addition, our in vivo studies in hMPV-infected mice indicated that MDA5 alters viral clearance, enhances disease severity and pulmonary inflammation, and regulates the production of cytokines and chemokines in response to hMPV. These findings are relevant for a better understanding of the pathogenesis of hMPV infection.
Collapse
|
50
|
Chakraborty K, Zhou Z, Wakamatsu N, Guerrero-Plata A. Interleukin-12p40 modulates human metapneumovirus-induced pulmonary disease in an acute mouse model of infection. PLoS One 2012; 7:e37173. [PMID: 22606349 PMCID: PMC3351396 DOI: 10.1371/journal.pone.0037173] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 04/16/2012] [Indexed: 11/18/2022] Open
Abstract
The mechanisms that regulate the host immune response induced by human metapneumovirus (hMPV), a newly-recognized member of the Paramyxoviridae family, are largely unknown. Cytokines play an important role in modulating inflammatory responses during viral infections. IL-12p40, a known important mediator in limiting lung inflammation, is induced by hMPV and its production is sustained after the resolution phase of infection suggesting that this cytokine plays a role in the immune response against hMPV. In this work, we demonstrated that in mice deficient in IL-12p40, hMPV infection induced an exacerbated pulmonary inflammatory response and mucus production, altered cytokine response, and decreased lung function. However, hMPV infection in these mice does not have an effect on viral replication. These results identify an important regulatory role of IL-12p40 in hMPV infection.
Collapse
Affiliation(s)
- Krishnendu Chakraborty
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Zehua Zhou
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Nobuko Wakamatsu
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Antonieta Guerrero-Plata
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
- Center for Experimental Infectious Disease Research, Louisiana State University, Baton Rouge, Louisiana, United States of America
- * E-mail:
| |
Collapse
|