1
|
Georgakopoulou VE. Insights from respiratory virus co-infections. World J Virol 2024; 13:98600. [DOI: 10.5501/wjv.v13.i4.98600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 10/18/2024] Open
Abstract
Respiratory viral co-infections present significant challenges in clinical settings due to their impact on disease severity and patient outcomes. Current diagnostic methods often miss these co-infections, complicating the epidemiology and management of these cases. Research, primarily conducted in vitro and in vivo, suggests that co-infections can lead to more severe illnesses, increased hospitalization rates, and greater healthcare utilization, especially in high-risk groups such as children, the elderly, and immunocompromised individuals. Common co-infection patterns, risk factors, and their impact on disease dynamics highlight the need for advanced diagnostic techniques and tailored therapeutic strategies. Understanding the virological interactions and immune response modulation during co-infections is crucial for developing effective public health interventions and improving patient outcomes. Future research should focus on the molecular mechanisms of co-infection and the development of specific therapies to mitigate the adverse effects of these complex infections.
Collapse
Affiliation(s)
- Vasiliki E Georgakopoulou
- Department of Pathophysiology, Laiko General Hospital, Medical School of National and Kapodistrian University of Athens, Athens 11527, Greece
| |
Collapse
|
2
|
Bergeron HC, Crabtree J, Nagy T, Martin DE, Tripp RA. Probenecid Inhibits Human Metapneumovirus (HMPV) Replication In Vitro and in BALB/c Mice. Viruses 2024; 16:1087. [PMID: 39066249 PMCID: PMC11281683 DOI: 10.3390/v16071087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/19/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Human metapneumovirus (HMPV) is an important cause of acute respiratory tract infection and causes significant morbidity and mortality. There is no specific antiviral drug to treat HMPV or vaccine to prevent HMPV. This study determined if probenecid, a host-targeting antiviral drug, had prophylactic (pre-virus) or therapeutic (post-virus) efficacy to inhibit HMPV replication in LLC-MK2 cells in vitro and in the lungs of BALB/c mice. This study showed that ≥0.5 μM probenecid significantly inhibited HMPV replication in vitro, and 2-200 mg/kg probenecid prophylaxis or treatment reduced HMPV replication in BALB/c mice.
Collapse
Affiliation(s)
- Harrison C. Bergeron
- Department of Infectious Diseases, University of Georgia, Athens, GA 30605, USA; (H.C.B.)
| | - Jackelyn Crabtree
- Department of Infectious Diseases, University of Georgia, Athens, GA 30605, USA; (H.C.B.)
| | - Tamas Nagy
- Department of Pathology, University of Georgia, Athens, GA 30605, USA
| | | | - Ralph A. Tripp
- Department of Infectious Diseases, University of Georgia, Athens, GA 30605, USA; (H.C.B.)
- TrippBio, Inc., Jacksonville, FL 32256, USA;
| |
Collapse
|
3
|
Zhang Y, Xu J, Miranda-Katz M, Sojati J, Tollefson SJ, Manni ML, Alcorn JF, Sarkar SN, Williams JV. Distinct roles for type I and type III interferons in virulent human metapneumovirus pathogenesis. PLoS Pathog 2024; 20:e1011840. [PMID: 38315735 PMCID: PMC10868789 DOI: 10.1371/journal.ppat.1011840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 02/15/2024] [Accepted: 11/17/2023] [Indexed: 02/07/2024] Open
Abstract
Human metapneumovirus (HMPV) is an important cause of acute lower respiratory infection in children and adults worldwide. There are four genetic subgroups of HMPV and both neutralizing antibodies and T cells contribute to protection. However, little is known about mechanisms of pathogenesis and most published work is based on a few extensively passaged, laboratory-adapted strains of HMPV. In this study, we isolated and characterized a panel of low passage HMPV clinical isolates representing all four genetic subgroups. The clinical isolates exhibited lower levels of in vitro replication compared to a lab-adapted strain. We compared disease phenotypes using a well-established mouse model. Several virulent isolates caused severe weight loss, lung pathology, airway dysfunction, and fatal disease in mice, which was confirmed in three inbred mouse strains. Disease severity did not correlate with lung viral titer, as virulent strains exhibited restricted replication in the lower airway. Virulent HMPV isolates were associated with markedly increased proinflammatory cytokine production and neutrophil influx; however, depletion of neutrophils or genetic ablation of inflammasome components did not reverse disease. Virulent clinical isolates induced markedly increased type I and type III interferon (IFN) secretion in vitro and in vivo. STAT1/2-deficient mice lacking both type I and type III IFN signaling showed reduced disease severity and increased lung viral replication. Inhibition of type I IFN signaling using a blocking antibody or genetic ablation of the type I IFN receptor reduced pathology with minimal effect on viral replication. Conversely, blockade of type III IFN signaling with a neutralizing antibody or genetic ablation of the IFN-lambda receptor had no effect on pathogenesis but restored viral replication. Collectively, these results demonstrate distinct roles for type I and type III IFN in HMPV pathogenesis and immunity.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jiuyang Xu
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Tsinghua University School of Medicine, Beijing, China
| | - Margot Miranda-Katz
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jorna Sojati
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Sharon J. Tollefson
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Michelle L. Manni
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - John F. Alcorn
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Saumendra N. Sarkar
- Department of Microbiology & Molecular Genetics, University of Pittsburgh School of Medicine, Pennsylvania, United States of America
| | - John V. Williams
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology & Molecular Genetics, University of Pittsburgh School of Medicine, Pennsylvania, United States of America
- Institute for Infection, Inflammation, and Immunity in Children, University of Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
4
|
Yim KC, Mousa JJ, Blanco JCG, Kim S, Boukhvalova MS. Human Metapneumovirus (hMPV) Infection and MPV467 Treatment in Immunocompromised Cotton Rats Sigmodon hispidus. Viruses 2023; 15:476. [PMID: 36851691 PMCID: PMC9966515 DOI: 10.3390/v15020476] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Human metapneumovirus (hMPV) is an important cause of respiratory disease in immunocompromised individuals, yet hMPV infection has not been modeled before in immunocompromised animals. In this work, cotton rats S. hispidus immunosuppressed by cyclophosphamide were infected with hMPV, and viral replication and pulmonary inflammation in these animals were compared to those in normal hMPV-infected S. hispidus. The efficacy of prophylactic and therapeutic administration of the anti-hMPV antibody MPV467 was also evaluated. Immunosuppressed animals had higher pulmonary and nasal titers of hMPV on day 5 post-infection compared to normal animals, and large amounts of hMPV were still present in the respiratory tract of immunosuppressed animals on days 7 and 9 post-infection, indicating prolonged viral replication. Immunosuppression was accompanied by reduced pulmonary histopathology in hMPV-infected cotton rats compared to normal animals; however, a delayed increase in pathology and pulmonary chemokine expression was seen in immunosuppressed cotton rats. Prophylactic and therapeutic MPV467 treatments protected both upper and lower respiratory tracts against hMPV infection. The lung pathology and pulmonary expression of IP-10 and MIP-1α mRNA were reduced by therapeutic MPV467 administration. These results indicate that immunosuppressed cotton rats represent a useful model for studying hMPV pathogenesis and for evaluating therapeutics that could alleviate hMPV-induced disease in immunocompromised subjects.
Collapse
Affiliation(s)
- Kevin C. Yim
- Sigmovir Biosystems, Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD 20850, USA
| | - Jarrod J. Mousa
- Center for Vaccines and Immunology, Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
- Department of Biochemistry and Molecular Biology, Franklin College of Arts and Sciences, University of Georgia, Athens, GA 30602, USA
| | - Jorge C. G. Blanco
- Sigmovir Biosystems, Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD 20850, USA
| | - Sonnie Kim
- NIH/NIAID, Respiratory Diseases Branch, Division of Microbiology and Infectious Diseases, Rockville, MD 20852, USA
| | - Marina S. Boukhvalova
- Sigmovir Biosystems, Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD 20850, USA
| |
Collapse
|
5
|
Chu JT, Gu H, Sun W, Fan RL, Nicholls JM, Valkenburg SA, Poon LL. Heterosubtypic immune pressure accelerates emergence of influenza A virus escape phenotypes in mice. Virus Res 2023; 323:198991. [PMID: 36302472 PMCID: PMC10194115 DOI: 10.1016/j.virusres.2022.198991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 11/07/2022]
Abstract
Rapid antigenic evolution of the influenza A virus surface antigen hemagglutinin undermines protection conferred by seasonal vaccines. Protective correlates targeted by universal vaccines such as cytotoxic T cells or HA stem directed broadly neutralizing antibodies have been shown to select for immune escape mutants during infection. We developed an in vivo serial passage mouse model for viral adaptation and used next generation sequencing to evaluate full genome viral evolution in the context of broadly protective immunity. Heterosubtypic immune pressure increased the incidence of genome-wide single nucleotide variants, though mutations found in early adapted populations were predominantly stochastic in nature. Prolonged adaptation under heterosubtypic immune selection resulted in the manifestation of highly virulent phenotypes that ablated vaccine mediated protection from mortality. High frequency mutations unique to escape phenotypes were identified within the polymerase encoding segments. These findings suggest that a suboptimial usage of population-wide universal influenza vaccine may drive formation of escape variants attributed to polygenic changes.
Collapse
Affiliation(s)
- Julie Ts Chu
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Haogao Gu
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Wanying Sun
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Rebecca Ly Fan
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - John M Nicholls
- Department of Pathology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Sophie A Valkenburg
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Leo Lm Poon
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China; HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China; Centre for Immunology & Infection, Hong Kong Science Park, Hong Kong Special Administrative Region, China.
| |
Collapse
|
6
|
Strickland BA, Rajagopala SV, Kamali A, Shilts MH, Pakala SB, Boukhvalova MS, Yooseph S, Blanco JCG, Das SR. Species-specific transcriptomic changes upon respiratory syncytial virus infection in cotton rats. Sci Rep 2022; 12:16579. [PMID: 36195733 PMCID: PMC9531660 DOI: 10.1038/s41598-022-19810-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Abstract
The cotton rat (Sigmodon) is the gold standard pre-clinical small animal model for respiratory viral pathogens, especially for respiratory syncytial virus (RSV). However, without a reference genome or a published transcriptome, studies requiring gene expression analysis in cotton rats are severely limited. The aims of this study were to generate a comprehensive transcriptome from multiple tissues of two species of cotton rats that are commonly used as animal models (Sigmodon fulviventer and Sigmodon hispidus), and to compare and contrast gene expression changes and immune responses to RSV infection between the two species. Transcriptomes were assembled from lung, spleen, kidney, heart, and intestines for each species with a contig N50 > 1600. Annotation of contigs generated nearly 120,000 gene annotations for each species. The transcriptomes of S. fulviventer and S. hispidus were then used to assess immune response to RSV infection. We identified 238 unique genes that are significantly differentially expressed, including several genes implicated in RSV infection (e.g., Mx2, I27L2, LY6E, Viperin, Keratin 6A, ISG15, CXCL10, CXCL11, IRF9) as well as novel genes that have not previously described in RSV research (LG3BP, SYWC, ABEC1, IIGP1, CREB1). This study presents two comprehensive transcriptome references as resources for future gene expression analysis studies in the cotton rat model, as well as provides gene sequences for mechanistic characterization of molecular pathways. Overall, our results provide generalizable insights into the effect of host genetics on host-virus interactions, as well as identify new host therapeutic targets for RSV treatment and prevention.
Collapse
Affiliation(s)
- Britton A Strickland
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Seesandra V Rajagopala
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, 1211 21st Avenue South, S2108 Medical Center North, Nashville, TN, 37232, USA
| | - Arash Kamali
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
| | - Meghan H Shilts
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, 1211 21st Avenue South, S2108 Medical Center North, Nashville, TN, 37232, USA
| | - Suman B Pakala
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, 1211 21st Avenue South, S2108 Medical Center North, Nashville, TN, 37232, USA
| | - Marina S Boukhvalova
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
| | - Shibu Yooseph
- Department of Computer Science, Genomics and Bioinformatics Cluster, University of Central Florida, Orlando, FL, USA
| | - Jorge C G Blanco
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA.
| | - Suman R Das
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, 1211 21st Avenue South, S2108 Medical Center North, Nashville, TN, 37232, USA.
| |
Collapse
|
7
|
Zoonotic Origins of Human Metapneumovirus: A Journey from Birds to Humans. Viruses 2022; 14:v14040677. [PMID: 35458407 PMCID: PMC9028271 DOI: 10.3390/v14040677] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 01/13/2023] Open
Abstract
Metapneumoviruses, members of the family Pneumoviridae, have been identified in birds (avian metapneumoviruses; AMPV’s) and humans (human metapneumoviruses; HMPV’s). AMPV and HMPV are closely related viruses with a similar genomic organization and cause respiratory tract illnesses in birds and humans, respectively. AMPV can be classified into four subgroups, A–D, and is the etiological agent of turkey rhinotracheitis and swollen head syndrome in chickens. Epidemiological studies have indicated that AMPV also circulates in wild bird species which may act as reservoir hosts for novel subtypes. HMPV was first discovered in 2001, but retrospective studies have shown that HMPV has been circulating in humans for at least 50 years. AMPV subgroup C is more closely related to HMPV than to any other AMPV subgroup, suggesting that HMPV has evolved from AMPV-C following zoonotic transfer. In this review, we present a historical perspective on the discovery of metapneumoviruses and discuss the host tropism, pathogenicity, and molecular characteristics of the different AMPV and HMPV subgroups to provide increased focus on the necessity to better understand the evolutionary pathways through which HMPV emerged as a seasonal endemic human respiratory virus.
Collapse
|
8
|
Chupin C, Pizzorno A, Traversier A, Brun P, Ogonczyk-Makowska D, Padey B, Milesi C, Dulière V, Laurent E, Julien T, Galloux M, Lina B, Eléouët JF, Moreau K, Hamelin ME, Terrier O, Boivin G, Dubois J, Rosa-Calatrava M. Avian Cell Line DuckCelt ®-T17 Is an Efficient Production System for Live-Attenuated Human Metapneumovirus Vaccine Candidate Metavac ®. Vaccines (Basel) 2021; 9:vaccines9101190. [PMID: 34696298 PMCID: PMC8540687 DOI: 10.3390/vaccines9101190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 12/14/2022] Open
Abstract
The development of a live-attenuated vaccine (LAV) for the prevention of human metapneumovirus (HMPV) infection is often hampered by the lack of highly efficient and scalable cell-based production systems that support eventual global vaccine production. Avian cell lines cultivated in suspension compete with traditional cell platforms used for viral vaccine manufacture. We investigated whether the DuckCelt®-T17 avian cell line (Vaxxel), previously described as an efficient production system for several influenza strains, could also be used to produce a new HMPV LAV candidate (Metavac®, SH gene-deleted A1/C-85473 HMPV). To that end, we characterized the operational parameters of MOI, cell density, and trypsin addition to achieve the optimal production of Metavac®, and demonstrated that the DuckCelt®-T17 cell line is permissive and well-adapted to the production of the wild-type A1/C-85473 HMPV and the Metavac® vaccine candidate. Moreover, our results confirmed that the LAV candidate produced in DuckCelt®-T17 cells conserves its advantageous replication properties in LLC-MK2 and 3D-reconstituted human airway epithelium models, and its capacity to induce efficient neutralizing antibodies in a BALB/c mouse model. Our results suggest that the DuckCelt®-T17 avian cell line is a very promising platform for the scalable in-suspension serum-free production of the HMPV-based LAV candidate Metavac®.
Collapse
Affiliation(s)
- Caroline Chupin
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (C.C.); (A.P.); (A.T.); (P.B.); (B.P.); (C.M.); (V.D.); (E.L.); (T.J.); (B.L.); (O.T.)
- Vaxxel, 43 Boulevard du Onze Novembre 1918, 69100 Villeurbanne, France
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
| | - Andrés Pizzorno
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (C.C.); (A.P.); (A.T.); (P.B.); (B.P.); (C.M.); (V.D.); (E.L.); (T.J.); (B.L.); (O.T.)
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
| | - Aurélien Traversier
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (C.C.); (A.P.); (A.T.); (P.B.); (B.P.); (C.M.); (V.D.); (E.L.); (T.J.); (B.L.); (O.T.)
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France
| | - Pauline Brun
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (C.C.); (A.P.); (A.T.); (P.B.); (B.P.); (C.M.); (V.D.); (E.L.); (T.J.); (B.L.); (O.T.)
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France
| | - Daniela Ogonczyk-Makowska
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Québec, QC G1V 4G2, Canada
| | - Blandine Padey
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (C.C.); (A.P.); (A.T.); (P.B.); (B.P.); (C.M.); (V.D.); (E.L.); (T.J.); (B.L.); (O.T.)
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
| | - Cédrine Milesi
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (C.C.); (A.P.); (A.T.); (P.B.); (B.P.); (C.M.); (V.D.); (E.L.); (T.J.); (B.L.); (O.T.)
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France
| | - Victoria Dulière
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (C.C.); (A.P.); (A.T.); (P.B.); (B.P.); (C.M.); (V.D.); (E.L.); (T.J.); (B.L.); (O.T.)
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France
| | - Emilie Laurent
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (C.C.); (A.P.); (A.T.); (P.B.); (B.P.); (C.M.); (V.D.); (E.L.); (T.J.); (B.L.); (O.T.)
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France
| | - Thomas Julien
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (C.C.); (A.P.); (A.T.); (P.B.); (B.P.); (C.M.); (V.D.); (E.L.); (T.J.); (B.L.); (O.T.)
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France
| | - Marie Galloux
- Unité de Virologie et Immunologie Moléculaires, UVSQ, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (M.G.); (J.-F.E.)
| | - Bruno Lina
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (C.C.); (A.P.); (A.T.); (P.B.); (B.P.); (C.M.); (V.D.); (E.L.); (T.J.); (B.L.); (O.T.)
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
| | - Jean-François Eléouët
- Unité de Virologie et Immunologie Moléculaires, UVSQ, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (M.G.); (J.-F.E.)
| | - Karen Moreau
- CIRI, Centre International de Recherche en Infectiologie, Team STAPHPATH, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France;
| | - Marie-Eve Hamelin
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Québec, QC G1V 4G2, Canada
| | - Olivier Terrier
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (C.C.); (A.P.); (A.T.); (P.B.); (B.P.); (C.M.); (V.D.); (E.L.); (T.J.); (B.L.); (O.T.)
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
| | - Guy Boivin
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Québec, QC G1V 4G2, Canada
| | - Julia Dubois
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (C.C.); (A.P.); (A.T.); (P.B.); (B.P.); (C.M.); (V.D.); (E.L.); (T.J.); (B.L.); (O.T.)
- Vaxxel, 43 Boulevard du Onze Novembre 1918, 69100 Villeurbanne, France
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
- Correspondence: (J.D.); (M.R.-C.)
| | - Manuel Rosa-Calatrava
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (C.C.); (A.P.); (A.T.); (P.B.); (B.P.); (C.M.); (V.D.); (E.L.); (T.J.); (B.L.); (O.T.)
- International Associated Laboratory RespiVir (LIA VirPath-LVMC France-Québec), Université Laval, Québec, QC G1V 4G2, Canada, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France; (D.O.-M.); (M.-E.H.); (G.B.)
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France
- Correspondence: (J.D.); (M.R.-C.)
| |
Collapse
|
9
|
Bhattacharya S, Agarwal S, Shrimali NM, Guchhait P. Interplay between hypoxia and inflammation contributes to the progression and severity of respiratory viral diseases. Mol Aspects Med 2021; 81:101000. [PMID: 34294412 PMCID: PMC8287505 DOI: 10.1016/j.mam.2021.101000] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/07/2021] [Accepted: 07/16/2021] [Indexed: 02/07/2023]
Abstract
History of pandemics is dominated by viral infections and specifically respiratory viral diseases like influenza and COVID-19. Lower respiratory tract infection is the fourth leading cause of death worldwide. Crosstalk between resultant inflammation and hypoxic microenvironment may impair ventilatory response of lungs. This reduces arterial partial pressure of oxygen, termed as hypoxemia, which is observed in a section of patients with respiratory virus infections including SARS-CoV-2 (COVID-19). In this review, we describe the interplay between inflammation and hypoxic microenvironment in respiratory viral infection and its contribution to disease pathogenesis.
Collapse
Affiliation(s)
- Sulagna Bhattacharya
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India; School of Biotechnology, Kalinga Institute of Industrial Technology, Orissa, India
| | - Sakshi Agarwal
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Nishith M Shrimali
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Prasenjit Guchhait
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India.
| |
Collapse
|
10
|
Strickland BA, Patel MC, Shilts MH, Boone HH, Kamali A, Zhang W, Stylos D, Boukhvalova MS, Rosas-Salazar C, Yooseph S, Rajagopala SV, Blanco JCG, Das SR. Microbial community structure and composition is associated with host species and sex in Sigmodon cotton rats. Anim Microbiome 2021; 3:29. [PMID: 33863395 PMCID: PMC8051552 DOI: 10.1186/s42523-021-00090-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/20/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The cotton rat (genus Sigmodon) is an essential small animal model for the study of human infectious disease and viral therapeutic development. However, the impact of the host microbiome on infection outcomes has not been explored in this model, partly due to the lack of a comprehensive characterization of microbial communities across different cotton rat species. Understanding the dynamics of their microbiome could significantly help to better understand its role when modeling viral infections in this animal model. RESULTS We examined the bacterial communities of the gut and three external sites (skin, ear, and nose) of two inbred species of cotton rats commonly used in research (S. hispidus and S. fulviventer) by using 16S rRNA gene sequencing, constituting the first comprehensive characterization of the cotton rat microbiome. We showed that S. fulviventer maintained higher alpha diversity and richness than S. hispidus at external sites (skin, ear, nose), but there were no differentially abundant genera. However, S. fulviventer and S. hispidus had distinct fecal microbiomes composed of several significantly differentially abundant genera. Whole metagenomic shotgun sequencing of fecal samples identified species-level differences between S. hispidus and S. fulviventer, as well as different metabolic pathway functions as a result of differential host microbiome contributions. Furthermore, the microbiome composition of the external sites showed significant sex-based differences while fecal communities were not largely different. CONCLUSIONS Our study shows that host genetic background potentially exerts homeostatic pressures, resulting in distinct microbiomes for two different inbred cotton rat species. Because of the numerous studies that have uncovered strong relationships between host microbiome, viral infection outcomes, and immune responses, our findings represent a strong contribution for understanding the impact of different microbial communities on viral pathogenesis. Furthermore, we provide novel cotton rat microbiome data as a springboard to uncover the full therapeutic potential of the microbiome against viral infections.
Collapse
Affiliation(s)
- Britton A Strickland
- Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mira C Patel
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
- Present Address: Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Meghan H Shilts
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Helen H Boone
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Arash Kamali
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Zhang
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
| | - Daniel Stylos
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
| | - Marina S Boukhvalova
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
| | | | - Shibu Yooseph
- Department of Computer Science, Genomics and Bioinformatics Cluster, University of Central Florida, Orlando, FL, USA
| | | | - Jorge C G Blanco
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA.
| | - Suman R Das
- Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Division of Infectious Diseases, Vanderbilt University Medical Center, 1211 21st Avenue South, S2108 Medical Center North, Nashville, TN, 37232, USA.
| |
Collapse
|
11
|
Host Components That Modulate the Disease Caused by hMPV. Viruses 2021; 13:v13030519. [PMID: 33809875 PMCID: PMC8004172 DOI: 10.3390/v13030519] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
Human metapneumovirus (hMPV) is one of the main pathogens responsible for acute respiratory infections in children up to 5 years of age, contributing substantially to health burden. The worldwide economic and social impact of this virus is significant and must be addressed. The structural components of hMPV (either proteins or genetic material) can be detected by several receptors expressed by host cells through the engagement of pattern recognition receptors. The recognition of the structural components of hMPV can promote the signaling of the immune response to clear the infection, leading to the activation of several pathways, such as those related to the interferon response. Even so, several intrinsic factors are capable of modulating the immune response or directly inhibiting the replication of hMPV. This article will discuss the current knowledge regarding the innate and adaptive immune response during hMPV infections. Accordingly, the host intrinsic components capable of modulating the immune response and the elements capable of restricting viral replication during hMPV infections will be examined.
Collapse
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
Pilaev M, Shen Y, Carbonneau J, Venable MC, Rhéaume C, Lavigne S, Couture C, Guarné A, Hamelin MÈ, Boivin G. Evaluation of pre- and post-fusion Human metapneumovirus F proteins as subunit vaccine candidates in mice. Vaccine 2020; 38:2122-2127. [DOI: 10.1016/j.vaccine.2020.01.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/11/2019] [Accepted: 01/16/2020] [Indexed: 11/24/2022]
|
17
|
Synergistic PA and HA mutations confer mouse adaptation of a contemporary A/H3N2 influenza virus. Sci Rep 2019; 9:16616. [PMID: 31719554 PMCID: PMC6851088 DOI: 10.1038/s41598-019-51877-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 10/07/2019] [Indexed: 12/25/2022] Open
Abstract
The mouse is the most widely used animal model for influenza virus research. However, the susceptibility of mice to seasonal influenza virus depends on the strain of mouse and on the strain of the influenza virus. Seasonal A/H3N2 influenza viruses do not replicate well in mice and therefore they need to be adapted to this animal model. In this study, we generated a mouse-adapted A/H3N2 virus (A/Switzerland/9715293/2013 [MA-H3N2]) by serial passaging in mouse lungs that exhibited greater virulence compared to the wild-type virus (P0-H3N2). Seven mutations were found in the genome of MA-H3N2: PA(K615E), NP(G384R), NA(G320E) and HA(N122D, N144E, N246K, and A304T). Using reverse genetics, two synergistically acting genes were found as determinants of the pathogenicity in mice. First, the HA substitutions were shown to enhanced viral replication in vitro and, second, the PA-K615E substitution increased polymerase activity, although did not alter virus replication in vitro or in mice. Notably, single mutations had only limited effects on virulence in vitro. In conclusion, a co-contribution of HA and PA mutations resulted in a lethal mouse model of seasonal A/H3N2 virus. Such adapted virus is an excellent tool for evaluation of novel drugs or vaccines and for study of influenza pathogenesis.
Collapse
|
18
|
Dubois J, Pizzorno A, Cavanagh MH, Padey B, Nicolas de Lamballerie C, Uyar O, Venable MC, Carbonneau J, Traversier A, Julien T, Lavigne S, Couture C, Lina B, Hamelin MÈ, Terrier O, Rosa-Calatrava M, Boivin G. Strain-Dependent Impact of G and SH Deletions Provide New Insights for Live-Attenuated HMPV Vaccine Development. Vaccines (Basel) 2019; 7:vaccines7040164. [PMID: 31671656 PMCID: PMC6963613 DOI: 10.3390/vaccines7040164] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/23/2019] [Accepted: 10/23/2019] [Indexed: 01/15/2023] Open
Abstract
Human metapneumovirus (HMPV) is a major pediatric respiratory pathogen with currently no specific treatment or licensed vaccine. Different strategies to prevent this infection have been evaluated, including live-attenuated vaccines (LAV) based on SH and/or G protein deletions. This approach showed promising outcomes but has not been evaluated further using different viral strains. In that regard, we previously showed that different HMPV strains harbor distinct in vitro fusogenic and in vivo pathogenic phenotypes, possibly influencing the selection of vaccine strains. In this study, we investigated the putative contribution of the low conserved SH or G accessory proteins in such strain-dependent phenotypes and generated recombinant wild type (WT) and SH- or G-deleted viruses derived from two different patient-derived HMPV strains, A1/C-85473 and B2/CAN98-75. The ΔSH and ΔG deletions led to different strain-specific phenotypes in both LLC-MK2 cell and reconstituted human airway epithelium models. More interestingly, the ΔG-85473 and especially ΔSH-C-85473 recombinant viruses conferred significant protection against HMPV challenge and induced immunogenicity against a heterologous strain. In conclusion, our results show that the viral genetic backbone should be considered in the design of live-attenuated HMPV vaccines, and that a SH-deleted virus based on the A1/C-85473 HMPV strain could be a promising LAV candidate as it is both attenuated and protective in mice while being efficiently produced in a cell-based system.
Collapse
Affiliation(s)
- Julia Dubois
- Laboratoire de Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France.
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, QC G1V 4G2, Canada.
| | - Andrés Pizzorno
- Laboratoire de Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France.
| | - Marie-Hélène Cavanagh
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, QC G1V 4G2, Canada.
| | - Blandine Padey
- Laboratoire de Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France.
| | - Claire Nicolas de Lamballerie
- Laboratoire de Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France.
| | - Olus Uyar
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, QC G1V 4G2, Canada.
| | - Marie-Christine Venable
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, QC G1V 4G2, Canada.
| | - Julie Carbonneau
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, QC G1V 4G2, Canada.
| | - Aurélien Traversier
- Laboratoire de Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France.
| | - Thomas Julien
- Laboratoire de Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France.
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France.
| | - Sophie Lavigne
- Quebec Heart and Lung Institute, Laval University, Quebec City, QC G1V 4G5, Canada.
| | - Christian Couture
- Quebec Heart and Lung Institute, Laval University, Quebec City, QC G1V 4G5, Canada.
| | - Bruno Lina
- Laboratoire de Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France.
- Laboratoire de Virologie, Centre National de Référence des virus Influenza, Institut des Agents Infectieux, Groupement Hospitalier Nord, Hospices Civils de Lyon, 69004 Lyon, France.
| | - Marie-Ève Hamelin
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, QC G1V 4G2, Canada.
| | - Olivier Terrier
- Laboratoire de Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France.
| | - Manuel Rosa-Calatrava
- Laboratoire de Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France.
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008 Lyon, France.
| | - Guy Boivin
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, QC G1V 4G2, Canada.
| |
Collapse
|
19
|
Human metapneumovirus activates NOD-like receptor protein 3 inflammasome via its small hydrophobic protein which plays a detrimental role during infection in mice. PLoS Pathog 2019; 15:e1007689. [PMID: 30964929 PMCID: PMC6474638 DOI: 10.1371/journal.ppat.1007689] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 04/19/2019] [Accepted: 03/08/2019] [Indexed: 12/30/2022] Open
Abstract
NOD-like receptor protein 3 (NLRP3) inflammasome activation triggers caspase-1 activation-induced maturation of interleukin (IL)-1β and IL-18 and therefore is important for the development of the host defense against various RNA viral diseases. However, the implication of this protein complex in human metapneumovirus (HMPV) disease has not been fully studied. Herein, we report that NLRP3 inflammasome plays a detrimental role during HMPV infection because NLRP3 inflammasome inhibition protected mice from mortality and reduced weight loss and inflammation without impacting viral replication. We also demonstrate that NLRP3 inflammasome exerts its deleterious effect via IL-1β production since we observed reduced mortality, weight loss and inflammation in IL-1β-deficient (IL-1β-/-) mice, as compared to wild-type animals during HMPV infection. Moreover, the effect on these evaluated parameters was not different in IL-1β-/- and wild-type mice treated with an NLRP3 inflammasome inhibitor. The production of IL-1β was also abrogated in bone marrow derived macrophages deficient for NLRP3. Finally, we show that small hydrophobic protein-deleted recombinant HMPV (HMPV ΔSH) failed to activate caspase-1, which is responsible for IL-1β cleavage and maturation. Furthermore, HMPV ΔSH-infected mice had less weight loss, showed no mortality and reduced inflammation, as compared to wild-type HMPV-infected mice. Thus, NLRP3 inflammasome activation seems to be triggered by HMPV SH protein in HMPV disease. In summary, once activated by the HMPV SH protein, NLRP3 inflammasome promotes the maturation of IL-1β, which exacerbates HMPV-induced inflammation. Therefore, the blockade of IL-1β production by using NLRP3 inflammasome inhibitors might be a novel potential strategy for the therapy and prevention of HMPV infection. Human metapneumovirus (HMPV), a negative-stranded, enveloped RNA virus, is recognized as one of the leading causes of acute respiratory disease in children since its discovery in 2001. Nevertheless, there is currently no licensed vaccine for the prevention of HMPV infection and treatment modalities are limited to the use of ribavirin, a weak antiviral agent or immunoglobulins. NOD-like receptor protein 3 (NLRP3) inflammasome has been shown to be involved in the pathogenesis of several RNA viral diseases but its role during HMPV infection has not been fully studied. Here, we report for the first time that NLRP3 inflammasome is activated by the small hydrophobic protein of HMPV, leading to the release of IL-1β, which has the potential to exacerbate inflammation. However, NLRP3 inflammasome has no direct influence on viral replication. Thus, IL-1β-mediated inflammatory process plays an important role during HMPV infection and, therefore, anti-IL-1β strategies such as the use of NLRP3 inhibitors may be a novel potential approach for the prevention and therapy of HMPV disease.
Collapse
|
20
|
Bernal LJ, Velandia-Romero M, Guevara C, Castellanos JE. Human Metapneumovirus: Laboratory Methods for Isolation, Propagation, and Plaque Titration. Intervirology 2019; 61:301-306. [PMID: 30917376 DOI: 10.1159/000497309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 01/24/2019] [Indexed: 11/19/2022] Open
Abstract
The human metapneumovirus (hMPV) is an important viral agent associated with severe infections of the upper and lower airways, especially in young children and immunosuppressed subjects. Nevertheless, in vitro studies of hMPV are very difficult due to the little knowledge we have on its laboratory manipulation. OBJECTIVE The aim of this study was to isolate and propagate hMPV from patients, and to establish a method to quantify the virus by plaque assay. METHOD As part of a Latin American respiratory virus surveillance study, 12 nasal secretion samples - hMPV-positive by direct fluorescence - were inoculated on LLC-MK2 cells to isolate the virus. The supernatants were re-inoculated and the cytopathic effect and syncytium formation were evaluated daily; the infection was confirmed by immunofluorescence and RT-PCR. A protocol to titrate the harvested virus was established inoculating serial dilutions on LLC-MK2 cells, and agarose was then added as an overlay. After different time periods, the monolayers were fixed and stained with Naphthol blue/black or crystal violet and finally the viral titer was obtained. RESULTS Eight out of 12 hMPV-positive respiratory samples were positive for the isolation and confirmed by RT-PCR and immunofluorescence, but the cytopathic effect and syncytium formation were observed only in 5 cultures. One out of 8 viral isolates was used for propagation and plaque assay standardization. We found that incubation for 7 days in the semisolid overlay yielded plaques with appropriate size and shape to be counted, although crystal violet staining showed slightly larger plaques than those seen with Naphthol blue/black staining. CONCLUSIONS The isolation and propagation from patient-derived hMPV and the standardization of a practical, reliable, and inexpensive method of detection and quantification of hMPV were carried out, without the additional use of antibodies that had not been reported previously. These results offer some important insights for future studies of cellular and molecular biology of hMPV.
Collapse
Affiliation(s)
- Lilia J Bernal
- Facultad de Odontología, Universidad Nacional de Colombia, Bogotá, Colombia
| | | | | | - Jaime E Castellanos
- Facultad de Odontología, Universidad Nacional de Colombia, Bogotá, Colombia, .,Grupo de Virología, Universidad El Bosque, Bogotá, Colombia,
| |
Collapse
|
21
|
Han M, Rajput C, Ishikawa T, Jarman CR, Lee J, Hershenson MB. Small Animal Models of Respiratory Viral Infection Related to Asthma. Viruses 2018; 10:E682. [PMID: 30513770 PMCID: PMC6316391 DOI: 10.3390/v10120682] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/21/2018] [Accepted: 11/29/2018] [Indexed: 12/20/2022] Open
Abstract
Respiratory viral infections are strongly associated with asthma exacerbations. Rhinovirus is most frequently-detected pathogen; followed by respiratory syncytial virus; metapneumovirus; parainfluenza virus; enterovirus and coronavirus. In addition; viral infection; in combination with genetics; allergen exposure; microbiome and other pathogens; may play a role in asthma development. In particular; asthma development has been linked to wheezing-associated respiratory viral infections in early life. To understand underlying mechanisms of viral-induced airways disease; investigators have studied respiratory viral infections in small animals. This report reviews animal models of human respiratory viral infection employing mice; rats; guinea pigs; hamsters and ferrets. Investigators have modeled asthma exacerbations by infecting mice with allergic airways disease. Asthma development has been modeled by administration of virus to immature animals. Small animal models of respiratory viral infection will identify cell and molecular targets for the treatment of asthma.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Charu Rajput
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Tomoko Ishikawa
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Caitlin R Jarman
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Julie Lee
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Marc B Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
22
|
Lê BV, Jandrot-Perrus M, Couture C, Checkmahomed L, Venable MC, Hamelin MÈ, Boivin G. Evaluation of anticoagulant agents for the treatment of human metapneumovirus infection in mice. J Gen Virol 2018; 99:1367-1380. [PMID: 30102144 DOI: 10.1099/jgv.0.001135] [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: 12/30/2022] Open
Abstract
Thrombin has been demonstrated to be involved in several viral diseases including human metapneumovirus (hMPV) infections. We previously showed that immediate administration of thrombin inhibitor argatroban post-infection protected mice against hMPV disease. This current work aims at determining whether warfarin and heparin, two other anticoagulants inhibiting thrombin formation and activities, may also be used for treatment against hMPV in vivo. We found that immediate injections of argatroban, warfarin or heparin after virus challenge protected mice against hMPV infection, as evidenced by decreased or no mortality, less weight loss, reduced viral load and attenuated inflammation. However, delayed treatments starting 1 day post-infection with argatroban or warfarin almost did not impact the survival whereas delayed treatment with heparin induced an increased mortality during infection. Moreover, these treatments also did not reduce weight loss, viral replication and inflammation. In agreement with these results, thrombin generation was decreased upon immediate anticoagulant treatments but was unaltered upon delayed treatments. Thus, thrombin generation occurs at the onset of hMPV infection and thrombin inhibition may be only useful for the treatment of this disease when initiated in the early stage. In this case, heparin is not recommended because of its reduced efficacy on mortality in infected mice whereas argatroban and warfarin appear as safe and effective drugs for the treatment of hMPV disease. The antiviral and anti-inflammatory effects of argatroban occur via thrombin-dependent pathways whereas the mechanisms by which warfarin exerts its beneficial effects against hMPV infection were not elucidated and need to be further studied.
Collapse
Affiliation(s)
- Ba Vuong Lê
- 1Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | | | - Christian Couture
- 3Quebec Heart and Lung Institute, Laval University, Quebec City, Quebec, Canada
| | - Liva Checkmahomed
- 1Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | | | - Marie-Ève Hamelin
- 1Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Guy Boivin
- 1Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
23
|
Liu Y, Qin T, Zhao X, Dong S, Zhu J, Peng D, Zhong J, Li T, Chen X. Skewed balance of regulatory T cell and inflammatory T cell in IL-17 defect with human metapneumovirus infection. Cell Immunol 2018; 331:161-167. [PMID: 30077332 DOI: 10.1016/j.cellimm.2018.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/25/2018] [Accepted: 06/18/2018] [Indexed: 01/01/2023]
Abstract
Human metapneumovirus (hMPV) is a common cause of respiratory infections in children. However, the precise mechanisms underlying the development of hMPV-induced pulmonary pathology remain unknown. Studies show that IL-17 plays an important role in some inflammatory diseases of the airways, including asthma and chronic obstructive pulmonary disease. Here, we generated an IL-17 KO murine model of hMPV infection and used it to characterize the role of IL-17 hMPV-induced pulmonary inflammation. The results demonstrated that the defect in IL-17 resulted in less neutrophil influx into the lungs, along with reduced ventilatory function. Meanwhile, viral infection in IL-17 KO mice increased regulatory T cells (Tregs) and reduced Th1 and Th2 cells in the lung, suggesting that lack of IL-17 skews the immune response in the lung toward an anti-inflammatory profile, as exhibited by a greater number of Treg cells and fewer Th1 and Th2 cells.
Collapse
Affiliation(s)
- Yuhang Liu
- Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Tao Qin
- Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xiaodong Zhao
- Research Center for Immunologic and Infectious Diseases, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Division of Immunology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing 400014, China
| | - Shifang Dong
- Division of Flow Cytometry, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Jin Zhu
- Division of Pathology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Donghong Peng
- Division of Respiratory, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Jiarong Zhong
- General Medical Wards, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Tingyu Li
- Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xin Chen
- General Medical Wards, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.
| |
Collapse
|
24
|
Xu J, Zhang Y, Williams JV. Development and optimization of a direct plaque assay for trypsin-dependent human metapneumovirus strains. J Virol Methods 2018; 259:1-9. [PMID: 29807042 DOI: 10.1016/j.jviromet.2018.05.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/24/2018] [Accepted: 05/24/2018] [Indexed: 02/07/2023]
Abstract
Human metapneumovirus (HMPV) is a non-segmented, negative strand RNA virus belonging to the family Pneumoviridae, previously a subfamily of Paramyxoviridae. It is a leading cause of lower respiratory tract infection in infants, children, and adults with underlying medical conditions. HMPV grows poorly in cell culture and requires trypsin to cleave and mature the virus particles, which adds to the challenge of HMPV research. Currently, an indirect immuno-staining assay is commonly used to titrate HMPV, which is time-consuming and costly. In order to simplify virus quantification for HMPV, a direct plaque assay was developed. By optimizing trypsin concentration and other supplements in the agarose overlay, it was found that HMPV strains from all four subgroups formed clear and countable plaques 5-7 days post-infection. Animal tissue homogenate can also be directly titrated with this assay. Compared with the traditional assay, the direct plaque assay yields similar titer result, but saves time and eliminates the use of antibodies. Potentially, it can also be applied to plaque purification for HMPV clinical isolates. The direct plaque assay will be a valuable tool in HMPV research.
Collapse
Affiliation(s)
- Jiuyang Xu
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, 15224, USA; Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing 100084, China
| | - Yu Zhang
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, 15224, USA
| | - John V Williams
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, 15224, USA.
| |
Collapse
|
25
|
Cheemarla NR, Guerrero-Plata A. How does the human metapneumovirus regulate neutrophil infiltration into the airways? Future Virol 2018; 13:233-235. [PMID: 30976289 DOI: 10.2217/fvl-2018-0001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Nagarjuna R Cheemarla
- 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
|
26
|
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
|
27
|
Cheemarla NR, Guerrero-Plata A. Human Metapneumovirus Attachment Protein Contributes to Neutrophil Recruitment into the Airways of Infected Mice. Viruses 2017; 9:v9100310. [PMID: 29065494 PMCID: PMC5691661 DOI: 10.3390/v9100310] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 09/26/2017] [Accepted: 10/21/2017] [Indexed: 12/22/2022] Open
Abstract
Human Metapneumovirus (HMPV) is a leading respiratory pathogen that causes lower respiratory tract infections worldwide. Acute HMPV infection induces an exacerbated inflammatory neutrophilic response leading to bronchiolitis and pneumonia. However, the mechanism by which the virus regulates neutrophil infiltration into the airways still remains unexplored. In this work, we used an experimental mouse model of HMPV infection to demonstrate that the attachment (G) protein of HMPV contributes to the recruitment of neutrophils into the airways and modulate the production of neutrophil chemoattractants and Type I IFN responses, specifically IFN-α. These findings provide the first evidence that the HMPV G protein contributes to the in vivo neutrophilic response to HMPV infection and furthers our understanding on virus induced inflammatory responses in the airways.
Collapse
Affiliation(s)
- Nagarjuna R Cheemarla
- 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
|
28
|
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
|
29
|
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
|
30
|
Ehlen L, Tödtmann J, Specht S, Kallies R, Papies J, Müller MA, Junglen S, Drosten C, Eckerle I. Epithelial cell lines of the cotton rat (Sigmodon hispidus) are highly susceptible in vitro models to zoonotic Bunya-, Rhabdo-, and Flaviviruses. Virol J 2016; 13:74. [PMID: 27142375 PMCID: PMC4855710 DOI: 10.1186/s12985-016-0531-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/24/2016] [Indexed: 01/27/2023] Open
Abstract
Background Small mammals such as bats and rodents have been increasingly recognized as reservoirs of novel potentially zoonotic pathogens. However, few in vitro model systems to date allow assessment of zoonotic viruses in a relevant host context. The cotton rat (Sigmodon hispidus) is a New World rodent species that has a long-standing history as an experimental animal model due to its unique susceptibility to human viruses. Furthermore, wild cotton rats are associated with a large variety of known or potentially zoonotic pathogens. Methods A method for the isolation and culture of airway epithelial cell lines recently developed for bats was applied for the generation of rodent airway and renal epithelial cell lines from the cotton rat. Continuous cell lines were characterized for their epithelial properties as well as for their interferon competence. Susceptibility to members of zoonotic Bunya-, Rhabdo-, and Flaviviridae, in particular Rift Valley fever virus (RVFV), vesicular stomatitis virus (VSV), West Nile virus (WNV), and tick-borne encephalitis virus (TBEV) was tested. Furthermore, novel arthropod-derived viruses belonging to the families Bunya-, Rhabdo-, and Mesoniviridae were tested. Results We successfully established airway and kidney epithelial cell lines from the cotton rat, and characterized their epithelial properties. Cells were shown to be interferon-competent. Viral infection assays showed high-titre viral replication of RVFV, VSV, WNV, and TBEV, as well as production of infectious virus particles. No viral replication was observed for novel arthropod-derived members of the Bunya-, Rhabdo-, and Mesoniviridae families in these cell lines. Conclusion In the current study, we showed that newly established cell lines from the cotton rat can serve as host-specific in vitro models for viral infection experiments. These cell lines may also serve as novel tools for virus isolation, as well as for the investigation of virus-host interactions in a relevant host species.
Collapse
Affiliation(s)
- Lukas Ehlen
- Institute of Virology, University of Bonn Medical Centre, Sigmund-Freud-Strasse 25, 53127, Bonn, Germany
| | - Jan Tödtmann
- Institute of Virology, University of Bonn Medical Centre, Sigmund-Freud-Strasse 25, 53127, Bonn, Germany
| | - Sabine Specht
- Institute for Medical Microbiology, Immunology & Parasitology (IMMIP), University of Bonn Medical Centre, Sigmund-Freud-Strasse 25, 53127, Bonn, Germany.,Present address: Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - René Kallies
- Institute of Virology, University of Bonn Medical Centre, Sigmund-Freud-Strasse 25, 53127, Bonn, Germany.,Present address: Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Jan Papies
- Institute of Virology, University of Bonn Medical Centre, Sigmund-Freud-Strasse 25, 53127, Bonn, Germany
| | - Marcel A Müller
- Institute of Virology, University of Bonn Medical Centre, Sigmund-Freud-Strasse 25, 53127, Bonn, Germany
| | - Sandra Junglen
- Institute of Virology, University of Bonn Medical Centre, Sigmund-Freud-Strasse 25, 53127, Bonn, Germany
| | - Christian Drosten
- Institute of Virology, University of Bonn Medical Centre, Sigmund-Freud-Strasse 25, 53127, Bonn, Germany
| | - Isabella Eckerle
- Institute of Virology, University of Bonn Medical Centre, Sigmund-Freud-Strasse 25, 53127, Bonn, Germany.
| |
Collapse
|
31
|
Foster MW, Gerhardt G, Robitaille L, Plante PL, Boivin G, Corbeil J, Moseley MA. Targeted Proteomics of Human Metapneumovirus in Clinical Samples and Viral Cultures. Anal Chem 2015; 87:10247-54. [PMID: 26376123 DOI: 10.1021/acs.analchem.5b01544] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The rapid, sensitive, and specific identification of infectious pathogens from clinical isolates is a critical need in the hospital setting. Mass spectrometry (MS) has been widely adopted for identification of bacterial pathogens, although polymerase chain reaction remains the mainstay for the identification of viral pathogens. Here, we explored the capability of MS for the detection of human metapneumovirus (HMPV), a common cause of respiratory tract infections in children. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) sequencing of a single HMPV reference strain (CAN97-83) was used to develop a multiple reaction monitoring (MRM) assay that employed stable isotope-labeled peptide internal standards for quantitation of HMPV. Using this assay, we confirmed the presence of HMPV in viral cultures from 10 infected patients and further assigned genetic lineage based on the presence/absence of variant peptides belonging to the viral matrix and nucleoproteins. Similar results were achieved for primary clinical samples (nasopharyngeal aspirates) from the same individuals. As validation, virus lineages, and variant coding sequences, were confirmed by next-generation sequencing of viral RNA obtained from the culture samples. Finally, separate dilution series of HMPV A and B lineages were used to further refine and assess the robustness of the assay and to determine limits of detection in nasopharyngeal aspirates. Our results demonstrate the applicability of MRM for identification of HMPV, and assignment of genetic lineage, from both viral cultures and clinical samples. More generally, this approach should prove tractable as an alternative to nucleic-acid based sequencing for the multiplexed identification of respiratory virus infections.
Collapse
Affiliation(s)
| | - Geoff Gerhardt
- Waters Corporation , Milford, Massachusetts 01757, United States
| | - Lynda Robitaille
- Department of Molecular Medicine, Department of Microbiology, Immunology and Infectious Diseases, Université Laval , Québec City, Québec, Canada G1V 0A6
| | - Pier-Luc Plante
- Department of Molecular Medicine, Department of Microbiology, Immunology and Infectious Diseases, Université Laval , Québec City, Québec, Canada G1V 0A6
| | - Guy Boivin
- Department of Molecular Medicine, Department of Microbiology, Immunology and Infectious Diseases, Université Laval , Québec City, Québec, Canada G1V 0A6
| | - Jacques Corbeil
- Department of Molecular Medicine, Department of Microbiology, Immunology and Infectious Diseases, Université Laval , Québec City, Québec, Canada G1V 0A6
| | | |
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
|
Aerts L, Cavanagh MH, Dubois J, Carbonneau J, Rhéaume C, Lavigne S, Couture C, Hamelin MÈ, Boivin G. Effect of in vitro syncytium formation on the severity of human metapneumovirus disease in a murine model. PLoS One 2015; 10:e0120283. [PMID: 25803584 PMCID: PMC4372586 DOI: 10.1371/journal.pone.0120283] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/05/2015] [Indexed: 12/17/2022] Open
Abstract
Human metapneumovirus (HMPV) is an important cause of acute respiratory tract infections (ARTI) in children, elderly individuals and immunocompromised patients. In vitro, different HMPV strains can induce variable cytopathic effects ranging from large multinucleated syncytia to focal cell rounding. In this study, we investigated the impact of different in vitro phenotypes of two HMPV strains on viral replication and disease severity in a BALB/c mouse model. We first generated two recombinant GFP-expressing HMPV viruses: C-85473, a syncytium-inducing strain (rC-85473) belonging to the A1 subtype and CAN98-75, a focal cell rounding-inducing strain (rCAN98-75) of the B2 subtype. We subsequently exchanged the F genes of both strains to create the chimeric viruses rC-85473_F and rCAN98-75_F. We demonstrated that the F protein was the sole protein responsible for the syncytium phenotype and that viruses carrying a syncytium-inducing F protein replicated to significantly higher titers in vitro. In vivo, however, the virulence and replicative capacity of the different HMPV strains did not appear to be solely dependent on the F gene but also on the viral background, with the strains containing the C-85473 background inducing more weight loss as well as increased lung viral titers, pro-inflammatory cytokines and inflammation than strains containing the CAN98-75 background. In conclusion, the F protein is the main determinant of syncytium formation and replication kinetics in vitro, although it is not the only factor implicated in HMPV disease severity in mice.
Collapse
Affiliation(s)
- Laetitia Aerts
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, Quebec, Canada
| | - Marie-Hélène Cavanagh
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, Quebec, Canada
| | - Julia Dubois
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, Quebec, Canada
| | - Julie Carbonneau
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, Quebec, Canada
| | - Chantal Rhéaume
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, Quebec, Canada
| | - Sophie Lavigne
- Anatomopathologie et cytologie, Institut Universitaire de Cardiologie et de Pneumologie de Québec and Université Laval, Quebec City, QC, Canada
| | - Christian Couture
- Anatomopathologie et cytologie, Institut Universitaire de Cardiologie et de Pneumologie de Québec and Université Laval, Quebec City, QC, Canada
| | - Marie-Ève Hamelin
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, Quebec, Canada
| | - Guy Boivin
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, Quebec, Canada
- * E-mail:
| |
Collapse
|
34
|
Aerts L, Rhéaume C, Carbonneau J, Lavigne S, Couture C, Hamelin MÈ, Boivin G. Adjuvant effect of the human metapneumovirus (HMPV) matrix protein in HMPV subunit vaccines. J Gen Virol 2014; 96:767-774. [PMID: 25519171 DOI: 10.1099/vir.0.000031] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The human metapneumovirus (HMPV) fusion (F) protein is the most immunodominant protein, yet subunit vaccines containing only this protein do not confer complete protection. The HMPV matrix (M) protein induces the maturation of antigen-presenting cells in vitro. The inclusion of the M protein into an F protein subunit vaccine might therefore provide an adjuvant effect. We administered the F protein twice intramuscularly, adjuvanted with alum, the M protein or both, to BALB/c mice at 3 week intervals. Three weeks after the boost, mice were infected with HMPV and monitored for 14 days. At day 5 post-challenge, pulmonary viral titres, histopathology and cytokine levels were analysed. Mice immunized with F+alum and F+M+alum generated significantly more neutralizing antibodies than mice immunized with F only [titres of 47 ± 7 (P<0.01) and 147 ± 13 (P<0.001) versus 17 ± 2]. Unlike F only [1.6 ± 0.5 × 10(3) TCID50 (g lung)(-1)], pulmonary viral titres in mice immunized with F+M and F+M+alum were undetectable. Mice immunized with F+M presented the most important reduction in pulmonary inflammation and the lowest T-helper Th2/Th1 cytokine ratio. In conclusion, addition of the HMPV-M protein to an F protein-based vaccine modulated both humoral and cellular immune responses to subsequent infection, thereby increasing the protection conferred by the vaccine.
Collapse
Affiliation(s)
- Laetitia Aerts
- Centre de Recherche en Infectiologie du Centre Hospitalier Universitaire de Québec and Université Laval, Quebec City, Quebec G1V 4G2, Canada
| | - Chantal Rhéaume
- Centre de Recherche en Infectiologie du Centre Hospitalier Universitaire de Québec and Université Laval, Quebec City, Quebec G1V 4G2, Canada
| | - Julie Carbonneau
- Centre de Recherche en Infectiologie du Centre Hospitalier Universitaire de Québec and Université Laval, Quebec City, Quebec G1V 4G2, Canada
| | - Sophie Lavigne
- Department of Anatomo-pathology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec G1V 4G5, Canada
| | - Christian Couture
- Department of Anatomo-pathology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec G1V 4G5, Canada
| | - Marie-Ève Hamelin
- Centre de Recherche en Infectiologie du Centre Hospitalier Universitaire de Québec and Université Laval, Quebec City, Quebec G1V 4G2, Canada
| | - Guy Boivin
- Centre de Recherche en Infectiologie du Centre Hospitalier Universitaire de Québec and Université Laval, Quebec City, Quebec G1V 4G2, Canada
| |
Collapse
|
35
|
Small Animal Models for Human Metapneumovirus: Cotton Rat is More Permissive than Hamster and Mouse. Pathogens 2014; 3:633-55. [PMID: 25438015 PMCID: PMC4243432 DOI: 10.3390/pathogens3030633] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 06/13/2014] [Accepted: 06/30/2014] [Indexed: 11/17/2022] Open
Abstract
Human metapneumovirus (hMPV) is the second most prevalent causative agent of pediatric respiratory infections worldwide. Currently, there are no vaccines or antiviral drugs against this virus. One of the major hurdles in hMPV research is the difficulty to identify a robust small animal model to accurately evaluate the efficacy and safety of vaccines and therapeutics. In this study, we compared the replication and pathogenesis of hMPV in BALB/c mice, Syrian golden hamsters, and cotton rats. It was found that BALB/c mice are not permissive for hMPV infection despite the use of a high dose (6.5 log10 PFU) of virus for intranasal inoculation. In hamsters, hMPV replicated efficiently in nasal turbinates but demonstrated only limited replication in lungs. In cotton rats, hMPV replicated efficiently in both nasal turbinate and lung when intranasally administered with three different doses (4, 5, and 6 log10 PFU) of hMPV. Lungs of cotton rats infected by hMPV developed interstitial pneumonia with mononuclear cells infiltrates and increased lumen exudation. By immunohistochemistry, viral antigens were detected at the luminal surfaces of the bronchial epithelial cells in lungs. Vaccination of cotton rats with hMPV completely protected upper and lower respiratory tract from wildtype challenge. The immunization also elicited elevated serum neutralizing antibody. Collectively, these results demonstrated that cotton rat is a robust small animal model for hMPV infection.
Collapse
|
36
|
Acute clearance of human metapneumovirus occurs independently of natural killer cells. J Virol 2014; 88:10963-9. [PMID: 24965465 DOI: 10.1128/jvi.01558-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human metapneumovirus (HMPV) is a major cause of respiratory disease. The role of NK cells in protection against HMPV is unclear. We show that while HMPV-infected C57BL/6 mice had higher numbers of functional lung NK cells than mock-treated mice, comparing NK cell-depleted and control mice did not reveal differences in lung viral titers, histopathology, cytokine levels, or T cell numbers or function. These data indicate that NK cells are not required for host control of HMPV.
Collapse
|
37
|
Schuster JE, Cox RG, Hastings AK, Boyd KL, Wadia J, Chen Z, Burton DR, Williamson RA, Williams JV. A broadly neutralizing human monoclonal antibody exhibits in vivo efficacy against both human metapneumovirus and respiratory syncytial virus. J Infect Dis 2014; 211:216-25. [PMID: 24864121 DOI: 10.1093/infdis/jiu307] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Human metapneumovirus (HMPV) is a leading cause of acute respiratory tract infection, with significant morbidity and mortality. No licensed vaccines or therapeutic agents exist. Monoclonal antibodies (mAbs) are effective at preventing other infectious diseases and could be used against HMPV in high-risk hosts. METHODS In vitro assays were performed to assess the neutralizing activity and affinity kinetics of human mAb 54G10. A new mouse model was developed to assess prophylactic and therapeutic efficacy in vivo. The epitope of 54G10 was identified by generating mAb-resistant mutants (MARMs). RESULTS At low concentrations, 54G10 neutralized all 4 subgroups of HMPV in vitro and had subnanomolar affinity for the fusion protein. DBA/2 mice were permissive for all 4 HMPV subgroups, and 54G10 was effective both prophylactically and therapeutically against HMPV in vivo. Sequencing of HMPV MARMs identified the 54G10 epitope, which was similar to an antigenic site on respiratory syncytial virus (RSV). 54G10 also exhibited in vitro neutralizing activity and in vivo protective and therapeutic efficacy against RSV. CONCLUSIONS Human mAb 54G10 has broad neutralizing activity against HMPV and could have prophylactic and therapeutic utility clinically. The conserved epitope could represent a structural vaccine target for HMPV and RSV.
Collapse
Affiliation(s)
| | - Reagan G Cox
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Andrew K Hastings
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Kelli L Boyd
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Jay Wadia
- Crucell Vaccine Institute, San Diego
| | - Zhifeng Chen
- Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California
| | - Dennis R Burton
- Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California
| | | | - John V Williams
- Department of Pediatrics Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
38
|
Wei L, Zhu S, She R, Hu F, Wang J, Yan X, Zhang C, Liu S, Quan R, Li Z, Du F, Wei T, Liu J. Viral replication and lung lesions in BALB/c mice experimentally inoculated with avian metapneumovirus subgroup C isolated from chickens. PLoS One 2014; 9:e92136. [PMID: 24637582 PMCID: PMC3956885 DOI: 10.1371/journal.pone.0092136] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 02/18/2014] [Indexed: 11/18/2022] Open
Abstract
Avian metapneumovirus (aMPV) emerged as an important respiratory pathogen causing acute respiratory tract infection in avian species. Here we used a chicken aMPV subgroup C (aMPV/C) isolate to inoculate experimentally BALB/c mice and found that the aMPV/C can efficiently replicate and persist in the lungs of mice for at least 21 days with a peak viral load at day 6 postinoculation. Lung pathological changes were characterized by increased inflammatory cells. Immunochemical assay showed the presence of viral antigens in the lungs and significant upregulation of pulmonary inflammatory cytokines and chemokines including MCP-1, MIP-1α, RANTES, IL-1β, IFN-γ, and TNF-α were detected following inoculation. These results indicate for the first time that chicken aMPV/C may replicate in the lung of mice. Whether aMPV/C has potential as zoonotic pathogen, further investigation will be required.
Collapse
Affiliation(s)
- Li Wei
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, People’s Republic of China
| | - Shanshan Zhu
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, People’s Republic of China
| | - Ruiping She
- College of Veterinary Medicine, China Agricultural University, Beijing, People’s Republic of China
| | - Fengjiao Hu
- College of Veterinary Medicine, China Agricultural University, Beijing, People’s Republic of China
| | - Jing Wang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, People’s Republic of China
| | - Xu Yan
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, People’s Republic of China
| | - Chunyan Zhang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, People’s Republic of China
| | - Shuhang Liu
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, People’s Republic of China
| | - Rong Quan
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, People’s Republic of China
| | - Zixuan Li
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, People’s Republic of China
| | - Fang Du
- College of Veterinary Medicine, China Agricultural University, Beijing, People’s Republic of China
| | - Ting Wei
- Molecular Virology Laboratory, QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Jue Liu
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, People’s Republic of China
| |
Collapse
|
39
|
Nel HJ, du Plessis N, Kleynhans L, Loxton AG, van Helden PD, Walzl G. Mycobacterium bovis BCG infection severely delays Trichuris muris expulsion and co-infection suppresses immune responsiveness to both pathogens. BMC Microbiol 2014; 14:9. [PMID: 24433309 PMCID: PMC3898725 DOI: 10.1186/1471-2180-14-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 01/10/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The global epidemiology of parasitic helminths and mycobacterial infections display extensive geographical overlap, especially in the rural and urban communities of developing countries. We investigated whether co-infection with the gastrointestinal tract-restricted helminth, Trichuris muris, and the intracellular bacterium, Mycobacterium bovis (M. bovis) BCG, would alter host immune responses to, or the pathological effect of, either infection. RESULTS We demonstrate that both pathogens are capable of negatively affecting local and systemic immune responses towards each other by modifying cytokine phenotypes and by inducing general immune suppression. T. muris infection influenced non-specific and pathogen-specific immunity to M. bovis BCG by down-regulating pulmonary TH1 and Treg responses and inducing systemic TH2 responses. However, co-infection did not alter mycobacterial multiplication or dissemination and host pulmonary histopathology remained unaffected compared to BCG-only infected mice. Interestingly, prior M. bovis BCG infection significantly delayed helminth clearance and increased intestinal crypt cell proliferation in BALB/c mice. This was accompanied by a significant reduction in systemic helminth-specific TH1 and TH2 cytokine responses and significantly reduced local TH1 and TH2 responses in comparison to T. muris-only infected mice. CONCLUSION Our data demonstrate that co-infection with pathogens inducing opposing immune phenotypes, can have differential effects on compartmentalized host immune protection to either pathogen. In spite of local and systemic decreases in TH1 and increases in TH2 responses co-infected mice clear M. bovis BCG at the same rate as BCG only infected animals, whereas prior mycobacterial infection initiates prolonged worm infestation in parallel to decreased pathogen-specific TH2 cytokine production.
Collapse
Affiliation(s)
- Hendrik J Nel
- Division of Molecular Biology and Human Genetics, MRC Centre for Molecular and Cellular Biology, NRF/DST Centre of Excellence in Biomedical TB Research, Faculty Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
- University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Nelita du Plessis
- Division of Molecular Biology and Human Genetics, MRC Centre for Molecular and Cellular Biology, NRF/DST Centre of Excellence in Biomedical TB Research, Faculty Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Leanie Kleynhans
- Division of Molecular Biology and Human Genetics, MRC Centre for Molecular and Cellular Biology, NRF/DST Centre of Excellence in Biomedical TB Research, Faculty Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - André G Loxton
- Division of Molecular Biology and Human Genetics, MRC Centre for Molecular and Cellular Biology, NRF/DST Centre of Excellence in Biomedical TB Research, Faculty Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Paul D van Helden
- Division of Molecular Biology and Human Genetics, MRC Centre for Molecular and Cellular Biology, NRF/DST Centre of Excellence in Biomedical TB Research, Faculty Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Gerhard Walzl
- Division of Molecular Biology and Human Genetics, MRC Centre for Molecular and Cellular Biology, NRF/DST Centre of Excellence in Biomedical TB Research, Faculty Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
40
|
Wei L, Zhu S, Yan X, Wang J, Zhang C, Liu S, She R, Hu F, Quan R, Liu J. Avian metapneumovirus subgroup C infection in chickens, China. Emerg Infect Dis 2014; 19:1092-4. [PMID: 23763901 PMCID: PMC3903454 DOI: 10.3201/eid1907.121126] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Avian metapneumovirus causes acute respiratory tract infection and reductions in egg production in various avian species. We isolated and characterized an increasingly prevalent avian metapneumovirus subgroup C strain from meat-type commercial chickens with severe respiratory signs in China. Culling of infected flocks could lead to economic consequences.
Collapse
Affiliation(s)
- Li Wei
- Beijing Municipal Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Blanco JC, Boukhvalova MS, Perez DR, Vogel SN, Kajon A. Modeling Human Respiratory Viral Infections in the Cotton Rat ( Sigmodon hispidus). ACTA ACUST UNITED AC 2014; 6:40-42. [PMID: 25635205 DOI: 10.4172/jaa.1000093] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
For over three decades, cotton rats have been a preferred model for human Respiratory Syncytial Virus (RSV) infection and pathogenesis, and a reliable model for an impressive list of human respiratory pathogens including adenoviruses, para influenza virus, measles, and human metapneumo virus. The most significant contribution of the cotton rat to biomedical research has been the development of anti-RSV antibodies for prophylactic use in high-risk infants. More recently, however, the cotton rat model has been further explored as a model for infection with other respiratory viral pathogens including influenza and rhinovirus.Together with RSV, these viruses inflict the greatest impact on the human respiratory health.This review will focus on the characteristics of these new models and their potential contribution to the development of new therapies.
Collapse
Affiliation(s)
| | | | - Daniel R Perez
- Department of Veterinary Medicine, University of Maryland, Collage Park, USA
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, USA
| | - Adriana Kajon
- Infectious Disease Program, Lovelace Respiratory Research Institute, Albuquerque, USA
| |
Collapse
|
42
|
Green MG, Huey D, Niewiesk S. The cotton rat (Sigmodon hispidus) as an animal model for respiratory tract infections with human pathogens. Lab Anim (NY) 2013; 42:170-6. [PMID: 23604159 DOI: 10.1038/laban.188] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 10/22/2012] [Indexed: 11/09/2022]
Abstract
Respiratory viral infection is a great human health concern, resulting in disease, death and economic losses. Cotton rats (Sigmodon hispidus) have been particularly useful in the study of the pathogenesis of human respiratory virus infections, including the development and testing of antiviral compounds and vaccines. In this article, the authors outline the advantages of the cotton rat compared with the mouse as a model for infection with measles virus, respiratory syncytial virus, influenza virus, human parainfluenza virus and human metapneumovirus. From the literature and their own experience, the authors summarize guidelines for handling, maintaining and breeding cotton rats. In addition, they offer technical tips for carrying out infection experiments and provide information about the large array of immunological assays and reagents available for the study of immune responses (macrophages, dendritic cells, T cells, B cells, antibodies, chemokines and cytokines) in cotton rats.
Collapse
Affiliation(s)
- M Gia Green
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | | | | |
Collapse
|
43
|
du Plessis N, Kleynhans L, Thiart L, van Helden PD, Brombacher F, Horsnell WGC, Walzl G. Acute helminth infection enhances early macrophage mediated control of mycobacterial infection. Mucosal Immunol 2013; 6:931-41. [PMID: 23250274 DOI: 10.1038/mi.2012.131] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 11/20/2012] [Indexed: 02/04/2023]
Abstract
Co-infection with mycobacteria and helminths is widespread in developing countries, but how this alters host immunological control of each pathogen is not comprehensively understood. In this study, we demonstrate that acute Nippostrongylus brasiliensis (Nb) murine infection reduce early pulmonary mycobacterial colonization. This Nb-associated reduction in pulmonary Mycobacterium tuberculosis colony-forming units was associated with early and increased activation of pulmonary CD4 T cells and increased T helper type 1 (Th1) and Th2 cytokine secretion. An accelerated and transient augmentation of neutrophils and alveolar macrophages (AMs) was also observed in co-infected animals. AMs displayed markers of both classical and alternative activation. Intranasal transfer of pulmonary macrophages obtained from donor mice 5 days after Nb infection significantly reduced pulmonary Mycobacterium bovis Bacille Calmette-Guérin clearance in recipient mice. These data demonstrate that early stage Nb infection elicits a macrophage response, which is protective during the early stages of subsequent mycobacterial infection.
Collapse
Affiliation(s)
- N du Plessis
- Division of Molecular Biology and Human Genetics, MRC Centre for Molecular and Cellular Biology, DST/NRF Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | | | | | | | | | | | | |
Collapse
|
44
|
Aerts L, Hamelin MÈ, Rhéaume C, Lavigne S, Couture C, Kim W, Susan-Resiga D, Prat A, Seidah NG, Vergnolle N, Riteau B, Boivin G. Modulation of protease activated receptor 1 influences human metapneumovirus disease severity in a mouse model. PLoS One 2013; 8:e72529. [PMID: 24015257 PMCID: PMC3755973 DOI: 10.1371/journal.pone.0072529] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 07/10/2013] [Indexed: 11/19/2022] Open
Abstract
Human metapneumovirus (hMPV) infection causes acute respiratory tract infections (RTI) which can result in hospitalization of both children and adults. To date, no antiviral or vaccine is available for this common viral infection. Immunomodulators could represent an interesting strategy for the treatment of severe viral infection. Recently, the role of protease-activated receptors (PAR) in inflammation, coagulation and infection processes has been of growing interest. Herein, the effects of a PAR1 agonist and a PAR1 antagonist on hMPV infection were investigated in BALB/c mice. Intranasal administration of the PAR1 agonist resulted in increased weight loss and mortality of infected mice. Conversely, the PAR1 antagonist was beneficial to hMPV infection by decreasing weight loss and clinical signs and by significantly reducing pulmonary inflammation, pro-inflammatory cytokine levels (including IL-6, KC and MCP-1) and recruitment of immune cells to the lungs. In addition, a significant reduction in pulmonary viral titers was also observed in the lungs of PAR1 antagonist-treated mice. Despite no apparent direct effect on virus replication during in vitro experiments, an important role for PAR1 in the regulation of furin expression in the lungs was shown for the first time. Further experiments indicated that the hMPV fusion protein can be cleaved by furin thus suggesting that PAR1 could have an effect on viral infectivity in addition to its immunomodulatory properties. Thus, inhibition of PAR1 by selected antagonists could represent an interesting strategy for decreasing the severity of paramyxovirus infections.
Collapse
Affiliation(s)
- Laetitia Aerts
- Centre de Recherche en Infectiologie du Centre Hospitalier Universitaire de Québec and Université Laval, Quebec, Canada
| | - Marie-Ève Hamelin
- Centre de Recherche en Infectiologie du Centre Hospitalier Universitaire de Québec and Université Laval, Quebec, Canada
| | - Chantal Rhéaume
- Centre de Recherche en Infectiologie du Centre Hospitalier Universitaire de Québec and Université Laval, Quebec, Canada
| | - Sophie Lavigne
- Department of Anatomo-pathology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec, Canada
| | - Christian Couture
- Department of Anatomo-pathology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec, Canada
| | - WooJin Kim
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal, Montreal, Canada
| | - Delia Susan-Resiga
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal, Montreal, Canada
| | - Annik Prat
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal, Montreal, Canada
| | - Nabil G. Seidah
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal, Montreal, Canada
| | - Nathalie Vergnolle
- Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Université de Toulouse, Université Paul Sabatier, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France
- Department of Physiology and Pharmacology, University of Calgary, Alberta, Canada
| | - Beatrice Riteau
- Virologie et Pathologie Humaine, Université Lyon, Faculté de Médecine RTH Laennec, Lyon, France
- Centre de Tours-Nouzilly Institut National de la Recherche Agronomique, Nouzilly, France
| | - Guy Boivin
- Centre de Recherche en Infectiologie du Centre Hospitalier Universitaire de Québec and Université Laval, Quebec, Canada
| |
Collapse
|
45
|
Lévy C, Aerts L, Hamelin MÈ, Granier C, Szécsi J, Lavillette D, Boivin G, Cosset FL. Virus-like particle vaccine induces cross-protection against human metapneumovirus infections in mice. Vaccine 2013; 31:2778-85. [PMID: 23583815 DOI: 10.1016/j.vaccine.2013.03.051] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 01/17/2013] [Accepted: 03/28/2013] [Indexed: 11/28/2022]
Abstract
Human metapneumovirus (HMPV) is a paramyxovirus that causes acute respiratory-tract infections in children and adults worldwide. A safe and effective vaccine could decrease the burden of disease associated with this novel pathogen. We engineered HMPV viral-like particles (HMPV-VLPs) derived from retroviral core particles that mimic the properties of the viral surface of two HMPV viruses of either lineage A or B. These VLPs functionally display F and G HMPV surface glycoproteins. When injected in mice, HMPV-VLPs induce strong humoral immune response against both homologous and heterologous strains. Moreover, the induced neutralizing antibodies prevented mortality upon subsequent infection of the lungs with both homologous and heterologous viruses. Upon challenge, viral titers in the lungs of immunized animals were significantly reduced as compared to those of control animals. In conclusion, a HMPV-VLP vaccine that induces cross-protective immunity in mice is a promising approach to prevent HMPV infections.
Collapse
Affiliation(s)
- Camille Lévy
- CIRI, International Center for Infectiology Research, EVIR Team, Université de Lyon, Lyon, France
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Edwards KM, Zhu Y, Griffin MR, Weinberg GA, Hall CB, Szilagyi PG, Staat MA, Iwane M, Prill MM, Williams JV. Burden of human metapneumovirus infection in young children. N Engl J Med 2013; 368:633-43. [PMID: 23406028 PMCID: PMC3662802 DOI: 10.1056/nejmoa1204630] [Citation(s) in RCA: 230] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND The inpatient and outpatient burden of human metapneumovirus (HMPV) infection among young children has not been well established. METHODS We conducted prospective, population-based surveillance for acute respiratory illness or fever among inpatient and outpatient children less than 5 years of age in three U.S. counties from 2003 through 2009. Clinical and demographic data were obtained from parents and medical records, HMPV was detected by means of a reverse-transcriptase polymerase-chain-reaction assay, and population-based rates of hospitalization and estimated rates of outpatient visits associated with HMPV infection were determined. RESULTS HMPV was detected in 200 of 3490 hospitalized children (6%), 222 of 3257 children in outpatient clinics (7%), 224 of 3001 children in the emergency department (7%), and 10 of 770 asymptomatic controls (1%). Overall annual rates of hospitalization associated with HMPV infection were 1 per 1000 children less than 5 years of age, 3 per 1000 infants less than 6 months of age, and 2 per 1000 children 6 to 11 months of age. Children hospitalized with HMPV infection, as compared with those hospitalized without HMPV infection, were older and more likely to receive a diagnosis of pneumonia or asthma, to require supplemental oxygen, and to have a longer stay in the intensive care unit. The estimated annual burden of outpatient visits associated with HMPV infection was 55 clinic visits and 13 emergency department visits per 1000 children. The majority of HMPV-positive inpatient and outpatient children had no underlying medical conditions, although premature birth and asthma were more frequent among hospitalized children with HMPV infection than among those without HMPV infection. CONCLUSIONS HMPV infection is associated with a substantial burden of hospitalizations and outpatient visits among children throughout the first 5 years of life, especially during the first year. Most children with HMPV infection were previously healthy. (Funded by the Centers for Disease Control and Prevention and the National Institutes of Health.).
Collapse
Affiliation(s)
- Kathryn M Edwards
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232-2581, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Haas LEM, Thijsen SFT, van Elden L, Heemstra KA. Human metapneumovirus in adults. Viruses 2013; 5:87-110. [PMID: 23299785 PMCID: PMC3564111 DOI: 10.3390/v5010087] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 12/17/2012] [Accepted: 12/19/2012] [Indexed: 11/16/2022] Open
Abstract
Human metapneumovirus (HMPV) is a relative newly described virus. It was first isolated in 2001 and currently appears to be one of the most significant and common human viral infections. Retrospective serologic studies demonstrated the presence of HMPV antibodies in humans more than 50 years earlier. Although the virus was primarily known as causative agent of respiratory tract infections in children, HMPV is an important cause of respiratory infections in adults as well. Almost all children are infected by HMPV below the age of five; the repeated infections throughout life indicate transient immunity. HMPV infections usually are mild and self-limiting, but in the frail elderly and the immunocompromised patients, the clinical course can be complicated. Since culturing the virus is relatively difficult, diagnosis is mostly based on a nucleic acid amplification test, such as reverse transcriptase polymerase chain reaction. To date, no vaccine is available and treatment is supportive. However, ongoing research shows encouraging results. The aim of this paper is to review the current literature concerning HMPV infections in adults, and discuss recent development in treatment and vaccination.
Collapse
Affiliation(s)
- Lenneke E. M. Haas
- Department of Intensive Care Medicine, Diakonessenhuis, Utrecht, 3582 KE, The Netherlands
| | - Steven F. T. Thijsen
- Department of Microbiology, Diakonessenhuis, Utrecht, 3582 KE, The Netherlands; E-Mails: (S.F.T.T.); (K.A.H.)
| | - Leontine van Elden
- Department of Pulmonary Diseases, Diakonessenhuis, Utrecht, 3582 KE, The Netherlands; E-Mail:
| | - Karen A. Heemstra
- Department of Microbiology, Diakonessenhuis, Utrecht, 3582 KE, The Netherlands; E-Mails: (S.F.T.T.); (K.A.H.)
| |
Collapse
|
48
|
Abstract
It has been 10 years since human metapneumovirus (HMPV) was identified as a causative agent of respiratory illness in humans. Since then, numerous studies have contributed to a substantial body of knowledge on many aspects of HMPV. This review summarizes our current knowledge on HMPV, HMPV disease pathogenesis, and disease intervention strategies and identifies a number of areas with key questions to be addressed in the future.
Collapse
|
49
|
Kwon D, Shin K, Shin JY, Lee JY, Ha Y, Lee NJ, Oh HB, Chae C, Kang C. Pathogenesis and Chronologic Localization of the Human Influenza A (H1N1) Virus in Cotton Rats. Osong Public Health Res Perspect 2011; 2:15-22. [PMID: 24159445 PMCID: PMC3766908 DOI: 10.1016/j.phrp.2011.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 03/14/2011] [Accepted: 03/29/2011] [Indexed: 01/20/2023] Open
Abstract
Objectives We aimed to evaluate the pathogenesis and chronologic localization of human influenza A (H1N1) virus in experimentally infected cotton rats. Methods The animals were intranasally inoculated with 107 plaque-forming units of A/Solomon Islands/3/2006 (H1N1) influenza virus and evaluated for pathogenicity for a period of 28 days. Virus replication kinetics and pathological properties were assessed chronologically. Acute antiviral responses were evaluated by mean of real-time polymerase chain reaction. Results Cotton rats infected with A/Solomon Islands/3/2006 virus lost weight until 6 days post-inoculation (DPI) and showed decreased activity until 3 DPI. At necropsy, focal areas of redness and consolidation of lungs were evident at 1, 2, and 3 DPI. Lung histopathology showed moderate to severe interstitial pneumonia, alveolitis and bronchiolitis. Influenza A specific viral protein was detected in bronchiolar epithelial cells, alveolar septa and pneumocytes. Influenza viruses were recovered from the lungs during the early period of infection and the titer peaked at 1 DPI. Viral proteins were detected from 4 hours to 6 hours DPI. These trends correlate with the up-regulation of mRNA expression of the IFN-α, Mx1, and Mx2 genes that play critical roles in the anti-influenza response at the early stage of infection. Conclusion Our results provide evidence that supports the use of cotton rats for the study of influenza virus pathogenesis and the immune response.
Collapse
Affiliation(s)
- Donghyok Kwon
- Division of Influenza Virus, Center for Infectious Diseases, Korea National Institute of Health, Osong, Korea
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Kyeongcheol Shin
- Division of Influenza Virus, Center for Infectious Diseases, Korea National Institute of Health, Osong, Korea
| | - Jin-Young Shin
- Division of Influenza Virus, Center for Infectious Diseases, Korea National Institute of Health, Osong, Korea
| | - Joo-Yeon Lee
- Division of Influenza Virus, Center for Infectious Diseases, Korea National Institute of Health, Osong, Korea
| | - Yooncheol Ha
- Division of Influenza Virus, Center for Infectious Diseases, Korea National Institute of Health, Osong, Korea
| | - Nam-Joo Lee
- Division of Influenza Virus, Center for Infectious Diseases, Korea National Institute of Health, Osong, Korea
| | - Hee-Bok Oh
- Division of Influenza Virus, Center for Infectious Diseases, Korea National Institute of Health, Osong, Korea
| | - Chanhee Chae
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
- Corresponding author.
| | - Chun Kang
- Division of Influenza Virus, Center for Infectious Diseases, Korea National Institute of Health, Osong, Korea
- Corresponding author.
| |
Collapse
|
50
|
Ludewick HP, Aerts L, Hamelin ME, Boivin G. Long-term impairment of Streptococcus pneumoniae lung clearance is observed after initial infection with influenza A virus but not human metapneumovirus in mice. J Gen Virol 2011; 92:1662-1665. [PMID: 21411678 DOI: 10.1099/vir.0.030825-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human metapneumovirus (hMPV) is a paramyxovirus responsible for respiratory tract infections in humans. Our objective was to investigate whether hMPV could predispose to long-term bacterial susceptibility, such as previously observed with influenza viruses. BALB/c mice were infected with hMPV or influenza A and, 14 days following viral infection, challenged with Streptococcus pneumoniae. Only mice previously infected with influenza A demonstrated an 8% weight loss of their body weight 72 h following S. pneumoniae infection, which correlated with an enhanced lung bacterial replication of >7 log(10) compared with pneumococcus infection alone. This enhanced bacterial replication was not related to altered macrophage or neutrophil recruitment or deficient production of critical cytokines. However, bacterial challenge induced the production of gamma interferon in bronchoalveolar lavages of influenza-infected mice, but not in those of hMPV-infected animals. In conclusion, hMPV does not cause long-term impairment of pneumococcus lung clearance, in contrast to influenza A virus.
Collapse
Affiliation(s)
- Herbert P Ludewick
- Research Center in Infectious Diseases of the 'Centre Hospitalier Universitaire de Québec' and Laval University, Québec City, QC, G1V 4G2, Canada
| | - Laetitia Aerts
- Research Center in Infectious Diseases of the 'Centre Hospitalier Universitaire de Québec' and Laval University, Québec City, QC, G1V 4G2, Canada
| | - Marie-Eve Hamelin
- Research Center in Infectious Diseases of the 'Centre Hospitalier Universitaire de Québec' and Laval University, Québec City, QC, G1V 4G2, Canada
| | - Guy Boivin
- Research Center in Infectious Diseases of the 'Centre Hospitalier Universitaire de Québec' and Laval University, Québec City, QC, G1V 4G2, Canada
| |
Collapse
|