1
|
Girma A. Biology of human respiratory syncytial virus: Current perspectives in immune response and mechanisms against the virus. Virus Res 2024; 350:199483. [PMID: 39396572 PMCID: PMC11513633 DOI: 10.1016/j.virusres.2024.199483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/30/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Human respiratory syncytial virus (hRSV) remains a leading cause of morbidity and mortality in infants, young children, and older adults. hRSV infection's limited treatment and vaccine options significantly increase bronchiolitis' morbidity rates. The severity and outcome of viral infection hinge on the innate immune response. Developing vaccines and identifying therapeutic interventions suitable for young children, older adults, and pregnant women relies on comprehending the molecular mechanisms of viral PAMP recognition, genetic factors of the inflammatory response, and antiviral defense. This review covers fundamental elements of hRSV biology, diagnosis, pathogenesis, and the immune response, highlighting prospective options for vaccine development.
Collapse
Affiliation(s)
- Abayeneh Girma
- Department of Biology, College of Natural and Computational Sciences, Mekdela Amba University, P.O. Box 32, Tulu Awuliya, Ethiopia.
| |
Collapse
|
2
|
Zhao C, Bai Y, Wang W, Amonkar GM, Mou H, Olejnik J, Hume AJ, Mühlberger E, Lukacs NW, Fearns R, Lerou PH, Ai X. Activation of STAT3-mediated ciliated cell survival protects against severe infection by respiratory syncytial virus. J Clin Invest 2024; 134:e183978. [PMID: 39484716 PMCID: PMC11527452 DOI: 10.1172/jci183978] [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: 06/17/2024] [Accepted: 08/15/2024] [Indexed: 11/03/2024] Open
Abstract
Respiratory syncytial virus (RSV) selectively targets ciliated cells in human bronchial epithelium and can cause bronchiolitis and pneumonia, mostly in infants. To identify molecular targets of intervention during RSV infection in infants, we investigated how age regulates RSV interaction with the bronchial epithelium barrier. Employing precision-cut lung slices and air-liquid interface cultures generated from infant and adult human donors, we found robust RSV virus spread and extensive apoptotic cell death only in infant bronchial epithelium. In contrast, adult bronchial epithelium showed no barrier damage and limited RSV infection. Single nuclear RNA-Seq revealed age-related insufficiency of an antiapoptotic STAT3 activation response to RSV infection in infant ciliated cells, which was exploited to facilitate virus spread via the extruded apoptotic ciliated cells carrying RSV. Activation of STAT3 and blockade of apoptosis rendered protection against severe RSV infection in infant bronchial epithelium. Lastly, apoptotic inhibitor treatment of a neonatal mouse model of RSV infection mitigated infection and inflammation in the lung. Taken together, our findings identify a STAT3-mediated antiapoptosis pathway as a target to battle severe RSV disease in infants.
Collapse
Affiliation(s)
- Caiqi Zhao
- Division of Newborn Medicine, Department of Pediatrics and
| | - Yan Bai
- Division of Newborn Medicine, Department of Pediatrics and
| | - Wei Wang
- Division of Newborn Medicine, Department of Pediatrics and
| | | | - Hongmei Mou
- The Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, Massachusetts, USA
| | - Judith Olejnik
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Adam J. Hume
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Elke Mühlberger
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Nicholas W. Lukacs
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Rachel Fearns
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Paul H. Lerou
- Division of Newborn Medicine, Department of Pediatrics and
| | - Xingbin Ai
- Division of Newborn Medicine, Department of Pediatrics and
| |
Collapse
|
3
|
Gay ACA, Banchero M, Carpaij O, Kole TM, Apperloo L, van Gosliga D, Fajar PA, Koppelman GH, Bont L, Hendriks RW, van den Berge M, Nawijn MC. Airway epithelial cell response to RSV is mostly impaired in goblet and multiciliated cells in asthma. Thorax 2024; 79:811-821. [PMID: 38373824 PMCID: PMC11347251 DOI: 10.1136/thorax-2023-220230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 11/27/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND In patients with asthma, respiratory syncytial virus (RSV) infections can cause disease exacerbation by infecting the epithelial layer of the airways, inducing subsequent immune response. The type I interferon antiviral response of epithelial cells upon RSV infection is found to be reduced in asthma in most-but not all-studies. Moreover, the molecular mechanisms causing the differences in the asthmatic bronchial epithelium in response to viral infection are poorly understood. METHODS Here, we investigated the transcriptional response to RSV infection of primary bronchial epithelial cells (pBECs) from patients with asthma (n=8) and healthy donors (n=8). The pBECs obtained from bronchial brushes were differentiated in air-liquid interface conditions and infected with RSV. After 3 days, cells were processed for single-cell RNA sequencing. RESULTS A strong antiviral response to RSV was observed for all cell types, for all samples (p<1e-48). Most (1045) differentially regulated genes following RSV infection were found in cells transitioning to secretory cells. Goblet cells from patients with asthma showed lower expression of genes involved in the interferon response (false discovery rate <0.05), including OASL, ICAM1 and TNFAIP3. In multiciliated cells, an impairment of the signalling pathways involved in the response to RSV in asthma was observed. CONCLUSION Our results highlight that the response to RSV infection of the bronchial epithelium in asthma and healthy airways was largely similar. However, in asthma, the response of goblet and multiciliated cells is impaired, highlighting the need for studying airway epithelial cells at high resolution in the context of asthma exacerbation.
Collapse
Affiliation(s)
- Aurore C A Gay
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| | - Martin Banchero
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| | - Orestes Carpaij
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Tessa M Kole
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Leonie Apperloo
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| | - Djoke van Gosliga
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Putri Ayu Fajar
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| | - Gerard H Koppelman
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Louis Bont
- Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
- Division of Infectious Diseases, Department of Pediatrics, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Maarten van den Berge
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Martijn C Nawijn
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
4
|
Felicetti T, Sarnari C, Gaito R, Tabarrini O, Manfroni G. Recent Progress toward the Discovery of Small Molecules as Novel Anti-Respiratory Syncytial Virus Agents. J Med Chem 2024; 67:11543-11579. [PMID: 38970494 DOI: 10.1021/acs.jmedchem.4c00630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2024]
Abstract
Respiratory syncytial virus (RSV) stands as the foremost cause of infant hospitalization globally, ranking second only to malaria in terms of infant mortality. Although three vaccines have recently been approved for the prophylaxis of adults aged 60 and above, and pregnant women, there is currently no effective antiviral drug for treating RSV infections. The only preventive measure for infants at high risk of severe RSV disease is passive immunization through monoclonal antibodies. This Perspective offers an overview of the latest advancements in RSV drug discovery of small molecule antivirals, with particular focus on the promising findings from agents targeting the fusion and polymerase proteins. A comprehensive reflection on the current state of RSV research is also given, drawing inspiration from the lessons gleaned from HCV and HIV, while also considering the impact of the recent approval of the three vaccines.
Collapse
Affiliation(s)
- Tommaso Felicetti
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Chiara Sarnari
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Roberta Gaito
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Giuseppe Manfroni
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| |
Collapse
|
5
|
Zhao C, Wang W, Bai Y, Amonkar G, Mou H, Olejnik J, Hume AJ, Mühlberger E, Fang Y, Que J, Fearns R, Ai X, Lerou PH. Age-related STAT3 signaling regulates severity of respiratory syncytial viral infection in human bronchial epithelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.20.558606. [PMID: 37781574 PMCID: PMC10541147 DOI: 10.1101/2023.09.20.558606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Respiratory syncytial virus (RSV) can cause severe disease especially in infants; however, mechanisms of age-associated disease severity remain elusive. Here, employing human bronchial epithelium models generated from tracheal aspirate-derived basal stem cells of neonates and adults, we investigated whether age regulates RSV-epithelium interaction to determine disease severity. We show that following RSV infection, only neonatal epithelium model exhibited cytopathy and mucus hyperplasia, and neonatal epithelium had more robust viral spread and inflammatory responses than adult epithelium. Mechanistically, RSV-infected neonatal ciliated cells displayed age-related impairment of STAT3 activation, rendering susceptibility to apoptosis, which facilitated viral spread. In contrast, SARS-CoV-2 infection of ciliated cells had no effect on STAT3 activation and was not affected by age. Taken together, our findings identify an age-related and RSV-specific interaction with neonatal bronchial epithelium that critically contributes to severity of infection, and STAT3 activation offers a potential strategy to battle severe RSV disease in infants.
Collapse
|
6
|
Duan W, Cen Y, Lin C, Ouyang H, Du K, Kumar A, Wang B, Avolio J, Grasemann H, Moraes TJ. Inflammatory epithelial cytokines after in vitro respiratory syncytial viral infection are associated with reduced lung function. ERJ Open Res 2021; 7:00365-2021. [PMID: 34527729 PMCID: PMC8435810 DOI: 10.1183/23120541.00365-2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 06/11/2021] [Indexed: 11/27/2022] Open
Abstract
Respiratory syncytial virus (RSV) infections in early life predispose children with cystic fibrosis (CF) to more severe lung function decline in later life. The mechanisms explaining the associations between RSV and progression of CF lung disease are not clear. In this study, a human bronchial epithelial cell line and primary human nasal epithelial cells (PNECs) from individuals with CF and healthy control donors were infected with RSV. Real-time PCR, plaque assay, cytokine detection, immunofluorescence and Western blot analyses were performed. RSV is replicated to a higher degree in CF epithelial cells as compared to control cells; however, no defects in innate immune pathways were identified in CF cells. Rather, primary p.Phe508del cystic fibrosis transmembrane conductance regulator PNECs produced more cytokines after RSV infection than control cells. Moreover, interleukin-8 and tumour necrosis factor-α production post RSV negatively correlated with lung function (% predicted forced expiratory volume in 1 s) in the individuals who donated the cells. These data suggest that CF epithelium has a dysfunctional response to RSV allowing for enhanced viral replication and an exaggerated inflammatory response that ultimately may predispose to greater airway inflammation and reduced lung function. This work demonstrates an association between epithelial inflammatory cytokines after in vitro viral infection and lung function in cystic fibrosis, and reinforces the importance of studying innate immune epithelial cell function in cystic fibrosishttps://bit.ly/3gDNwwo
Collapse
Affiliation(s)
- Wenming Duan
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Yuchen Cen
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada.,Dept of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Cindy Lin
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Hong Ouyang
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Kai Du
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Anushree Kumar
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Borui Wang
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Julie Avolio
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Hartmut Grasemann
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada.,Division of Respiratory Medicine, Dept of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
| | - Theo J Moraes
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada.,Dept of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Division of Respiratory Medicine, Dept of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
7
|
A Fragile Balance: Does Neutrophil Extracellular Trap Formation Drive Pulmonary Disease Progression? Cells 2021; 10:cells10081932. [PMID: 34440701 PMCID: PMC8394734 DOI: 10.3390/cells10081932] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Neutrophils act as the first line of defense during infection and inflammation. Once activated, they are able to fulfil numerous tasks to fight inflammatory insults while keeping a balanced immune response. Besides well-known functions, such as phagocytosis and degranulation, neutrophils are also able to release "neutrophil extracellular traps" (NETs). In response to most stimuli, the neutrophils release decondensed chromatin in a NADPH oxidase-dependent manner decorated with histones and granule proteins, such as neutrophil elastase, myeloperoxidase, and cathelicidins. Although primarily supposed to prevent microbial dissemination and fight infections, there is increasing evidence that an overwhelming NET response correlates with poor outcome in many diseases. Lung-related diseases especially, such as bacterial pneumonia, cystic fibrosis, chronic obstructive pulmonary disease, aspergillosis, influenza, and COVID-19, are often affected by massive NET formation. Highly vascularized areas as in the lung are susceptible to immunothrombotic events promoted by chromatin fibers. Keeping this fragile equilibrium seems to be the key for an appropriate immune response. Therapies targeting dysregulated NET formation might positively influence many disease progressions. This review highlights recent findings on the pathophysiological influence of NET formation in different bacterial, viral, and non-infectious lung diseases and summarizes medical treatment strategies.
Collapse
|
8
|
Contribution of Pro-Inflammatory Molecules Induced by Respiratory Virus Infections to Neurological Disorders. Pharmaceuticals (Basel) 2021; 14:ph14040340. [PMID: 33917837 PMCID: PMC8068239 DOI: 10.3390/ph14040340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 12/19/2022] Open
Abstract
Neurobehavioral alterations and cognitive impairment are common phenomena that represent neuropsychiatric disorders and can be triggered by an exacerbated immune response against pathogens, brain injury, or autoimmune diseases. Pro-inflammatory molecules, such as cytokines and chemokines, are produced in the brain by resident cells, mainly by microglia and astrocytes. Brain infiltrating immune cells constitutes another source of these molecules, contributing to an impaired neurological synapse function, affecting typical neurobehavioral and cognitive performance. Currently, there is increasing evidence supporting the notion that behavioral alterations and cognitive impairment can be associated with respiratory viral infections, such as human respiratory syncytial virus, influenza, and SARS-COV-2, which are responsible for endemic, epidemic, or pandemic outbreak mainly in the winter season. This article will review the brain′s pro-inflammatory response due to infection by three highly contagious respiratory viruses that are the leading cause of acute respiratory illness, morbidity, and mobility in infants, immunocompromised and elderly population. How these respiratory viral pathogens induce increased secretion of pro-inflammatory molecules and their relationship with the alterations at a behavioral and cognitive level will be discussed.
Collapse
|
9
|
González-García LD, Martínez-Castillo M, Vargas-Pavía TA, Ulloa-Aguilar JM, Arévalo-Romero H, Léon-Reyes G, Helguera-Repetto AC, García-Cordero J, León-Juárez M. Inhibition of AMP-activated protein kinase in respiratory syncytial virus infection activates lipid metabolism. Arch Virol 2021; 166:1177-1182. [PMID: 33580381 DOI: 10.1007/s00705-021-04974-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 12/10/2020] [Indexed: 11/25/2022]
Abstract
Respiratory syncytial virus (RSV) is most commonly associated with upper respiratory tract infections during childhood. The lipid composition of cells and lipogenic enzymes play an important role in RSV infection. There are controversial data about whether lipid biosynthesis regulators such as AMP-activated protein kinase (AMPK) are deregulated by RSV. Hence, we examined whether the activation state of AMPK is altered in RSV-infected HEp-2 cells. Our data show that RSV infection inhibits AMPK activity, favoring the activation of downstream lipogenic effectors and cellular lipid anabolism in HEp-2 cells.
Collapse
Affiliation(s)
- Luis Didier González-García
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Montes Urales 800, Col. Lomas Virreyes, CP 11000, Ciudad de México, México
| | - Macario Martínez-Castillo
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Casco de Santo Tomas, 11340, Ciudad de México, México
| | - Tania Allin Vargas-Pavía
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Montes Urales 800, Col. Lomas Virreyes, CP 11000, Ciudad de México, México
| | - José Manuel Ulloa-Aguilar
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Montes Urales 800, Col. Lomas Virreyes, CP 11000, Ciudad de México, México
| | - Haruki Arévalo-Romero
- Laboratorio de Inmunología y Microbiología Molecular, División Académica multidisciplinaria de Jalpa de Méndez, Departamento de Genómica, Universidad Juárez Autónoma de Tabasco, Jalpa de Méndez, México
| | - Guadalupe Léon-Reyes
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica, Ciudad de México, México
| | - Addy Cecilia Helguera-Repetto
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Montes Urales 800, Col. Lomas Virreyes, CP 11000, Ciudad de México, México
| | - Julio García-Cordero
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del I.P.N., Ciudad de México, México
| | - Moisés León-Juárez
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Montes Urales 800, Col. Lomas Virreyes, CP 11000, Ciudad de México, México.
| |
Collapse
|
10
|
Bianchini S, Silvestri E, Argentiero A, Fainardi V, Pisi G, Esposito S. Role of Respiratory Syncytial Virus in Pediatric Pneumonia. Microorganisms 2020; 8:microorganisms8122048. [PMID: 33371276 PMCID: PMC7766387 DOI: 10.3390/microorganisms8122048] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022] Open
Abstract
Respiratory viral infections represent the leading cause of hospitalization in infants and young children worldwide and the second leading cause of infant mortality. Among these, Respiratory Syncytial Virus (RSV) represents the main cause of lower respiratory tract infections (LRTIs) in young children worldwide. RSV manifestation can range widely from mild upper respiratory infections to severe respiratory infections, mainly bronchiolitis and pneumonia, leading to hospitalization, serious complications (such as respiratory failure), and relevant sequalae in childhood and adulthood (wheezing, asthma, and hyperreactive airways). There are no specific clinical signs or symptoms that can distinguish RSV infection from other respiratory pathogens. New multiplex platforms offer the possibility to simultaneously identify different pathogens, including RSV, with an accuracy similar to that of single polymerase chain reaction (PCR) in the majority of cases. At present, the treatment of RSV infection relies on supportive therapy, mainly consisting of oxygen and hydration. Palivizumab is the only prophylactic method available for RSV infection. Advances in technology and scientific knowledge have led to the creation of different kinds of vaccines and drugs to treat RSV infection. Despite the good level of these studies, there are currently few registered strategies to prevent or treat RSV due to difficulties related to the unpredictable nature of the disease and to the specific target population.
Collapse
Affiliation(s)
- Sonia Bianchini
- Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy; (S.B.); (E.S.)
- Pediatric Unit, ASST Santi Carlo e Paolo, 20142 Milan, Italy
| | - Ettore Silvestri
- Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy; (S.B.); (E.S.)
| | - Alberto Argentiero
- Pediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (A.A.); (V.F.); (G.P.)
| | - Valentina Fainardi
- Pediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (A.A.); (V.F.); (G.P.)
| | - Giovanna Pisi
- Pediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (A.A.); (V.F.); (G.P.)
| | - Susanna Esposito
- Pediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (A.A.); (V.F.); (G.P.)
- Correspondence: ; Tel.: +39-0521-704790
| |
Collapse
|
11
|
The diverse roles of RIP kinases in host-pathogen interactions. Semin Cell Dev Biol 2020; 109:125-143. [PMID: 32859501 PMCID: PMC7448748 DOI: 10.1016/j.semcdb.2020.08.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/09/2020] [Accepted: 08/09/2020] [Indexed: 12/16/2022]
Abstract
Receptor Interacting Protein Kinases (RIPKs) are cellular signaling molecules that are critical for homeostatic signaling in both communicable and non-communicable disease processes. In particular, RIPK1, RIPK2, RIPK3 and RIPK7 have emerged as key mediators of intracellular signal transduction including inflammation, autophagy and programmed cell death, and are thus essential for the early control of many diverse pathogenic organisms. In this review, we discuss the role of each RIPK in host responses to bacterial and viral pathogens, with a focus on studies that have used pathogen infection models rather than artificial stimulation with purified pathogen associated molecular patterns. We also discuss the intricate mechanisms of host evasion by pathogens that specifically target RIPKs for inactivation, and finally, we will touch on the controversial issue of drug development for kinase inhibitors to treat chronic inflammatory and neurological disorders, and the implications this may have on the outcome of pathogen infections.
Collapse
|
12
|
Hu M, Bogoyevitch MA, Jans DA. Impact of Respiratory Syncytial Virus Infection on Host Functions: Implications for Antiviral Strategies. Physiol Rev 2020; 100:1527-1594. [PMID: 32216549 DOI: 10.1152/physrev.00030.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Respiratory syncytial virus (RSV) is one of the leading causes of viral respiratory tract infection in infants, the elderly, and the immunocompromised worldwide, causing more deaths each year than influenza. Years of research into RSV since its discovery over 60 yr ago have elucidated detailed mechanisms of the host-pathogen interface. RSV infection elicits widespread transcriptomic and proteomic changes, which both mediate the host innate and adaptive immune responses to infection, and reflect RSV's ability to circumvent the host stress responses, including stress granule formation, endoplasmic reticulum stress, oxidative stress, and programmed cell death. The combination of these events can severely impact on human lungs, resulting in airway remodeling and pathophysiology. The RSV membrane envelope glycoproteins (fusion F and attachment G), matrix (M) and nonstructural (NS) 1 and 2 proteins play key roles in modulating host cell functions to promote the infectious cycle. This review presents a comprehensive overview of how RSV impacts the host response to infection and how detailed knowledge of the mechanisms thereof can inform the development of new approaches to develop RSV vaccines and therapeutics.
Collapse
Affiliation(s)
- MengJie Hu
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - Marie A Bogoyevitch
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - David A Jans
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
13
|
Wu W, Choi EJ, Lee I, Lee YS, Bao X. Non-Coding RNAs and Their Role in Respiratory Syncytial Virus (RSV) and Human Metapneumovirus (hMPV) Infections. Viruses 2020; 12:v12030345. [PMID: 32245206 PMCID: PMC7150941 DOI: 10.3390/v12030345] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/14/2020] [Accepted: 03/18/2020] [Indexed: 12/17/2022] Open
Abstract
Recent high-throughput sequencing revealed that only 2% of the transcribed human genome codes for proteins, while the majority of transcriptional products are non-coding RNAs (ncRNAs). Herein, we review the current knowledge regarding ncRNAs, both host- and virus-derived, and their role in respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) infections. RSV is known as the most common cause of lower respiratory tract infection (LRTI) in children, while hMPV is also a significant contributor to LRTI in the pediatrics population. Although RSV and hMPV are close members, belonging to the Pneumoviridae family, they induce distinct changes in the ncRNA profile. Several types of host ncRNAs, including long ncRNA (lncRNA), microRNAs (miRNAs), and transfer RNA (tRNA)-derived RNA fragments (tRFs), are involved as playing roles in RSV and/or hMPV infection. Given the importance of ncRNAs in regulating the expression and functions of genes and proteins, comprehensively understanding the roles of ncRNAs in RSV/hMPV infection could shed light upon the disease mechanisms of RSV and hMPV, potentially providing insights into the development of prevention strategies and antiviral therapy. The presence of viral-derived RNAs and the potential of using ncRNAs as diagnostic biomarkers are also discussed in this review.
Collapse
Affiliation(s)
- Wenzhe Wu
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (W.W.); (E.-J.C.)
| | - Eun-Jin Choi
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (W.W.); (E.-J.C.)
| | | | - Yong Sun Lee
- Department of Cancer System Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si Gyeonggi-do 10408, Korea;
| | - Xiaoyong Bao
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (W.W.); (E.-J.C.)
- Sealy Center for Molecular Medicine, The University of Texas Medical Branch, Galveston, TX 77555, USA
- The Institute of Translational Sciences, The University of Texas Medical Branch, Galveston, TX 77555, USA
- The Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, TX 77555, USA
- Correspondence: ; Tel.: +409-772-1777
| |
Collapse
|
14
|
Sastre B, García-García ML, Calvo C, Casas I, Rodrigo-Muñoz JM, Cañas JA, Mora I, del Pozo V. Immune recovery following bronchiolitis is linked to a drop in cytokine and LTC4 levels. Pediatr Res 2020; 87:581-587. [PMID: 31600771 PMCID: PMC7086521 DOI: 10.1038/s41390-019-0606-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/25/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Bronchiolitis is the main cause of hospitalization of children younger than 1 year; however, the immune mechanism of bronchiolitis is not completely understood. The aim of this study was to analyze the recovery of immune response after a bronchiolitis episode. METHODS Forty-nine infants hospitalized with bronchiolitis diagnosis were enrolled. Nasopharyngeal aspirates (NPAs) were processed. Twenty-seven pro-inflammatory biomarkers linked to innate immunity, inflammation, and epithelial damage, as well as nitrites and lipid mediators, were evaluated in the NPA supernatant by ELISA (enzyme-linked immunosorbent assay) and Luminex. Also, 11 genes were analyzed in NPA cells by quantitative PCR. RESULTS A widespread statistically significant decline of multiple pro-inflammatory parameters and cytokines were detected in the recovery period after respiratory infection: interferon-α2 (IFNα2), IFNγ, interleukin-10 (IL-10), IL-1β, IL-8, IFN-γ-inducible protein-10, vascular endothelial growth factor, monocyte chemoattractant protein-1, macrophage inflammatory protein-1α (MIP-1α), and MIP-1β. Supporting these results, a decreased nuclear factor-κB gene expression was observed (P = 0.0116). A significant diminution of cysteinyl leukotriene C4 (LTC4) soluble levels (P = 0.0319) and cyclooxygenase-2 (COX-2) gene expression were observed in the recovery sample. In children classified by post-bronchiolitis wheezing, LTC4 remains elevated in the NPA supernatant. CONCLUSIONS After bronchiolitis, cytokines and biomarkers linked to innate immune response in NPA decrease significantly in the recovery period accompanied by a drop in LTC4 levels; however, this reduction was lower in infants with post-bronchiolitis wheezing.
Collapse
Affiliation(s)
- Beatriz Sastre
- grid.419651.eDepartment of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain ,0000 0000 9314 1427grid.413448.eCIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - María Luz García-García
- 0000 0001 0635 4617grid.411361.0Pediatrics Department, Severo Ochoa Hospital, Leganés, Spain ,Translational Research Network in Pediatric Infectious Diseases (RITIP), Madrid, Spain ,0000 0001 2323 8386grid.464699.0Alfonso X El Sabio University, Madrid, Spain
| | - Cristina Calvo
- Translational Research Network in Pediatric Infectious Diseases (RITIP), Madrid, Spain ,0000 0001 2323 8386grid.464699.0Alfonso X El Sabio University, Madrid, Spain ,0000 0000 8970 9163grid.81821.32Pediatric Infectious Diseases Department, Hospital Universitario La Paz, Madrid, Spain ,Fundación IdiPaz, Madrid, Spain ,TEDDY Network (European Network of Excellence for Pediatric Clinical Research), Madrid, Spain
| | - Inmaculada Casas
- Translational Research Network in Pediatric Infectious Diseases (RITIP), Madrid, Spain ,Respiratory Virus and Influenza Unit, National Microbiology Center (ISCIII), Madrid, Spain
| | - José Manuel Rodrigo-Muñoz
- grid.419651.eDepartment of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain ,0000 0000 9314 1427grid.413448.eCIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - José Antonio Cañas
- grid.419651.eDepartment of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain ,0000 0000 9314 1427grid.413448.eCIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Inés Mora
- grid.419651.eDepartment of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Victoria del Pozo
- grid.419651.eDepartment of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain ,0000 0000 9314 1427grid.413448.eCIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|
15
|
Juliana A, Zonneveld R, Plötz FB, van Meurs M, Wilschut J. Neutrophil-endothelial interactions in respiratory syncytial virus bronchiolitis: An understudied aspect with a potential for prediction of severity of disease. J Clin Virol 2019; 123:104258. [PMID: 31931445 DOI: 10.1016/j.jcv.2019.104258] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/25/2019] [Accepted: 12/30/2019] [Indexed: 01/25/2023]
Abstract
Respiratory syncytial virus (RSV) lower respiratory tract infection (LRTI) causes significant morbidity and mortality among young infants worldwide. It is currently widely accepted that neutrophil influx into the airways is a hallmark of the pathophysiology. However, the exact mechanism of neutrophil migration from the vasculature into the alveolar space in RSV LRTI has received little attention. Data shows that endothelial cells become activated upon RSV infection, driving a 'pro-adhesive state' for circulating neutrophils with upregulation of endothelial intercellular adhesion molecule-1 (ICAM-1). During RSV LRTI different subsets of immature and mature neutrophils are present in the bloodstream, that upregulate integrins lymphocyte-function associated antigen (LFA)-1 and macrophage (Mac)-1, serving as ICAM-1 ligands. An alveolar gradient of interleukin-8 may serve as a potent chemoattractant for circulating neutrophils. Neutrophils from lung aspirates of RSV-infected infants show further signs of inflammatory and migratory activation, while soluble endothelial cell adhesion molecules (sCAMs), such as sICAM-1, have become measurable in the systemic circulation. Whether these mechanisms are solely responsible for neutrophil migration into the alveolar space remains under debate. However, data indicate that the currently postulated neutrophil influx into the lungs should rather be regarded as a neutrophil efflux from the vasculature, involving substantial neutrophil-endothelial interactions. Molecular patterns of these interactions may be clinically useful to predict outcomes of RSV LRTI and deserve further study.
Collapse
Affiliation(s)
- Amadu Juliana
- Academic Pediatric Center Suriname, Academic Hospital Paramaribo, Paramaribo, Suriname.
| | - Rens Zonneveld
- Department of Microbiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Frans B Plötz
- Department of Pediatrics, Tergooi Hospitals, Blaricum, The Netherlands
| | - Matijs van Meurs
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan Wilschut
- Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
16
|
Carvajal JJ, Avellaneda AM, Salazar-Ardiles C, Maya JE, Kalergis AM, Lay MK. Host Components Contributing to Respiratory Syncytial Virus Pathogenesis. Front Immunol 2019; 10:2152. [PMID: 31572372 PMCID: PMC6753334 DOI: 10.3389/fimmu.2019.02152] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the most prevalent viral etiological agent of acute respiratory tract infection. Although RSV affects people of all ages, the disease is more severe in infants and causes significant morbidity and hospitalization in young children and in the elderly. Host factors, including an immature immune system in infants, low lymphocyte levels in patients under 5 years old, and low levels of RSV-specific neutralizing antibodies in the blood of adults over 65 years of age, can explain the high susceptibility to RSV infection in these populations. Other host factors that correlate with severe RSV disease include high concentrations of proinflammatory cytokines such as interleukins (IL)-6, IL-8, tumor necrosis factor (TNF)-α, and thymic stromal lymphopoitein (TSLP), which are produced in the respiratory tract of RSV-infected individuals, accompanied by a strong neutrophil response. In addition, data from studies of RSV infections in humans and in animal models revealed that this virus suppresses adaptive immune responses that could eliminate it from the respiratory tract. Here, we examine host factors that contribute to RSV pathogenesis based on an exhaustive review of in vitro infection in humans and in animal models to provide insights into the design of vaccines and therapeutic tools that could prevent diseases caused by RSV.
Collapse
Affiliation(s)
- Jonatan J. Carvajal
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Andrea M. Avellaneda
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Camila Salazar-Ardiles
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Jorge E. Maya
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K. Lay
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad de Chile, Santiago, Chile
| |
Collapse
|
17
|
San-Juan-Vergara H, Peeples ME. Importance of Virus Characteristics in Respiratory Syncytial Virus-Induced Disease. Immunol Allergy Clin North Am 2019; 39:321-334. [PMID: 31284923 PMCID: PMC6879194 DOI: 10.1016/j.iac.2019.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Severe lower respiratory tract infection in infants and young children is most frequently caused by respiratory syncytial virus (RSV). RSV infects the smallest airways, making breathing difficult and in some infants requiring medical support. Severity is affected by viral dose, infant age, virus genotype, and effectiveness of the innate/adaptive immune responses. Severe disease correlates with later wheezing and asthma in some children. The adaptive immune response is protective but wanes after each infection, likely due to the ability of the RSV NS1/NS2 proteins to inhibit the innate immune response. Several vaccine approaches and candidates are currently in clinical trials.
Collapse
Affiliation(s)
- Homero San-Juan-Vergara
- Division of Health Sciences, Fundación Universidad del Norte, Universidad del Norte, Bloque de Salud, Cuarto Piso 4-25L4, Km 5. Via Puerto, Barranquilla 081007, Colombia
| | - Mark E Peeples
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
18
|
Norris MJ, Malhi M, Duan W, Ouyang H, Granados A, Cen Y, Tseng YC, Gubbay J, Maynes J, Moraes TJ. Targeting Intracellular Ion Homeostasis for the Control of Respiratory Syncytial Virus. Am J Respir Cell Mol Biol 2019; 59:733-744. [PMID: 30095982 DOI: 10.1165/rcmb.2017-0345oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of mortality in infants and young children. Despite the RSV disease burden, no vaccine is available, and treatment remains nonspecific. New drug candidates are needed to combat RSV. Toward this goal, we screened over 2,000 compounds to identify approved drugs with novel anti-RSV activity. Cardiac glycosides, inhibitors of the membrane-bound Na+/K+-ATPase, were identified to have anti-RSV activity. Cardiac glycosides diminished RSV infection in human epithelial type 2 cells and in primary human airway epithelial cells grown at an air-liquid interface. Digoxin, a U.S. Food and Drug Administration-approved cardiac glycoside, was also able to inhibit infection of primary nasal epithelial cells with community isolates of RSV. Our results suggest that the antiviral effects of cardiac glycosides may be dependent on changes in the intracellular Na+ and K+ composition. Consistent with this mechanism, we demonstrated that the ionophoric antibiotics salinomycin, valinomycin, and monensin inhibited RSV in human epithelial type 2 cells and primary nasal epithelial cells. Our data indicate that the K+/Na+-sensitive steps in the RSV life cycle occur within the initial 4 hours of viral infection but do not include virus binding/entry. Rather, our findings demonstrated a negative effect on the RSV transcription and/or replication process. Overall, this work suggests that targeting intracellular ion concentrations offers a novel antiviral strategy.
Collapse
Affiliation(s)
- Michael J Norris
- 1 Department of Laboratory Medicine and Pathobiology and.,2 Program in Translational Medicine
| | - Manpreet Malhi
- 3 Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.,4 Program in Molecular Medicine
| | | | | | - Andrea Granados
- 1 Department of Laboratory Medicine and Pathobiology and.,5 Public Health Ontario, Toronto, Ontario, Canada
| | | | | | | | - Jason Maynes
- 4 Program in Molecular Medicine.,6 Department of Anesthesia and Pain Medicine, and
| | - Theo J Moraes
- 1 Department of Laboratory Medicine and Pathobiology and.,2 Program in Translational Medicine.,7 Division of Respiratory Medicine, Department of Pediatrics, Hospital for Sick Children, Toronto, Ontario, Canada; and
| |
Collapse
|
19
|
Tognarelli EI, Bueno SM, González PA. Immune-Modulation by the Human Respiratory Syncytial Virus: Focus on Dendritic Cells. Front Immunol 2019; 10:810. [PMID: 31057543 PMCID: PMC6478035 DOI: 10.3389/fimmu.2019.00810] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/26/2019] [Indexed: 12/23/2022] Open
Abstract
The human respiratory syncytial virus (hRSV) is the leading cause of pneumonia in infants and produces a significant burden in the elderly. It can also infect and produce disease in otherwise healthy adults and recurrently infect those previously exposed to the virus. Importantly, recurrent infections are not necessarily a consequence of antigenic variability, as described for other respiratory viruses, but most likely due to the capacity of this virus to interfere with the host's immune response and the establishment of a protective and long-lasting immunity. Although some genes encoded by hRSV are known to have a direct participation in immune evasion, it seems that repeated infection is mainly given by its capacity to modulate immune components in such a way to promote non-optimal antiviral responses in the host. Importantly, hRSV is known to interfere with dendritic cell (DC) function, which are key cells involved in establishing and regulating protective virus-specific immunity. Notably, hRSV infects DCs, alters their maturation, migration to lymph nodes and their capacity to activate virus-specific T cells, which likely impacts the host antiviral response against this virus. Here, we review and discuss the most important and recent findings related to DC modulation by hRSV, which might be at the basis of recurrent infections in previously infected individuals and hRSV-induced disease. A focus on the interaction between DCs and hRSV will likely contribute to the development of effective prophylactic and antiviral strategies against this virus.
Collapse
Affiliation(s)
- Eduardo I Tognarelli
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
20
|
Kim MJ, Shim DH, Cha H, Moon K, Yang CM, Hwang SJ, Kim KW, Park JH, Lee C, Elias JA, Sohn MH, Lee JM. Chitinase 3-like 1 protein plays a critical role in respiratory syncytial virus-induced airway inflammation. Allergy 2019; 74:685-697. [PMID: 30402955 PMCID: PMC7159489 DOI: 10.1111/all.13661] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 09/09/2018] [Accepted: 09/19/2018] [Indexed: 12/28/2022]
Abstract
Background Chitinase 3‐like 1 protein (CHI3L1) (YKL‐40 in humans and breast regression protein [BRP]‐39 in mice) is required for optimal allergen sensitization and Th2 inflammation in various chronic inflammatory diseases including asthma. However, the role of CHI3L1 in airway inflammation induced by respiratory viruses has not been investigated. The aim of this study was to investigate the relationship between CHI3L1 and airway inflammation caused by respiratory syncytial virus (RSV) infection. Methods We measured YKL‐40 levels in human nasopharyngeal aspirate (NPA) from hospitalized children presenting with acute respiratory symptoms. Wild‐type (WT) and BRP‐39 knockout (KO) C57BL/6 mice were inoculated with live RSV (A2 strain). Bronchoalveolar lavage fluid and lung tissue samples were obtained on day 7 after inoculation to assess lung inflammation, airway reactivity, and expression of cytokines and BRP‐39. Results In human subjects, YKL‐40 and IL‐13 levels in NPA were higher in children with RSV infection than in control subjects. Expression of BRP‐39 and Th2 cytokines, IL‐13 in particular, was increased following RSV infection in mice. Airway inflammation caused by RSV infection was reduced in BRP‐39 KO mice as compared to WT mice. Th2 cytokine levels were not increased in the lungs of RSV‐infected BRP‐39 KO mice. BRP‐39 regulated M2 macrophage activation in RSV‐infected mice. Additionally, treatment with anti‐CHI3L1 antibody attenuated airway inflammation and Th2 cytokine production in RSV‐infected WT mice. Conclusion These findings suggest that CHI3L1 could contribute to airway inflammation induced by RSV infection. CHI3L1 could be a potential therapeutic candidate for attenuating Th2‐associated immunopathology during RSV infection.
Collapse
Affiliation(s)
- Min Jung Kim
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
- Department of Pediatrics Severance Hospital Institute of Allergy Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Doo Hee Shim
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Hye‐Ran Cha
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Kuk‐Young Moon
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Chang Mo Yang
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Su Jin Hwang
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Kyung Won Kim
- Department of Pediatrics Severance Hospital Institute of Allergy Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Jeon Han Park
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology Brown University Providence Rhode Island USA
| | - Jack A. Elias
- Department of Molecular Microbiology and Immunology Brown University Providence Rhode Island USA
| | - Myung Hyun Sohn
- Department of Pediatrics Severance Hospital Institute of Allergy Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Jae Myun Lee
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| |
Collapse
|
21
|
Muraro SP, De Souza GF, Gallo SW, Da Silva BK, De Oliveira SD, Vinolo MAR, Saraiva EM, Porto BN. Respiratory Syncytial Virus induces the classical ROS-dependent NETosis through PAD-4 and necroptosis pathways activation. Sci Rep 2018; 8:14166. [PMID: 30242250 PMCID: PMC6154957 DOI: 10.1038/s41598-018-32576-y] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/11/2018] [Indexed: 12/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of diseases of the respiratory tract in young children and babies, being mainly associated with bronchiolitis. RSV infection occurs primarily in pulmonary epithelial cells and, once infection is established, an immune response is triggered and neutrophils are recruited. In this study, we investigated the mechanisms underlying NET production induced by RSV. We show that RSV induced the classical ROS-dependent NETosis in human neutrophils and that RSV was trapped in DNA lattices coated with NE and MPO. NETosis induction by RSV was dependent on signaling by PI3K/AKT, ERK and p38 MAPK and required histone citrullination by PAD-4. In addition, RIPK1, RIPK3 and MLKL were essential to RSV-induced NETosis. MLKL was also necessary to neutrophil necrosis triggered by the virus, likely promoting membrane-disrupting pores, leading to neutrophil lysis and NET extrusion. Finally, we found that RSV infection of alveolar epithelial cells or lung fibroblasts triggers NET-DNA release by neutrophils, indicating that neutrophils can identify RSV-infected cells and respond to them by releasing NETs. The identification of the mechanisms responsible to mediate RSV-induced NETosis may prove valuable to the design of new therapeutic approaches to treat the inflammatory consequences of RSV bronchiolitis in young children.
Collapse
Affiliation(s)
- Stéfanie P Muraro
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, 90610-000, Brazil
| | - Gabriela F De Souza
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, 90610-000, Brazil
| | - Stephanie W Gallo
- Laboratory of Immunology and Microbiology, School of Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, 90610-000, Brazil
| | - Bruna K Da Silva
- Laboratory of Immunoinflammation, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083-862, Brazil
| | - Sílvia D De Oliveira
- Laboratory of Immunology and Microbiology, School of Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, 90610-000, Brazil
| | - Marco Aurélio R Vinolo
- Laboratory of Immunoinflammation, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083-862, Brazil
| | - Elvira M Saraiva
- Laboratory of Immunobiology of Leishmaniasis, Department of Immunology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, 21941-902, Brazil
| | - Bárbara N Porto
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, 90610-000, Brazil.
| |
Collapse
|
22
|
Differential Responses by Human Respiratory Epithelial Cell Lines to Respiratory Syncytial Virus Reflect Distinct Patterns of Infection Control. J Virol 2018; 92:JVI.02202-17. [PMID: 29769339 DOI: 10.1128/jvi.02202-17] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/02/2018] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) infects small foci of respiratory epithelial cells via infected droplets. Infection induces expression of type I and III interferons (IFNs) and proinflammatory cytokines, the balance of which may restrict viral replication and affect disease severity. We explored this balance by infecting two respiratory epithelial cell lines with low doses of recombinant RSV expressing green fluorescent protein (rgRSV). A549 cells were highly permissive, whereas BEAS-2B cells restricted infection to individual cells or small foci. After infection, A549 cells expressed higher levels of IFN-β-, IFN-λ-, and NF-κB-inducible proinflammatory cytokines. In contrast, BEAS-2B cells expressed higher levels of antiviral interferon-stimulated genes, pattern recognition receptors, and other signaling intermediaries constitutively and after infection. Transcriptome analysis revealed that constitutive expression of antiviral and proinflammatory genes predicted responses by each cell line. These two cell lines provide a model for elucidating critical mediators of local control of viral infection in respiratory epithelial cells.IMPORTANCE Airway epithelium is both the primary target of and the first defense against respiratory syncytial virus (RSV). Whether RSV replicates and spreads to adjacent epithelial cells depends on the quality of their innate immune responses. A549 and BEAS-2B are alveolar and bronchial epithelial cell lines, respectively, that are often used to study RSV infection. We show that A549 cells are permissive to RSV infection and express genes characteristic of a proinflammatory response. In contrast, BEAS-2B cells restrict infection and express genes characteristic of an antiviral response associated with expression of type I and III interferons. Transcriptome analysis of constitutive gene expression revealed patterns that may predict the response of each cell line to infection. This study suggests that restrictive and permissive cell lines may provide a model for identifying critical mediators of local control of infection and stresses the importance of the constitutive antiviral state for the response to viral challenge.
Collapse
|
23
|
Engystol reduces onset of experimental respiratory syncytial virus-induced respiratory inflammation in mice by modulating macrophage phagocytic capacity. PLoS One 2018; 13:e0195822. [PMID: 29672626 PMCID: PMC5909611 DOI: 10.1371/journal.pone.0195822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 03/30/2018] [Indexed: 01/21/2023] Open
Abstract
Background Respiratory viruses such as respiratory syncytial virus (RSV) or rhinovirus are one of the major causes for respiratory tract infections causing common cold disease. Respiratory viral infections range from mild symptoms in adults to serious illness especially in the very young or elderly as well as patients suffering from lung diseases or being immunocompromised due to other reasons. Engystol (EGY-2) is a multicomponent, multitarget preparation consisting of Vincetoxicum hirundinaria and Sulfur in various dilutions. The study objective was to test the effect of EGY-2 on the innate immune response during the early onset of respiratory viral infection in vivo as exemplified in a mouse model of RSV-induced respiratory inflammation. Methods Naïve BALB/c mice were infected with 1x106 infectious units RSV A2 intranasally to cause a mild respiratory infection. EGY-2 was administered daily per oral gavage starting seven days prior to RSV infection at doses of 0.4 to 5.1 tablets/kg. Control groups received placebo treatment. Animals were sacrificed 1 to 3 days post infection (p.i.) to analyse the infection and induced immune response in the lung. Viral load in bronchoalveolar lavage fluid (BALF) and lung homogenate was determined by TCID50 assay as well as immunofluorescence staining of BALF cells using anti-RSV antibody and microscopic analysis. The RSV induced immune response was assessed by evaluation of BALF differential cell count, BALF cytokine secretion and analysis of the phagocytic capacity of alveolar macrophages. Results EGY-2 significantly reduced the RSV induced neutrophil and early lymphocyte influx on day 1 p.i. in BALF. EGY-2 treatment significantly diminished the RSV induced secretion of pro-inflammatory cytokines such as IFN-γ, IL-1β, IL-6, KC and TNF-α at day 1. EGY-2 treatment was not protective for RSV infection per se, as no alteration in the viral load in lung and BALF was detected. Enhanced numbers of phagocytic-active macrophages were observed in EGY-2 treated animals on day 1 and this macrophage population showed strongly enhanced phagocytic activity on day 1 and day 3. Conclusion The data suggest a beneficial immunomodulatory effect of EGY-2 during early onset of respiratory viral infection in vivo, mediated by stimulation of macrophage phagocytosis, resulting in a reduced innate inflammatory response in terms of neutrophil and early lymphocyte infiltration as well as reduced inflammatory cytokine secretion.
Collapse
|
24
|
Gonzàlez-Parra G, De Ridder F, Huntjens D, Roymans D, Ispas G, Dobrovolny HM. A comparison of RSV and influenza in vitro kinetic parameters reveals differences in infecting time. PLoS One 2018; 13:e0192645. [PMID: 29420667 PMCID: PMC5805318 DOI: 10.1371/journal.pone.0192645] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 01/26/2018] [Indexed: 11/19/2022] Open
Abstract
Influenza and respiratory syncytial virus (RSV) cause acute infections of the respiratory tract. Since the viruses both cause illnesses with similar symptoms, researchers often try to apply knowledge gleaned from study of one virus to the other virus. This can be an effective and efficient strategy for understanding viral dynamics or developing treatment strategies, but only if we have a full understanding of the similarities and differences between the two viruses. This study used mathematical modeling to quantitatively compare the viral kinetics of in vitro RSV and influenza virus infections. Specifically, we determined the viral kinetics parameters for RSV A2 and three strains of influenza virus, A/WSN/33 (H1N1), A/Puerto Rico/8/1934 (H1N1), and pandemic H1N1 influenza virus. We found that RSV viral titer increases at a slower rate and reaches its peak value later than influenza virus. Our analysis indicated that the slower increase of RSV viral titer is caused by slower spreading of the virus from one cell to another. These results provide estimates of dynamical differences between influenza virus and RSV and help provide insight into the virus-host interactions that cause observed differences in the time courses of the two illnesses in patients.
Collapse
Affiliation(s)
- Gilberto Gonzàlez-Parra
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States of America
- Department of Mathematics, New Mexico Tech, Socorro, NM, United States of America
| | | | | | | | | | - Hana M. Dobrovolny
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States of America
- * E-mail:
| |
Collapse
|
25
|
Pickens JA, Tripp RA. Verdinexor Targeting of CRM1 is a Promising Therapeutic Approach against RSV and Influenza Viruses. Viruses 2018; 10:E48. [PMID: 29361733 PMCID: PMC5795461 DOI: 10.3390/v10010048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 12/11/2022] Open
Abstract
Two primary causes of respiratory tract infections are respiratory syncytial virus (RSV) and influenza viruses, both of which remain major public health concerns. There are a limited number of antiviral drugs available for the treatment of RSV and influenza, each having limited effectiveness and each driving selective pressure for the emergence of drug-resistant viruses. Novel broad-spectrum antivirals are needed to circumvent problems with current disease intervention strategies, while improving the cytokine-induced immunopathology associated with RSV and influenza infections. In this review, we examine the use of Verdinexor (KPT-335, a novel orally bioavailable drug that functions as a selective inhibitor of nuclear export, SINE), as an antiviral with multifaceted therapeutic potential. KPT-335 works to (1) block CRM1 (i.e., Chromosome Region Maintenance 1; exportin 1 or XPO1) mediated export of viral proteins critical for RSV and influenza pathogenesis; and (2) repress nuclear factor κB (NF-κB) activation, thus reducing cytokine production and eliminating virus-associated immunopathology. The repurposing of SINE compounds as antivirals shows promise not only against RSV and influenza virus but also against other viruses that exploit the nucleus as part of their viral life cycle.
Collapse
Affiliation(s)
- Jennifer A Pickens
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA.
| | - Ralph A Tripp
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
26
|
Molina IG, Josts I, Almeida Hernandez Y, Esperante S, Salgueiro M, Garcia Alai MM, de Prat-Gay G, Tidow H. Structure and stability of the Human respiratory syncytial virus M 2-1 RNA-binding core domain reveals a compact and cooperative folding unit. Acta Crystallogr F Struct Biol Commun 2018; 74:23-30. [PMID: 29372904 PMCID: PMC5947689 DOI: 10.1107/s2053230x17017381] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/04/2017] [Indexed: 11/10/2022] Open
Abstract
Human syncytial respiratory virus is a nonsegmented negative-strand RNA virus with serious implications for respiratory disease in infants, and has recently been reclassified into a new family, Pneumoviridae. One of the main reasons for this classification is the unique presence of a transcriptional antiterminator, called M2-1. The puzzling mechanism of action of M2-1, which is a rarity among antiterminators in viruses and is part of the RNA polymerase complex, relies on dissecting the structure and function of this multidomain tetramer. The RNA-binding activity is located in a monomeric globular `core' domain, a high-resolution crystal structure of which is now presented. The structure reveals a compact domain which is superimposable on the full-length M2-1 tetramer, with additional electron density for the C-terminal tail that was not observed in the previous models. Moreover, its folding stability was determined through chemical denaturation, which shows that the secondary and tertiary structure unfold concomitantly, which is indicative of a two-state equilibrium. These results constitute a further step in the understanding of this unique RNA-binding domain, for which there is no sequence or structural counterpart outside this virus family, in addition to its implications in transcription regulation and its likeliness as an antiviral target.
Collapse
Affiliation(s)
- Ivana G. Molina
- Protein Structure–Function and Engineering Laboratory, Fundación Instituto Leloir and IIBBA–CONICET, Avenida Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
| | - Inokentijs Josts
- The Hamburg Centre for Ultrafast Imaging and Department of Chemistry, Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Yasser Almeida Hernandez
- The Hamburg Centre for Ultrafast Imaging and Department of Chemistry, Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Sebastian Esperante
- Protein Structure–Function and Engineering Laboratory, Fundación Instituto Leloir and IIBBA–CONICET, Avenida Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
| | - Mariano Salgueiro
- Protein Structure–Function and Engineering Laboratory, Fundación Instituto Leloir and IIBBA–CONICET, Avenida Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
| | - Maria M. Garcia Alai
- European Molecular Biology Laboratory (EMBL), Hamburg Unit, Notkestrasse 85, 22607 Hamburg, Germany
| | - Gonzalo de Prat-Gay
- Protein Structure–Function and Engineering Laboratory, Fundación Instituto Leloir and IIBBA–CONICET, Avenida Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
| | - Henning Tidow
- The Hamburg Centre for Ultrafast Imaging and Department of Chemistry, Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| |
Collapse
|
27
|
New Insights Contributing to the Development of Effective Vaccines and Therapies to Reduce the Pathology Caused by hRSV. Int J Mol Sci 2017; 18:ijms18081753. [PMID: 28800119 PMCID: PMC5578143 DOI: 10.3390/ijms18081753] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 07/28/2017] [Accepted: 08/07/2017] [Indexed: 12/12/2022] Open
Abstract
Human Respiratory Syncytial Virus (hRSV) is one of the major causes of acute lower respiratory tract infections (ALRTI) worldwide, leading to significant levels of immunocompromisation as well as morbidity and mortality in infants. Its main target of infection is the ciliated epithelium of the lungs and the host immune responses elicited is ineffective at achieving viral clearance. It is thought that the lack of effective immunity against hRSV is due in part to the activity of several viral proteins that modulate the host immune response, enhancing a Th2-like pro-inflammatory state, with the secretion of cytokines that promote the infiltration of immune cells to the lungs, with consequent damage. Furthermore, the adaptive immunity triggered by hRSV infection is characterized by weak cytotoxic T cell responses and secretion of low affinity antibodies by B cells. These features of hRSV infection have meant that, to date, no effective and safe vaccines have been licensed. In this article, we will review in detail the information regarding hRSV characteristics, pathology, and host immune response, along with several prophylactic treatments and vaccine prototypes. We will also expose significant data regarding the newly developed BCG-based vaccine that promotes protective cellular and humoral response against hRSV infection, which is currently undergoing clinical evaluation.
Collapse
|
28
|
Rossi GA, Colin AA. Respiratory syncytial virus-Host interaction in the pathogenesis of bronchiolitis and its impact on respiratory morbidity in later life. Pediatr Allergy Immunol 2017; 28:320-331. [PMID: 28339145 DOI: 10.1111/pai.12716] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/13/2017] [Indexed: 02/06/2023]
Abstract
Respiratory syncytial virus (RSV) is the most common agent of severe airway disease in infants and young children. Large epidemiologic studies have demonstrated a clear relationship between RSV infection and subsequent recurrent wheezing and asthma into childhood, thought to be predominantly related to long-term changes in neuroimmune control of airway tone rather than to allergic sensitization. These changes appear to be governed by the severity of the first RSV infection in infancy which in term depends on viral characteristics and load, but perhaps as importantly, on the genetic susceptibility and on the constitutional characteristic of the host. A variety of viral and host factors and their interplay modify the efficiency of the response to infection, including viral replication and the magnitude of structural and functional damage to the respiratory structures, and ultimately the extent, severity, and duration of subsequent wheezing.
Collapse
Affiliation(s)
- Giovanni A Rossi
- Pulmonary and Allergy Disease Pediatric Unit and Cystic Fibrosis Center, Istituto Giannina Gaslini, Genoa, Italy
| | - Andrew A Colin
- Division of Pediatric Pulmonology, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
29
|
Porto BN, Stein RT. Neutrophil Extracellular Traps in Pulmonary Diseases: Too Much of a Good Thing? Front Immunol 2016; 7:311. [PMID: 27574522 PMCID: PMC4983612 DOI: 10.3389/fimmu.2016.00311] [Citation(s) in RCA: 230] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/02/2016] [Indexed: 12/30/2022] Open
Abstract
Neutrophil extracellular traps (NETs) arise from the release of granular and nuclear contents of neutrophils in the extracellular space in response to different classes of microorganisms, soluble factors, and host molecules. NETs are composed by decondensed chromatin fibers coated with antimicrobial granular and cytoplasmic proteins, such as myeloperoxidase, neutrophil elastase (NE), and α-defensins. Besides being expressed on NET fibers, NE and MPO also regulate NET formation. Furthermore, histone deimination by peptidylarginine deiminase 4 (PAD4) is a central step to NET formation. NET formation has been widely demonstrated to be an effective mechanism to fight against invading microorganisms, as deficiency in NET release or dismantling NET backbone by bacterial DNases renders the host susceptible to infections. Therefore, the primary role of NETs is to prevent microbial dissemination, avoiding overwhelming infections. However, an excess of NET formation has a dark side. The pathogenic role of NETs has been described for many human diseases, infectious and non-infectious. The detrimental effect of excessive NET release is particularly important to lung diseases, because NETs can expand more easily in the pulmonary alveoli, causing lung injury. Moreover, NETs and its associated molecules are able to directly induce epithelial and endothelial cell death. In this regard, massive NET formation has been reported in several pulmonary diseases, including asthma, chronic obstructive pulmonary disease, cystic fibrosis, respiratory syncytial virus bronchiolitis, influenza, bacterial pneumonia, and tuberculosis, among others. Thus, NET formation must be tightly regulated in order to avoid NET-mediated tissue damage. Recent development of therapies targeting NETs in pulmonary diseases includes DNA disintegration with recombinant human DNase, neutralization of NET proteins, with anti-histone antibodies and protease inhibitors. In this review, we summarize the recent knowledge on the pathophysiological role of NETs in pulmonary diseases as well as some experimental and clinical approaches to modulate their detrimental effects.
Collapse
Affiliation(s)
- Bárbara Nery Porto
- Laboratory of Clinical and Experimental Immunology, Infant Center, Institute of Biomedical Research, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Renato Tetelbom Stein
- Laboratory of Pediatric Respirology, Infant Center, Institute of Biomedical Research, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
30
|
Bohmwald K, Espinoza JA, Rey-Jurado E, Gómez RS, González PA, Bueno SM, Riedel CA, Kalergis AM. Human Respiratory Syncytial Virus: Infection and Pathology. Semin Respir Crit Care Med 2016; 37:522-37. [PMID: 27486734 PMCID: PMC7171722 DOI: 10.1055/s-0036-1584799] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The human respiratory syncytial virus (hRSV) is by far the major cause of acute lower respiratory tract infections (ALRTIs) worldwide in infants and children younger than 2 years. The overwhelming number of hospitalizations due to hRSV-induced ALRTI each year is due, at least in part, to the lack of licensed vaccines against this virus. Thus, hRSV infection is considered a major public health problem and economic burden in most countries. The lung pathology developed in hRSV-infected individuals is characterized by an exacerbated proinflammatory and unbalanced Th2-type immune response. In addition to the adverse effects in airway tissues, hRSV infection can also cause neurologic manifestations in the host, such as seizures and encephalopathy. Although the origins of these extrapulmonary symptoms remain unclear, studies with patients suffering from neurological alterations suggest an involvement of the inflammatory response against hRSV. Furthermore, hRSV has evolved numerous mechanisms to modulate and evade the immune response in the host. Several studies have focused on elucidating the interactions between hRSV virulence factors and the host immune system, to rationally design new vaccines and therapies against this virus. Here, we discuss about the infection, pathology, and immune response triggered by hRSV in the host.
Collapse
Affiliation(s)
- Karen Bohmwald
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Janyra A Espinoza
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Emma Rey-Jurado
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roberto S Gómez
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Departamento de Ciencias Biológicas y Facultad de Medicina, Millennium Institute on Immunology and Immunotherapy, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
31
|
Cervantes-Ortiz SL, Zamorano Cuervo N, Grandvaux N. Respiratory Syncytial Virus and Cellular Stress Responses: Impact on Replication and Physiopathology. Viruses 2016; 8:v8050124. [PMID: 27187445 PMCID: PMC4885079 DOI: 10.3390/v8050124] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/14/2016] [Accepted: 04/21/2016] [Indexed: 02/08/2023] Open
Abstract
Human respiratory syncytial virus (RSV), a member of the Paramyxoviridae family, is a major cause of severe acute lower respiratory tract infection in infants, elderly and immunocompromised adults. Despite decades of research, a complete integrated picture of RSV-host interaction is still missing. Several cellular responses to stress are involved in the host-response to many virus infections. The endoplasmic reticulum stress induced by altered endoplasmic reticulum (ER) function leads to activation of the unfolded-protein response (UPR) to restore homeostasis. Formation of cytoplasmic stress granules containing translationally stalled mRNAs is a means to control protein translation. Production of reactive oxygen species is balanced by an antioxidant response to prevent oxidative stress and the resulting damages. In recent years, ongoing research has started to unveil specific regulatory interactions of RSV with these host cellular stress responses. Here, we discuss the latest findings regarding the mechanisms evolved by RSV to induce, subvert or manipulate the ER stress, the stress granule and oxidative stress responses. We summarize the evidence linking these stress responses with the regulation of RSV replication and the associated pathogenesis.
Collapse
Affiliation(s)
- Sandra L Cervantes-Ortiz
- CRCHUM-Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X 0A9, Canada.
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada.
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montréal, QC H3C 3J7, Canada.
| | - Natalia Zamorano Cuervo
- CRCHUM-Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X 0A9, Canada.
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada.
| | - Nathalie Grandvaux
- CRCHUM-Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X 0A9, Canada.
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada.
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada.
| |
Collapse
|
32
|
Noval MG, Esperante SA, Molina IG, Chemes LB, Prat-Gay GD. Intrinsic Disorder to Order Transitions in the Scaffold Phosphoprotein P from the Respiratory Syncytial Virus RNA Polymerase Complex. Biochemistry 2016; 55:1441-54. [PMID: 26901160 DOI: 10.1021/acs.biochem.5b01332] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Intrinsic disorder is at the center of biochemical regulation and is particularly overrepresented among the often multifunctional viral proteins. Replication and transcription of the respiratory syncytial virus (RSV) relies on a RNA polymerase complex with a phosphoprotein cofactor P as the structural scaffold, which consists of a four-helix bundle tetramerization domain flanked by two domains predicted to be intrinsically disordered. Because intrinsic disorder cannot be reduced to a defined atomic structure, we tackled the experimental dissection of the disorder-order transitions of P by a domain fragmentation approach. P remains as a tetramer above 70 °C but shows a pronounced reversible secondary structure transition between 10 and 60 °C. While the N-terminal module behaves as a random coil-like IDP in a manner independent of tetramerization, the isolated C-terminal module displays a cooperative and reversible metastable transition. When linked to the tetramerization domain, the C-terminal module becomes markedly more structured and stable, with strong ANS binding. Therefore, the tertiary structure in the C-terminal module is not compact, conferring "late" molten globule-like IDP properties, stabilized by interactions favored by tetramerization. The presence of a folded structure highly sensitive to temperature, reversibly and almost instantly formed and broken, suggests a temperature sensing activity. The marginal stability allows for exposure of protein binding sites, offering a thermodynamic and kinetic fine-tuning in order-disorder transitions, essential for the assembly and function of the RSV RNA polymerase complex.
Collapse
Affiliation(s)
- María G Noval
- Protein Structure-Function and Engineering Laboratory, Fundación Instituto Leloir and IIBBA-CONICET , Av. Patricias Argentinas 435, 1405 Buenos Aires, Argentina
| | - Sebastian A Esperante
- Protein Structure-Function and Engineering Laboratory, Fundación Instituto Leloir and IIBBA-CONICET , Av. Patricias Argentinas 435, 1405 Buenos Aires, Argentina
| | - Ivana G Molina
- Protein Structure-Function and Engineering Laboratory, Fundación Instituto Leloir and IIBBA-CONICET , Av. Patricias Argentinas 435, 1405 Buenos Aires, Argentina
| | - Lucía B Chemes
- Protein Structure-Function and Engineering Laboratory, Fundación Instituto Leloir and IIBBA-CONICET , Av. Patricias Argentinas 435, 1405 Buenos Aires, Argentina
| | - Gonzalo de Prat-Gay
- Protein Structure-Function and Engineering Laboratory, Fundación Instituto Leloir and IIBBA-CONICET , Av. Patricias Argentinas 435, 1405 Buenos Aires, Argentina.,CNPq, Laboratório de Genômica Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro, RJ, Brazil
| |
Collapse
|
33
|
The Interferon Type I/III Response to Respiratory Syncytial Virus Infection in Airway Epithelial Cells Can Be Attenuated or Amplified by Antiviral Treatment. J Virol 2015; 90:1705-17. [PMID: 26608311 DOI: 10.1128/jvi.02417-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/16/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Human respiratory syncytial virus (RSV) is a single-stranded RNA virus that causes acute, and occasionally fatal, lower respiratory illness in young infants, the elderly, and immunocompromised patients. Therapeutic interventions able to cut short viral replication and quickly return the airways to normal function are needed. An understanding of antiviral activities and their effects on host defense mechanisms is important for the design of safe and effective therapy. We targeted functionally and temporally distinct steps within the viral life cycle using small-molecule RSV inhibitors and studied their antiviral activities and their effects on innate interferon responses of airway epithelial cells in vitro. Antivirals acting upstream of RSV polymerase activity (i.e., compounds targeting the fusion protein or the nucleoprotein) reduced viral load immediately postinfection and partially attenuated interferon responses. In contrast, antivirals directed to the RSV polymerase demonstrated activity throughout the viral replication cycle and specifically modulated the RIG-I/mitochondrial antiviral signaling protein (MAVS)/TBK1/IRF3/interferon-stimulated gene (ISG) axis, causing either an upregulation or a downregulation of interferon responses, depending on the mechanism of polymerase inhibition. Notably, polymerase inhibition leading to the accumulation of abortive RNA products correlated with the amplification of interferon-stimulated genes to up to 10 times above normal infection levels. Understanding how antiviral activities and their modulation of innate immunity may affect recovery from RSV infection will help guide the development of safe and effective therapies. IMPORTANCE RSV circulates seasonally, causing acute lower respiratory disease. Therapeutic interventions with efficacy throughout the viral replication cycle, rapid viral clearance, and prevention of potentially harmful inflammatory responses are desirable. Compounds targeting the RSV polymerase inhibited virus replication late in the viral life cycle and, depending on the functional domain targeted, either attenuated or amplified RIG-I and downstream interferon pathways in infected cells. These data will help guide the development of safe and effective therapies by providing new molecular evidence that the mechanism of inhibition by an antiviral compound can directly impact innate antiviral immune responses in the airway epithelium.
Collapse
|
34
|
Rivera CA, Gómez RS, Díaz RA, Céspedes PF, Espinoza JA, González PA, Riedel CA, Bueno SM, Kalergis AM. Novel therapies and vaccines against the human respiratory syncytial virus. Expert Opin Investig Drugs 2015; 24:1613-30. [DOI: 10.1517/13543784.2015.1099626] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
35
|
Cao H, Ouyang H, Ip W, Du K, Duan W, Avolio J, Wu J, Duan C, Yeger H, Bear CE, Gonska T, Hu J, Moraes TJ. Testing gene therapy vectors in human primary nasal epithelial cultures. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2015; 2:15034. [PMID: 26730394 PMCID: PMC4685663 DOI: 10.1038/mtm.2015.34] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 07/27/2015] [Accepted: 08/05/2015] [Indexed: 01/01/2023]
Abstract
Cystic fibrosis (CF) results from mutations in the CF transmembrane conductance regulator (CFTR) gene, which codes for a chloride/bicarbonate channel in the apical epithelial membranes. CFTR dysfunction results in a multisystem disease including the development of life limiting lung disease. The possibility of a cure for CF by replacing defective CFTR has led to different approaches for CF gene therapy; all of which ultimately have to be tested in preclinical model systems. Primary human nasal epithelial cultures (HNECs) derived from nasal turbinate brushing were used to test the efficiency of a helper-dependent adenoviral (HD-Ad) vector expressing CFTR. HD-Ad-CFTR transduction resulted in functional expression of CFTR at the apical membrane in nasal epithelial cells obtained from CF patients. These results suggest that HNECs can be used for preclinical testing of gene therapy vectors in CF.
Collapse
Affiliation(s)
- Huibi Cao
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Hong Ouyang
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Wan Ip
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Kai Du
- Programme in Molecular Structure & Function, Research Institute, Hospital for Sick Children , Toronto, Ontario, Canada
| | - Wenming Duan
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Julie Avolio
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Jing Wu
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Cathleen Duan
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Herman Yeger
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Christine E Bear
- Programme in Molecular Structure & Function, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Tanja Gonska
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children, Toronto, Onatrio, Canada; Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Jim Hu
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children, Toronto, Onatrio, Canada; Programme in Molecular Structure & Function, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Theo J Moraes
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children, Toronto, Onatrio, Canada; Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
36
|
Brown PM, Schneeberger DL, Piedimonte G. Biomarkers of respiratory syncytial virus (RSV) infection: specific neutrophil and cytokine levels provide increased accuracy in predicting disease severity. Paediatr Respir Rev 2015; 16:232-40. [PMID: 26074450 PMCID: PMC4656140 DOI: 10.1016/j.prrv.2015.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 12/17/2022]
Abstract
Despite fundamental advances in the research on respiratory syncytial virus (RSV) since its initial identification almost 60 years ago, recurring failures in developing vaccines and pharmacologic strategies effective in controlling the infection have allowed RSV to become a leading cause of global infant morbidity and mortality. Indeed, the burden of this infection on families and health care organizations worldwide continues to escalate and its financial costs are growing. Furthermore, strong epidemiologic evidence indicates that early-life lower respiratory tract infections caused by RSV lead to the development of recurrent wheezing and childhood asthma. While some progress has been made in the identification of reliable biomarkers for RSV bronchiolitis, a "one size fits all" biomarker capable of accurately and consistently predicting disease severity and post-acute outcomes has yet to be discovered. Therefore, it is of great importance on a global scale to identify useful biomarkers for this infection that will allow pediatricians to cost-effectively predict the clinical course of the disease, as well as monitor the efficacy of new therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Giovanni Piedimonte
- Center for Pediatric Research, Pediatric Institute and Children's Hospitals, The Cleveland Clinic.
| |
Collapse
|
37
|
Bertrand P, Lay MK, Piedimonte G, Brockmann PE, Palavecino CE, Hernández J, León MA, Kalergis AM, Bueno SM. Elevated IL-3 and IL-12p40 levels in the lower airway of infants with RSV-induced bronchiolitis correlate with recurrent wheezing. Cytokine 2015; 76:417-423. [PMID: 26299549 DOI: 10.1016/j.cyto.2015.07.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 12/30/2022]
Abstract
Respiratory Syncytial Virus (RSV) is the first cause of hospitalization due to bronchiolitis in infants. RSV bronchiolitis has been linked to asthma and recurrent wheezing, however the mechanisms behind this association have not been elucidated. Here, we evaluated the cytokine and chemokine profiles in the airways in infants with RSV bronchiolitis. Nasopharyngeal Aspirates (NPA) and Bronchoalveolar Lavage Fluids (BALF) from infants hospitalized due to RSV bronchiolitis and healthy controls were analyzed for cytokine and chemokine production. We observed elevated levels of Th2 cytokines (IL-3, IL-4, IL-10 and IL-13), pro-inflammatory cytokines and chemokines (IL-1β, IL-6, TNF-β, MCP-1/CCL2, MIP-1α/CCL3 and IL-8/CXCL8) in BALF from infants with RSV bronchiolitis, as compared to controls. We found a direct correlation of IL-3 and IL-12p40 levels with the development of recurrent wheezing later in life. These results suggest that IL-3 and IL-12p40 could be considered as molecular predictors for recurrent wheezing due to RSV infection.
Collapse
Affiliation(s)
- Pablo Bertrand
- División de Pediatría, Unidad de Enfermedades Respiratorias Pediátricas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K Lay
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Giovanni Piedimonte
- The Cleveland Clinic Pediatric Institute and Children's Hospital, Cleveland, OH, United States
| | - Pablo E Brockmann
- División de Pediatría, Unidad de Enfermedades Respiratorias Pediátricas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christian E Palavecino
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jury Hernández
- División de Pediatría, Unidad de Enfermedades Respiratorias Pediátricas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel A León
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Reumatología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; INSERM U1064, Nantes, France
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Reumatología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
38
|
Guo X, Liu T, Shi H, Wang J, Ji P, Wang H, Hou Y, Tan RX, Li E. Respiratory Syncytial Virus Infection Upregulates NLRC5 and Major Histocompatibility Complex Class I Expression through RIG-I Induction in Airway Epithelial Cells. J Virol 2015; 89:7636-45. [PMID: 25972545 PMCID: PMC4505675 DOI: 10.1128/jvi.00349-15] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/06/2015] [Indexed: 01/11/2023] Open
Abstract
UNLABELLED Respiratory syncytial virus (RSV) is the leading cause of acute respiratory tract viral infection in infants, causing bronchiolitis and pneumonia. The host antiviral response to RSV acts via retinoic acid-inducible gene I (RIG-I). We show here that RSV infection upregulates major histocompatibility complex class I (MHC-I) expression through the induction of NLRC5, a NOD-like, CARD domain-containing intracellular protein that has recently been identified as a class I MHC transactivator (CITA). RSV infection of A549 cells promotes upregulation of NLRC5 via beta interferon (IFN-β) production, since the NLRC5-inducing activity in a conditioned medium from RSV-infected A549 cells was removed by antibody to IFN-β, but not by antibody to IFN-γ. RSV infection resulted in RIG-I upregulation and induction of NLRC5 and MHC-I. Suppression of RIG-I induction significantly blocked NLRC5, as well as MHC-I, upregulation and diminished IRF3 activation. Importantly, Vero cells deficient in interferon production still upregulated MHC-I following introduction of the RSV genome by infection or transfection, further supporting a key role for RIG-I. A model is therefore proposed in which the host upregulates MHC-I expression during RSV infection directly via the induction of RIG-I and NLRC5 expression. Since elevated expression of MHC-I molecules can sensitize host cells to T lymphocyte-mediated cytotoxicity or immunopathologic damage, the results have significant implications for the modification of immunity in RSV disease. IMPORTANCE Human respiratory syncytial virus (RSV) is the leading cause of bronchiolitis and pneumonia in infants and young children worldwide. Infection early in life is linked to persistent wheezing and allergic asthma in later life, possibly related to upregulation of major histocompatibility class I (MHC-I) on the cell surface, which facilitates cytotoxic T cell activation and antiviral immunity. Here, we show that RSV infection of lung epithelial cells induces expression of RIG-I, resulting in induction of a class I MHC transactivator, NLRC5, and subsequent upregulation of MHC-I. Suppression of RIG-I induction blocked RSV-induced NLRC5 expression and MHC-I upregulation. Increased MHC-I expression may exacerbate the RSV disease condition due to immunopathologic damage, linking the innate immune response to RSV disease.
Collapse
Affiliation(s)
- Xuancheng Guo
- State Key Laboratory of Pharmaceutical Biotechnology and School of Medicine, Nanjing University, Nanjing, Jiangsu, China Jiangsu Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Taixiang Liu
- Jiangsu Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China Jiangsu Province Blood Center, Nanjing, Jiangsu, China
| | - Hengfei Shi
- State Key Laboratory of Pharmaceutical Biotechnology and School of Medicine, Nanjing University, Nanjing, Jiangsu, China Jiangsu Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Jingjing Wang
- State Key Laboratory of Pharmaceutical Biotechnology and School of Medicine, Nanjing University, Nanjing, Jiangsu, China College of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Ping Ji
- State Key Laboratory of Pharmaceutical Biotechnology and School of Medicine, Nanjing University, Nanjing, Jiangsu, China Jiangsu Province Blood Center, Nanjing, Jiangsu, China
| | - Hongwei Wang
- Jiangsu Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Yayi Hou
- State Key Laboratory of Pharmaceutical Biotechnology and School of Medicine, Nanjing University, Nanjing, Jiangsu, China Jiangsu Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Ren Xiang Tan
- State Key Laboratory of Pharmaceutical Biotechnology and School of Medicine, Nanjing University, Nanjing, Jiangsu, China College of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Erguang Li
- State Key Laboratory of Pharmaceutical Biotechnology and School of Medicine, Nanjing University, Nanjing, Jiangsu, China Jiangsu Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
39
|
Espinoza JA, Bohmwald K, Céspedes PF, Riedel CA, Bueno SM, Kalergis AM. Modulation of host adaptive immunity by hRSV proteins. Virulence 2015; 5:740-51. [PMID: 25513775 PMCID: PMC4189880 DOI: 10.4161/viru.32225] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Globally, the human respiratory syncytial virus (hRSV) is the major cause of lower respiratory tract infections (LRTIs) in infants and children younger than 2 years old. Furthermore, the number of hospitalizations due to LRTIs has shown a sustained increase every year due to the lack of effective vaccines against hRSV. Thus, this virus remains as a major public health and economic burden worldwide. The lung pathology developed in hRSV-infected humans is characterized by an exacerbated inflammatory and Th2 immune response. In order to rationally design new vaccines and therapies against this virus, several studies have focused in elucidating the interactions between hRSV virulence factors and the host immune system. Here, we discuss the main features of hRSV biology, the processes involved in virus recognition by the immune system and the most relevant mechanisms used by this pathogen to avoid the antiviral host response.
Collapse
Affiliation(s)
- Janyra A Espinoza
- a Millenium Institute on Immunology and Immunotherapy; Departamento de Genética Molecular y Microbiología; Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago, Chile
| | | | | | | | | | | |
Collapse
|
40
|
Lay MK, Bueno SM, Gálvez N, Riedel CA, Kalergis AM. New insights on the viral and host factors contributing to the airway pathogenesis caused by the respiratory syncytial virus. Crit Rev Microbiol 2015; 42:800-12. [PMID: 26119025 DOI: 10.3109/1040841x.2015.1055711] [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: 12/21/2022]
Abstract
The respiratory syncytial virus (RSV) is the most prevalent etiological agent of lower respiratory tract infections and the first cause of hospitalization in infants due to respiratory disease worldwide. However, efforts to develop safe and effective vaccines and antivirals have been challenged by an incomplete understanding of the RSV pathogenesis and the host immune response to RSV infection in the airways. Here, we discuss recent advances in understanding the interaction between RSV and the epithelium to induce pathogenesis in the airways, such as the role of the RSV NS2 protein in the airway epithelium, as well as the events involved in the RSV entry process. In addition, we summarize the cellular factors produced by airway epithelial cells (AECs) in response to RSV infection that lead to the activation of innate and adaptive immune responses, inducing lung inflammation and disease. Further, we discuss the possible contribution of a recently identified cytokine, thymic stromal lymphopoitein (TSLP), in the lung immunopathology caused by RSV.
Collapse
Affiliation(s)
- Margarita K Lay
- a Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Susan M Bueno
- a Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile , Santiago , Chile .,b INSERM U1064 , Nantes , France
| | - Nicolás Gálvez
- a Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Claudia A Riedel
- c Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas y Facultad de Medicina , Laboratorio de Biología Celular y Farmacología, Millennium Institute on Immunology and Immunotherapy, Universidad Andrés Bello , Santiago , Chile , and
| | - Alexis M Kalergis
- a Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile , Santiago , Chile .,b INSERM U1064 , Nantes , France .,d Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina , Pontificia Universidad Católica de Chile , Santiago , Chile
| |
Collapse
|
41
|
Hong Z, Xu Y, Yin JF, Jin J, Jiang Y, Du Q. Improving the effectiveness of (-)-epigallocatechin gallate (EGCG) against rabbit atherosclerosis by EGCG-loaded nanoparticles prepared from chitosan and polyaspartic acid. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:2182-9. [PMID: 25483592 DOI: 10.1021/jf404310y] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
(-)-Epigallocatechin gallate (EGCG) is the major bioactive compound in green tea. Its effect is limited by the harsh environment of the gastrointestinal tract. The present study investigates how the effectiveness of EGCG is influenced by its encapsulation into self-assembled nanoparticles of chitosan (CS) and aspartic acid (PAA). Blank nanoparticles with a mean diameter of ca. 93 nm were prepared from 30-50 kDa PAA and 3-5 kDa CS with a mass rate of 1:1. EGCG was loaded in the nanoparticles to yield EGCG-CS-PAA nanoparticles with an average diameter of 102 nm, which were pH-responsive and demonstrated different EGCG release profiles in simulated gastrointestinal tract media. The average ratio (%) of lipid deposition for EGCG-CS-PAA nanoparticles administered orally to rabbits was 16.9 ± 5.8%, which was close to that of oral simvastatin (15.6 ± 4.1%). Orally administered EGCG alone yielded an average ratio of lipid deposit area of 42.1 ± 4.0%, whereas this value was 65.3 ± 10.8% for the blank nanoparticles. The effectiveness of EGCG against rabbit atherosclerosis was significantly improved by incorporating EGCG into the nanoformulation.
Collapse
Affiliation(s)
- Zhiyong Hong
- Institute of Food Chemistry, Zhejiang A&F University , 88 Huanbei Road, Hangzhou, Zhejiang 311300, China
| | | | | | | | | | | |
Collapse
|
42
|
Bohmwald K, Espinoza JA, González PA, Bueno SM, Riedel CA, Kalergis AM. Central nervous system alterations caused by infection with the human respiratory syncytial virus. Rev Med Virol 2014; 24:407-19. [PMID: 25316031 DOI: 10.1002/rmv.1813] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 08/31/2014] [Accepted: 09/02/2014] [Indexed: 01/08/2023]
Abstract
Worldwide, the human respiratory syncytial virus (hRSV) is the leading cause of infant hospitalization because of acute respiratory tract infections, including severe bronchiolitis and pneumonia. Despite intense research, to date there is neither vaccine nor treatment available to control hRSV disease burden globally. After infection, an incubation period of 3-5 days is usually followed by symptoms, such as cough and low-grade fever. However, hRSV infection can also produce a larger variety of symptoms, some of which relate to the individual's age at infection. Indeed, infants can display severe symptoms, such as dyspnea and chest wall retractions. Upon examination, crackles and wheezes are also common features that suggest infection by hRSV. Additionally, infection in infants younger than 1 year is associated with several non-specific symptoms, such as failure to thrive, periodic breathing or apnea, and feeding difficulties that usually require hospitalization. Recently, neurological symptoms have also been associated with hRSV respiratory infection and include seizures, central apnea, lethargy, feeding or swallowing difficulties, abnormalities in muscle tone, strabismus, abnormalities in the CSF, and encephalopathy. Here, we discuss recent findings linking the neurological, extrapulmonary effects of hRSV with infection and functional impairment of the CNS.
Collapse
Affiliation(s)
- Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | | | |
Collapse
|
43
|
Foronjy RF, Dabo AJ, Cummins N, Geraghty P. Leukemia inhibitory factor protects the lung during respiratory syncytial viral infection. BMC Immunol 2014; 15:41. [PMID: 25277705 PMCID: PMC4189665 DOI: 10.1186/s12865-014-0041-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 09/15/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infects the lung epithelium where it stimulates the production of numerous host cytokines that are associated with disease burden and acute lung injury. Characterizing the host cytokine response to RSV infection, the regulation of host cytokines and the impact of neutralizing an RSV-inducible cytokine during infection were undertaken in this study. METHODS A549, primary human small airway epithelial (SAE) cells and wild-type, TIR-domain-containing adapter-inducing interferon-β (Trif) and mitochondrial antiviral-signaling protein (Mavs) knockout (KO) mice were infected with RSV and cytokine responses were investigated by ELISA, multiplex analysis and qPCR. Neutralizing anti-leukemia inhibitory factor (LIF) IgG or control IgG was administered to a group of wild-type animals prior to RSV infection. RESULTS AND DISCUSSION RSV-infected A549 and SAE cells release a network of cytokines, including newly identified RSV-inducible cytokines LIF, migration inhibitory factor (MIF), stem cell factor (SCF), CCL27, CXCL12 and stem cell growth factor beta (SCGF-β). These RSV-inducible cytokines were also observed in the airways of mice during an infection. To identify the regulation of RSV inducible cytokines, Mavs and Trif deficient animals were infected with RSV. In vivo induction of airway IL-1β, IL-4, IL-5, IL-6, IL-12(p40), IFN-γ, CCL2, CCL5, CCL3, CXCL1, IP-10/CXCL10, IL-22, MIG/CXCL9 and MIF were dependent on Mavs expression in mice. Loss of Trif expression in mice altered the RSV induction of IL-1β, IL-5, CXCL12, MIF, LIF, CXCL12 and IFN-γ. Silencing of retinoic acid-inducible gene-1 (RIG-I) expression in A549 cells had a greater impact on RSV-inducible cytokines than melanoma differentiation-associated protein 5 (MDA5) and laboratory of genetics and physiology 2 (LGP2), and Trif expression. To evaluate the role of LIF in the airways during RSV infection, animals were treated with neutralizing anti-LIF IgG, which enhanced RSV pathology observed with increased airspace protein content, apoptosis and airway hyperresponsiveness compared to control IgG treatment. CONCLUSIONS RSV infection in the epithelium induces a network of immune factors to counter infection, primarily in a RIG-I dependent manner. Expression of LIF protects the lung from lung injury and enhanced pathology during RSV infection.
Collapse
|
44
|
Gomez RS, Guisle-Marsollier I, Bohmwald K, Bueno SM, Kalergis AM. Respiratory Syncytial Virus: pathology, therapeutic drugs and prophylaxis. Immunol Lett 2014; 162:237-47. [PMID: 25268876 DOI: 10.1016/j.imlet.2014.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 08/21/2014] [Accepted: 09/08/2014] [Indexed: 11/16/2022]
Abstract
Human Respiratory Syncytial Virus (hRSV) is the leading cause of lower respiratory tract diseases, affecting particularly newborns and young children. This virus is able to modulate the immune response, generating a pro-inflammatory environment in the airways that causes obstruction and pulmonary alterations in the infected host. To date, no vaccines are available for human use and the first vaccine that reached clinical trials produced an enhanced hRSV-associated pathology 50 years ago, resulting in the death of two children. Currently, only two therapeutic approaches have been used to treat hRSV infection in high risk children: 1. Palivizumab, a humanized antibody against the F glycoprotein that reduces to half the number of hospitalized cases and 2. Ribavirin, which fails to have a significant therapeutic effect. A major caveat for these approaches is their high economical cost, which highlights the need of new and affordable therapeutic or prophylactic tools to treat or prevents hRSV infection. Accordingly, several efforts are in progress to understand the hRSV-associated pathology and to characterize the immune response elicited by this virus. Currently, preclinical and clinical trials are being conducted to evaluate safety and efficacy of several drugs and vaccines, which have shown promising results. In this article, we discuss the most important advances in the development of drugs and vaccines, which could eventually lead to better strategies to treat or prevent the detrimental inflammation triggered by hRSV infection.
Collapse
Affiliation(s)
- Roberto S Gomez
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Chile; INSERM U1064, Nantes, France
| | | | - Karen Bohmwald
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Chile
| | - Susan M Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Chile; INSERM U1064, Nantes, France
| | - Alexis M Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Chile; Departamento de Reumatología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile; INSERM U1064, Nantes, France.
| |
Collapse
|
45
|
Espinoza JA, Bueno SM, Riedel CA, Kalergis AM. Induction of protective effector immunity to prevent pathogenesis caused by the respiratory syncytial virus. Implications on therapy and vaccine design. Immunology 2014; 143:1-12. [PMID: 24801878 DOI: 10.1111/imm.12313] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 04/29/2014] [Accepted: 05/01/2014] [Indexed: 01/22/2023] Open
Abstract
Human respiratory syncytial virus (hRSV) is the leading cause of respiratory illness in infants and young children around the globe. This pathogen, which was discovered in 1956, continues to cause a huge number of hospitalizations due to respiratory disease and it is considered a health and economic burden worldwide, especially in developing countries. The immune response elicited by hRSV infection leads to lung and systemic inflammation, which results in lung damage but is not efficient at preventing viral replication. Indeed, natural hRSV infection induces a poor immune memory that allows recurrent infections. Here, we review the most recent knowledge about the lifecycle of hRSV, the immune response elicited by this virus and the subsequent pathology induced in response to infection in the airways. Novel findings about the alterations that this virus causes in the central nervous system and potential therapies and vaccines designed to treat or prevent hRSV infection are discussed.
Collapse
Affiliation(s)
- Janyra A Espinoza
- Millennium Institute on Immunology, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | |
Collapse
|
46
|
Characterization of a respiratory syncytial virus L protein inhibitor. Antimicrob Agents Chemother 2014; 58:3867-73. [PMID: 24777090 DOI: 10.1128/aac.02540-14] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The respiratory syncytial virus (RSV) L protein is a viral RNA-dependent RNA polymerase that contains multiple enzyme activities required for RSV replication. The RSV L inhibitors described in literature are limited by their cytotoxicity or the lack of RSV B subtype coverage. Here, we characterize a new RSV L inhibitor with strong antiviral activity against both RSV A and B subtypes and no detectable cytotoxicity. This compound, AZ-27, was equally active against RSV live viruses and subgenomic replicons and demonstrated advantages over other classes of RSV inhibitors in time-of-addition and cell line dependency studies. Resistance studies identified a dominant mutation in the putative capping enzyme domain of L protein, which conferred strong resistance to the AZ-27 series but not other classes of RSV inhibitors, supporting RSV L protein as the direct target for AZ-27. This novel and broad-spectrum RSV L polymerase inhibitor may pave the way toward an efficacious RSV therapeutic and provide a new tool for interrogation of the L protein function.
Collapse
|
47
|
Everitt AR, Clare S, McDonald JU, Kane L, Harcourt K, Ahras M, Lall A, Hale C, Rodgers A, Young DB, Haque A, Billker O, Tregoning JS, Dougan G, Kellam P. Defining the range of pathogens susceptible to Ifitm3 restriction using a knockout mouse model. PLoS One 2013; 8:e80723. [PMID: 24278312 PMCID: PMC3836756 DOI: 10.1371/journal.pone.0080723] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/16/2013] [Indexed: 12/22/2022] Open
Abstract
The interferon-inducible transmembrane (IFITM) family of proteins has been shown to restrict a broad range of viruses in vitro and in vivo by halting progress through the late endosomal pathway. Further, single nucleotide polymorphisms (SNPs) in its sequence have been linked with risk of developing severe influenza virus infections in humans. The number of viruses restricted by this host protein has continued to grow since it was first demonstrated as playing an antiviral role; all of which enter cells via the endosomal pathway. We therefore sought to test the limits of antimicrobial restriction by Ifitm3 using a knockout mouse model. We showed that Ifitm3 does not impact on the restriction or pathogenesis of bacterial (Salmonella typhimurium, Citrobacter rodentium, Mycobacterium tuberculosis) or protozoan (Plasmodium berghei) pathogens, despite in vitro evidence. However, Ifitm3 is capable of restricting respiratory syncytial virus (RSV) in vivo either through directly restricting RSV cell infection, or by exerting a previously uncharacterised function controlling disease pathogenesis. This represents the first demonstration of a virus that enters directly through the plasma membrane, without the need for the endosomal pathway, being restricted by the IFITM family; therefore further defining the role of these antiviral proteins.
Collapse
Affiliation(s)
- Aaron R. Everitt
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
- * E-mail:
| | - Simon Clare
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Jacqueline U. McDonald
- Mucosal Infection and Immunity Group, Section of Infectious Diseases, Department of Medicine, Imperial College London, London, United Kingdom
| | - Leanne Kane
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Katherine Harcourt
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Malika Ahras
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Amar Lall
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Christine Hale
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Angela Rodgers
- Medical Research Council National Institute for Medical Research, London, United Kingdom
| | - Douglas B. Young
- Medical Research Council National Institute for Medical Research, London, United Kingdom
| | - Ashraful Haque
- Malaria Immunology Laboratory, Queensland Institute of Medical Research and The Australian Centre for Vaccine Development, Herston, Brisbane, Queensland, Australia
| | - Oliver Billker
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - John S. Tregoning
- Mucosal Infection and Immunity Group, Section of Infectious Diseases, Department of Medicine, Imperial College London, London, United Kingdom
| | - Gordon Dougan
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Paul Kellam
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
- Department of Infection, University College London, London, United Kingdom
| |
Collapse
|
48
|
Roflumilast inhibits respiratory syncytial virus infection in human differentiated bronchial epithelial cells. PLoS One 2013; 8:e69670. [PMID: 23936072 PMCID: PMC3720563 DOI: 10.1371/journal.pone.0069670] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 06/11/2013] [Indexed: 11/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) causes acute exacerbations in COPD and asthma. RSV infects bronchial epithelial cells (HBE) that trigger RSV associated lung pathology. This study explores whether the phosphodiesterase 4 (PDE4) inhibitor Roflumilast N-oxide (RNO), alters RSV infection of well-differentiated HBE (WD-HBE) in vitro. WD-HBE were RSV infected in the presence or absence of RNO (0.1-100 nM). Viral infection (staining of F and G proteins, nucleoprotein RNA level), mRNA of ICAM-1, ciliated cell markers (digital high speed videomicroscopy, β-tubulin immunofluorescence, Foxj1 and Dnai2 mRNA), Goblet cells (PAS), mRNA of MUC5AC and CLCA1, mRNA and protein level of IL-13, IL-6, IL-8, TNFα, formation of H2O2 and the anti-oxidative armamentarium (mRNA of Nrf2, HO-1, GPx; total antioxidant capacity (TAC) were measured at day 10 or 15 post infection. RNO inhibited RSV infection of WD-HBE, prevented the loss of ciliated cells and markers, reduced the increase of MUC5AC and CLCA1 and inhibited the increase of IL-13, IL-6, IL-8, TNFα and ICAM-1. Additionally RNO reversed the reduction of Nrf2, HO-1 and GPx mRNA levels and consequently restored the TAC and reduced the H2O2 formation. RNO inhibits RSV infection of WD-HBE cultures and mitigates the cytopathological changes associated to this virus.
Collapse
|