1
|
Pickles RJ, Chen G, Randell SH. Enhanced susceptibility of pediatric airway epithelium to respiratory syncytial virus infection. J Clin Invest 2024; 134:e185689. [PMID: 39484717 PMCID: PMC11527439 DOI: 10.1172/jci185689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024] Open
Abstract
Immature innate and adaptive immunity and vulnerability of narrower airways to obstruction increase the susceptibility of infants to severe respiratory syncytial virus (RSV) disease. In this issue of the JCI, Zhao et al. illustrated greater intrinsic susceptibility of pediatric versus adult airway epithelial cells to RSV-induced cytopathology. Using precision cut lung slices (PCLS) and air-liquid interface (ALI) airway epithelial cell cultures, the authors showed that impaired STAT3 activation in RSV-infected pediatric multiciliated cells increased cell apoptosis and viral shedding, which enhanced the spread of infection. Bolstering STAT3 activation and treatment of neonatal mice with apoptosis inhibitors suppressed virus spread, suggesting that enhancing STAT3 activation may provide therapeutic benefit.
Collapse
Affiliation(s)
| | - Gang Chen
- Marsico Lung Institute
- Department of Pediatrics
| | - Scott H. Randell
- Marsico Lung Institute
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
2
|
Savino F, Dini M, Clemente A, Calvi C, Pau A, Galliano I, Gambarino S, Bergallo M. Nasopharyngeal and Peripheral Blood Type II Interferon Signature Evaluation in Infants during Respiratory Syncytial Virus Infection. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:259. [PMID: 38399546 PMCID: PMC10890591 DOI: 10.3390/medicina60020259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/22/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024]
Abstract
Background and Objectives: In this study, we applied one-step real time rt-PCR technology type II INF signature to blood and nasopharyngeal (NPS) swabs of acute early recovery children < 1 years hospitalized for bronchiolitis with laboratory-confirmed RSV infection. Materials and Methods: A prospective observational case-control study was conducted in 2021-2022. The study took place in Children Hospital "Regina Margherita", Torino Italy. The study included 66 infants, of which 30 patients were hospitalized for bronchiolitis due to RSV infection and 36 age-matched controls. Inclusion criteria included a positive RSV test for infants with bronchiolitis. We collected peripheral blood and nasopharyngeal swabs for relative quantification of type II Interferon signature by One-Step Multiplex PCR real time. Results: IFN levels were downregulated in the peripheral blood of bronchiolitis patients; these data were not confirmed in the nasopharyngeal swab. There was no correlation between NPS and the type II IFN score in peripheral blood. Conclusions: our study shows for the first time that type II IFN score was significant reduced in peripheral blood of infants with bronchiolitis by RSV compared to age-matched healthy controls; in the NPS swab this resulted downregulation was not statistically significant and the type II IFN score in the NPS swab can be used as marker of resolution of infection or improvement of clinical conditions.
Collapse
Affiliation(s)
- Francesco Savino
- Early Infancy Special Care Unit, Regina Margherita Children Hospital, A.O.U. Città della Salute e della Scienza di Torino, 10126 Torino, Italy;
| | - Maddalena Dini
- Paediatric Laboratory, Department of Public Health and Pediatric Sciences, Medical School, University of Turin, 10136 Turin, Italy; (M.D.); (A.C.); (C.C.); (A.P.); (I.G.)
- BioMole srl, Via Quarello 15/A, 10135 Turin, Italy
| | - Anna Clemente
- Paediatric Laboratory, Department of Public Health and Pediatric Sciences, Medical School, University of Turin, 10136 Turin, Italy; (M.D.); (A.C.); (C.C.); (A.P.); (I.G.)
| | - Cristina Calvi
- Paediatric Laboratory, Department of Public Health and Pediatric Sciences, Medical School, University of Turin, 10136 Turin, Italy; (M.D.); (A.C.); (C.C.); (A.P.); (I.G.)
- Department of Pediatrics, Infectious Diseases Unit, Regina Margherita Children’s Hospital, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
| | - Anna Pau
- Paediatric Laboratory, Department of Public Health and Pediatric Sciences, Medical School, University of Turin, 10136 Turin, Italy; (M.D.); (A.C.); (C.C.); (A.P.); (I.G.)
| | - Ilaria Galliano
- Paediatric Laboratory, Department of Public Health and Pediatric Sciences, Medical School, University of Turin, 10136 Turin, Italy; (M.D.); (A.C.); (C.C.); (A.P.); (I.G.)
- Department of Pediatrics, Infectious Diseases Unit, Regina Margherita Children’s Hospital, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
| | - Stefano Gambarino
- Paediatric Laboratory, Department of Public Health and Pediatric Sciences, Medical School, University of Turin, 10136 Turin, Italy; (M.D.); (A.C.); (C.C.); (A.P.); (I.G.)
- BioMole srl, Via Quarello 15/A, 10135 Turin, Italy
| | - Massimiliano Bergallo
- Paediatric Laboratory, Department of Public Health and Pediatric Sciences, Medical School, University of Turin, 10136 Turin, Italy; (M.D.); (A.C.); (C.C.); (A.P.); (I.G.)
- BioMole srl, Via Quarello 15/A, 10135 Turin, Italy
- Department of Pediatrics, Infectious Diseases Unit, Regina Margherita Children’s Hospital, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
| |
Collapse
|
3
|
Córdova-Dávalos LE, Hernández-Mercado A, Barrón-García CB, Rojas-Martínez A, Jiménez M, Salinas E, Cervantes-García D. Impact of genetic polymorphisms related to innate immune response on respiratory syncytial virus infection in children. Virus Genes 2022; 58:501-514. [PMID: 36085536 PMCID: PMC9462631 DOI: 10.1007/s11262-022-01932-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022]
Abstract
Respiratory syncytial virus (RSV) causes lower respiratory tract infections and bronchiolitis, mainly affecting children under 2 years of age and immunocompromised patients. Currently, there are no available vaccines or efficient pharmacological treatments against RSV. In recent years, tremendous efforts have been directed to understand the pathological mechanisms of the disease and generate a vaccine against RSV. Although RSV is highly infectious, not all the patients who get infected develop bronchiolitis and severe disease. Through various sequencing studies, single nucleotide polymorphisms (SNPs) have been discovered in diverse receptors, cytokines, and transcriptional regulators with crucial role in the activation of the innate immune response, which is implicated in the susceptibility to develop or protect from severe forms of the infection. In this review, we highlighted how variations in the key genes affect the development of innate immune response against RSV. This data would provide crucial information about the mechanisms of viral infection, and in the future, could help in generation of new strategies for vaccine development or generation of the pharmacological treatments.
Collapse
Affiliation(s)
- Laura Elena Córdova-Dávalos
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México
| | - Alicia Hernández-Mercado
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México
| | - Claudia Berenice Barrón-García
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México
| | - Augusto Rojas-Martínez
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Av. Morones Prieto 3000 Pte, Los Doctores, 64710, Monterrey, Nuevo León, México
| | - Mariela Jiménez
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México
| | - Eva Salinas
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México.
| | - Daniel Cervantes-García
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México. .,Consejo Nacional de Ciencia y Tecnología, 03940, Ciudad de México, México.
| |
Collapse
|
4
|
Martín-Vicente M, Resino S, Martínez I. Early innate immune response triggered by the human respiratory syncytial virus and its regulation by ubiquitination/deubiquitination processes. J Biomed Sci 2022; 29:11. [PMID: 35152905 PMCID: PMC8841119 DOI: 10.1186/s12929-022-00793-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/28/2022] [Indexed: 12/25/2022] Open
Abstract
The human respiratory syncytial virus (HRSV) causes severe lower respiratory tract infections in infants and the elderly. An exuberant inadequate immune response is behind most of the pathology caused by the HRSV. The main targets of HRSV infection are the epithelial cells of the respiratory tract, where the immune response against the virus begins. This early innate immune response consists of the expression of hundreds of pro-inflammatory and anti-viral genes that stimulates subsequent innate and adaptive immunity. The early innate response in infected cells is mediated by intracellular signaling pathways composed of pattern recognition receptors (PRRs), adapters, kinases, and transcriptions factors. These pathways are tightly regulated by complex networks of post-translational modifications, including ubiquitination. Numerous ubiquitinases and deubiquitinases make these modifications reversible and highly dynamic. The intricate nature of the signaling pathways and their regulation offers the opportunity for fine-tuning the innate immune response against HRSV to control virus replication and immunopathology.
Collapse
Affiliation(s)
- María Martín-Vicente
- Unidad de Infección Viral E Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III (Campus Majadahonda), Carretera Majadahonda-Pozuelo, Km 2.2, 28220 Majadahonda, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Salvador Resino
- Unidad de Infección Viral E Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III (Campus Majadahonda), Carretera Majadahonda-Pozuelo, Km 2.2, 28220 Majadahonda, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Isidoro Martínez
- Unidad de Infección Viral E Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III (Campus Majadahonda), Carretera Majadahonda-Pozuelo, Km 2.2, 28220 Majadahonda, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Th17/IL-17 Axis Regulated by Airway Microbes Get Involved in the Development of Asthma. Curr Allergy Asthma Rep 2020; 20:11. [PMID: 32172346 DOI: 10.1007/s11882-020-00903-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Bronchial asthma is a common respiratory disease induced by immune imbalance, characterized by chronic non-specific airway inflammation and airway hyperresponsiveness (AHR). Many factors induce asthma, among which respiratory infection is the important cause. In this review, we discuss how respiratory microbes participate in the occurrence and progression of asthma via Th17/IL-17 axis. RECENT FINDINGS Pathogenesis of asthma has been considered as closely related to the imbalance in number and function of Th1/Th2 in the CD4+ T lymphocyte subsets. Recent studies have shown that Th17 cell and its secretory IL-17 also play an important role in AHR. Respiratory virus, bacteria, fungi, and other respiratory microbial infections can directly or indirectly induce the differentiation of Th17 cell and the production of related cytokines to induce AHR. Respiratory microbial infection can affect the TH17/IL-17A axis through a variety of mechanisms, thereby promoting the occurrence and development of asthma, and these specific mechanisms may provide new effective therapeutic ideas for asthma.
Collapse
|
6
|
Antunes KH, Becker A, Franceschina C, de Freitas DDN, Lape I, da Cunha MD, Leitão L, Rigo MM, Pinto LA, Stein RT, de Souza APD. Respiratory syncytial virus reduces STAT3 phosphorylation in human memory CD8 T cells stimulated with IL-21. Sci Rep 2019; 9:17766. [PMID: 31780735 PMCID: PMC6882881 DOI: 10.1038/s41598-019-54240-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 11/07/2019] [Indexed: 12/21/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a common cause of childhood lower respiratory tract infections. The recent failure of a vaccine candidate based on recombinant F protein underlines the urgent need to better understand the protective human memory immune response against RSV. Signal transducer and activator of transcription 3 (STAT3) protein is a transcription factor that promotes the maturation of the memory CD8 T cell response in cooperation with IL-10 and IL-21. However, the role of STAT3 in the memory CD8 T cell response during RSV infection remains to be elucidated. We found that in infants with bronchiolitis infected with RSV, the expression of STAT3 detected in nasal washes is reduced when compared to that in infants infected by other viruses. In vitro, RSV impairs STAT3 phosphorylation induced by IL-21 in purified human memory CD8 T cells. In addition, RSV decreases granzyme B production by memory CD8 T cells, reducing its cytotoxic activity against RSV-infected epithelial pulmonary cell lines. Together, these data indicate that RSV modulates the IL-21/STAT3 pathway in human memory CD8 T cells, and this could be a mechanism to be further explored to improve the memory response against the infection.
Collapse
Affiliation(s)
- Krist Helen Antunes
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - André Becker
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Caroline Franceschina
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Deise do Nascimento de Freitas
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Isadora Lape
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Mariana D'Ávila da Cunha
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Lidiane Leitão
- Laboratory of Respiratory Physiology, Infant Center, School of Medicine, PUCRS, Porto Alegre, Brazil
| | - Mauricio M Rigo
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Leonardo Araújo Pinto
- Laboratory of Respiratory Physiology, Infant Center, School of Medicine, PUCRS, Porto Alegre, Brazil
| | - Renato T Stein
- Laboratory of Respiratory Physiology, Infant Center, School of Medicine, PUCRS, Porto Alegre, Brazil
| | - Ana Paula Duarte de Souza
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil. .,School of Health and Life Sciences, PUCRS, Porto Alegre, Brazil.
| |
Collapse
|
7
|
Chang Z, Wang Y, Zhou X, Long JE. STAT3 roles in viral infection: antiviral or proviral? Future Virol 2018; 13:557-574. [PMID: 32201498 PMCID: PMC7079998 DOI: 10.2217/fvl-2018-0033] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a transcription factor which can be activated by cytokines, growth factor receptors, and nonreceptor-like tyrosine kinase. An activated STAT3 translocates into the nucleus and combines with DNA to regulate the expression of target genes involved in cell proliferation, differentiation, apoptosis and metastasis. Recent studies have shown that STAT3 plays important roles in viral infection and pathogenesis. STAT3 exhibits a proviral function in several viral infections, including those of HBV, HCV, HSV-1, varicella zoster virus, human CMV and measles virus. However, in some circumstances, STAT3 has an antiviral function in other viral infections, such as enterovirus 71, severe acute respiratory syndrome coronavirus and human metapneumovirus. This review summarizes the roles of STAT3 in viral infection and pathogenesis, and briefly discusses the molecular mechanisms involved in these processes.
Collapse
Affiliation(s)
- Zhangmei Chang
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Kunshan Center For Disease Control & Prevention, 458 Tongfengxi Road, Kunshan, Jiangsu, 215301, PR China.,Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Kunshan Center For Disease Control & Prevention, 458 Tongfengxi Road, Kunshan, Jiangsu, 215301, PR China
| | - Yan Wang
- Department of Medical Microbiology & Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan R., Shanghai 200032, PR China.,Department of Medical Microbiology & Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan R., Shanghai 200032, PR China
| | - Xin Zhou
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | - Jian-Er Long
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Department of Medical Microbiology & Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan R., Shanghai 200032, PR China.,Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Department of Medical Microbiology & Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan R., Shanghai 200032, PR China
| |
Collapse
|
8
|
Do LAH, Pellet J, van Doorn HR, Tran AT, Nguyen BH, Tran TTL, Tran QH, Vo QB, Tran Dac NA, Trinh HN, Nguyen TTH, Le Binh BT, Nguyen HMK, Nguyen MT, Thai QT, Vo TV, Ngo NQM, Dang TKH, Cao NH, Tran TV, Ho LV, De Meulder B, Auffray C, Hofstra JJ, Farrar J, Bryant JE, de Jong M, Hibberd ML. Host Transcription Profile in Nasal Epithelium and Whole Blood of Hospitalized Children Under 2 Years of Age With Respiratory Syncytial Virus Infection. J Infect Dis 2017; 217:134-146. [PMID: 29029245 PMCID: PMC5853303 DOI: 10.1093/infdis/jix519] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 09/18/2017] [Accepted: 09/25/2017] [Indexed: 12/29/2022] Open
Abstract
Background Most insights into the cascade of immune events after acute respiratory syncytial virus (RSV) infection have been obtained from animal experiments or in vitro models. Methods In this study, we investigated host gene expression profiles in nasopharyngeal (NP) swabs and whole blood samples during natural RSV and rhinovirus (hRV) infection (acute versus early recovery phase) in 83 hospitalized patients <2 years old with lower respiratory tract infections. Results Respiratory syncytial virus infection induced strong and persistent innate immune responses including interferon signaling and pathways related to chemokine/cytokine signaling in both compartments. Interferon-α/β, NOTCH1 signaling pathways and potential biomarkers HIST1H4E, IL7R, ISG15 in NP samples, or BCL6, HIST2H2AC, CCNA1 in blood are leading pathways and hub genes that were associated with both RSV load and severity. The observed RSV-induced gene expression patterns did not differ significantly in NP swab and blood specimens. In contrast, hRV infection did not as strongly induce expression of innate immunity pathways, and significant differences were observed between NP swab and blood specimens. Conclusions We conclude that RSV induced strong and persistent innate immune responses and that RSV severity may be related to development of T follicular helper cells and antiviral inflammatory sequelae derived from high activation of BCL6.
Collapse
Affiliation(s)
- Lien Anh Ha Do
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, in partnership with the Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Johann Pellet
- Murdoch Children’s Research Institute, Melbourne, Australia
| | - H Rogier van Doorn
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, in partnership with the Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | | | | | | | | | - Quoc Bao Vo
- Children Hospital 2, Ho Chi Minh City, Vietnam
| | | | | | | | | | | | | | | | - Thanh Vu Vo
- Children Hospital 1, Ho Chi Minh City, Vietnam
| | | | | | | | | | - Lu Viet Ho
- Children Hospital 2, Ho Chi Minh City, Vietnam
| | | | - Charles Auffray
- European Institute for Systems Biology and Medicine, Lyon, France
| | - Jorrit-Jan Hofstra
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Jeremy Farrar
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, in partnership with the Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Juliet E Bryant
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, in partnership with the Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Menno de Jong
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, in partnership with the Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Martin L Hibberd
- Genome Institute of Singapore
- London School of Hygiene & Tropical Medicine, United Kingdom
| |
Collapse
|
9
|
Park BK, Choi SH, Kim YE, Park S, Lee Y, Lee KW, Kwon HJ. Monoclonal Antibodies Against the Human Respiratory Syncytial Virus Obtained by Immunization with Epitope Peptides and CpG-DNA-liposome Complex. Monoclon Antib Immunodiagn Immunother 2016; 34:101-9. [PMID: 25897608 DOI: 10.1089/mab.2014.0089] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the major cause of pulmonary inflammation in infants, young children, and immunocompromised adults. However, the RSV vaccine is not yet available commercially. The RSV-F glycoprotein mediates virus-host cell fusion, leading to syncytial formation; therefore, the RSV-F glycoprotein has been a treatment target for prevention and therapy of RSV infection. To produce the RSV-F-protein epitope-specific monoclonal antibody (MAb), BALB/c mice were immunized with a complex consisting of epitope peptide and MB-ODN 4531(O), encapsulated in a phosphatidyl-β-oleoyl-γ-palmitoyl ethanolamine (DOPE):cholesterol hemisuccinate (CHEMS) complex (Lipoplex(O)). Using conventional hybridoma technology, we obtained two clones able to produce antibodies reactive to two B-cell epitopes of RSV-F protein. Each anti-RSV-F glycoprotein MAb efficiently binds to each epitope. The F7-1A9D10 clone showed specific binding with RSV-F protein. There was no specific protein detected by Western blot analysis using F9 epitope-specific anti-RSV-F glycoprotein MAb (clone F9-1A6C8). However, based on confocal-image analysis, the antibody from the F9-1A6C8 clone showed specific binding with RSV-F protein. It is important that further study on possible applications for passive immunotherapy against RSV infection, such as therapeutic antibody production, is carried out.
Collapse
Affiliation(s)
- Byoung Kwon Park
- 1 Department of Microbiology, Hallym University , Chuncheon, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
10
|
Lee Y, Lee YS, Cho SY, Kwon HJ. Perspective of Peptide Vaccine Composed of Epitope Peptide, CpG-DNA, and Liposome Complex Without Carriers. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 99:75-97. [PMID: 26067817 DOI: 10.1016/bs.apcsb.2015.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The magnitude and specificity of cell-mediated and humoral immunity are critically determined by peptide sequences; peptides corresponding to the B- or T-cell receptor epitopes are sufficient to induce an effective immune response if delivered properly. Therefore, studies on the screening and application of peptide-based epitopes have been done extensively for the development of therapeutic antibodies and prophylactic vaccines. However, the efficacy of immune response and antibody production by peptide-based immunization is too limited for human application at the present. To improve the efficacy of vaccines, researchers formulated adjuvants such as alum, water-in-oil emulsion, and Toll-like receptor agonists. They also employed liposomes as delivering vehicles to stimulate immune responses. Here, we review our recent studies providing a potent method of epitope screening and antibody production without conventional carriers. We adopted Lipoplex(O), comprising a natural phosphodiester bond CpG-DNA and a specific liposome complex, as an adjuvant. Lipoplex(O) induces potent stimulatory activity in humans as well as in mice, and immunization of mice with several peptides along with Lipoplex(O) without general carriers induces significant production of each peptide-specific IgG2a. Immunization of peptide vaccines against virus-associated antigens in mice has protective effects against the viral infection. A peptide vaccine against carcinoma-associated antigen and the peptide-specific monoclonal antibody has functional effects against cancer cells in mouse models. In conclusion, we improved the efficacy of peptide vaccines in mice. Our strategy can be applied in development of therapeutic antibodies or in defense against pandemic infectious diseases through rapid screening of potent B-cell epitopes.
Collapse
Affiliation(s)
- Younghee Lee
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, South Korea
| | - Young Seek Lee
- Division of Molecular and Life Sciences, College of Science and Technology, Hanyang University, Ansan, South Korea
| | - Soo Young Cho
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Research Institute for Veterinary Science BK21, Program for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Hyung-Joo Kwon
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, South Korea; Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon, South Korea.
| |
Collapse
|
11
|
Effect of epitope-CpG-DNA-liposome complex without carriers on vaccination of respiratory syncytial virus infection. ACTA ACUST UNITED AC 2014. [DOI: 10.1007/s13765-014-4215-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
12
|
Hirakawa S, Kojima T, Obata K, Okabayashi T, Yokota SI, Nomura K, Obonai T, Fuchimoto J, Himi T, Tsutsumi H, Sawada N. Marked induction of matrix metalloproteinase-10 by respiratory syncytial virus infection in human nasal epithelial cells. J Med Virol 2013; 85:2141-50. [PMID: 24009192 DOI: 10.1002/jmv.23718] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2013] [Indexed: 11/10/2022]
Abstract
Respiratory syncytial virus (RSV) is an important pathogen of bronchiolitis, asthma, and severe lower respiratory tract disease in infants and young children. Matrix metalloproteinases (MMPs) play key roles in viral infection, inflammation and remodeling of the airway. However, the roles and regulation of MMPs in human nasal epithelial cells (HNECs) after RSV infection remain unclear. To investigate the regulation of MMP induced after RSV infection in HNECs, an RSV-infected model of HNECs in vitro was used. It was found that mRNA of MMP-10 was markedly increased in HNECs after RSV infection, together with induction of mRNAs of MMP-1, -7, -9, and -19. The amount of MMP-10 released from HNECs was also increased in a time-dependent manner after RSV infection as was that of chemokine RANTES. The upregulation of MMP-10 in HNECs after RSV infection was prevented by inhibitors of NF-κB and pan-PKC with inhibition of RSV replication, whereas it was prevented by inhibitors of JAK/STAT, MAPK, and EGF receptors without inhibition of RSV replication. In lung tissue of an infant with severe RSV infection in which a few RSV antibody-positive macrophages were observed, MMP-10 was expressed at the apical side of the bronchial epithelial cells and alveolar epithelial cells. In conclusion, MMP-10 induced by RSV infection in HNECs is regulated via distinct signal transduction pathways with or without relation to RSV replication. MMP-10 may play an important role in the pathogenesis of RSV diseases and it has the potential to be a novel marker and therapeutic target for RSV infection.
Collapse
Affiliation(s)
- Satoshi Hirakawa
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan; Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Garofalo RP, Kolli D, Casola A. Respiratory syncytial virus infection: mechanisms of redox control and novel therapeutic opportunities. Antioxid Redox Signal 2013; 18:186-217. [PMID: 22799599 PMCID: PMC3513983 DOI: 10.1089/ars.2011.4307] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Respiratory syncytial virus (RSV) is one of the most important causes of upper and lower respiratory tract infections in infants and young children, for which no effective treatment is currently available. Although the mechanisms of RSV-induced airway disease remain incompletely defined, the lung inflammatory response is thought to play a central pathogenetic role. In the past few years, we and others have provided increasing evidence of a role of reactive oxygen species (ROS) as important regulators of RSV-induced cellular signaling leading to the expression of key proinflammatory mediators, such as cytokines and chemokines. In addition, RSV-induced oxidative stress, which results from an imbalance between ROS production and airway antioxidant defenses, due to a widespread inhibition of antioxidant enzyme expression, is likely to play a fundamental role in the pathogenesis of RSV-associated lung inflammatory disease, as demonstrated by a significant increase in markers of oxidative injury, which correlate with the severity of clinical illness, in children with RSV infection. Modulation of ROS production and oxidative stress therefore represents a potential novel pharmacological approach to ameliorate RSV-induced lung inflammation and its long-term consequences.
Collapse
Affiliation(s)
- Roberto P Garofalo
- Department of Pediatrics, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | | | | |
Collapse
|
14
|
Abstract
Respiratory syncytial virus (RSV) is responsible for significant morbidity and mortality, particularly in infants younger than 18 months and in the elderly. To date, there are few effective treatment options available to prevent or treat RSV infections. Attractive therapeutic strategies include targeting host epithelial adhesion molecules required for RSV infection, enhancing localized cell-mediated immunity, interfering with RSV viral gene expression and developing a multigene DNA vaccine. The most recent data supporting the advantages and limitations of each of these approaches are discussed in detail. Several promising strategies offer hope for safe and effective prophylaxis and treatment of RSV infection.
Collapse
|
15
|
Levitz R, Wattier R, Phillips P, Solomon A, Lawler J, Lazar I, Weibel C, Kahn JS. Induction of IL-6 and CCL5 (RANTES) in human respiratory epithelial (A549) cells by clinical isolates of respiratory syncytial virus is strain specific. Virol J 2012; 9:190. [PMID: 22962966 PMCID: PMC3463437 DOI: 10.1186/1743-422x-9-190] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 08/24/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is the major respiratory pathogen of infants and young children. During each seasonal epidemic, multiple strains of both subgroup A and B viruses circulate in the community. Like other RNA viruses, RSV genome replication is prone to errors that results in a heterogeneous population of viral strains some of which may possess differences in virulence. We sought to determine whether clinical isolates of RSV differ in their capacity to induce inflammatory cytokines IL-6 and CCL5 (previously known as RANTES [regulated upon activation, normal T-cell expressed and secreted protein]), which are known to be induced in vitro and in vivo in response to RSV, during infection of A549 cells. RESULTS Screening of subgroup A and B isolates revealed heterogeneity among strains to induce IL-6 and CCL5. We chose two subgroup B strains, New Haven (NH)1067 and NH1125, for further analysis because of their marked differences in cytokine inducing properties and because subgroup B strains, in general, are less genetically heterogeneous as compared to subgroup A strains. At 12 and 24 hours post infection RSV strains, NH1067 and NH1125 differed in their capacity to induce IL-6 by an order of magnitude or more. The concentrations of IL-6 and CCL5 were dependent on the dose of infectious virus and the concentration of these cytokines induced by NH1125 was greater than that of those induced by NH1067 when the multiplicity of infection of NH1067 used was as much as 10-fold higher than that of NH1125. The induction of IL-6 was dependent on viable virus as infection with UV-inactivated virus did not induce IL-6. The difference in IL-6 induction most likely could not be explained by differences in viral replication kinetics. The intracellular level of RSV RNA, as determined by quantitative RT-PCR, was indistinguishable between the 2 strains though the titer of progeny virus produced by NH1125 was greater than that produced by NH1067 at 16, 24 and 36 hours but essentially equal at 48 and 72 hours. Full genome sequencing of the 2 strains revealed 193 polymorphisms and 4 insertions in NH1067 when compared to NH1125 (2 single base insertions in non-coding regions and 2 duplications of 3 and 60 bases in the RSV G gene). Of the polymorphisms, 147 occurred in coding regions and only 30 resulted in amino acid changes in 7 of the RSV genes. CONCLUSIONS These data suggest that RSV strains may not be homogeneous with regard to pathogenesis or virulence. Identification of the genetic polymorphisms associated with variations in cytokine induction may lead to insights into RSV disease and to the development of effective antiviral agents and vaccines.
Collapse
Affiliation(s)
- Ruth Levitz
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Boyapalle S, Wong T, Garay J, Teng M, San Juan-Vergara H, Mohapatra S, Mohapatra S. Respiratory syncytial virus NS1 protein colocalizes with mitochondrial antiviral signaling protein MAVS following infection. PLoS One 2012; 7:e29386. [PMID: 22383950 PMCID: PMC3288005 DOI: 10.1371/journal.pone.0029386] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2011] [Accepted: 11/28/2011] [Indexed: 12/24/2022] Open
Abstract
Respiratory syncytial virus (RSV) nonstructural protein 1(NS1) attenuates type-I interferon (IFN) production during RSV infection; however the precise role of RSV NS1 protein in orchestrating the early host-virus interaction during infection is poorly understood. Since NS1 constitutes the first RSV gene transcribed and the production of IFN depends upon RLR (RIG-I-like receptor) signaling, we reasoned that NS1 may interfere with this signaling. Herein, we report that NS1 is localized to mitochondria and binds to mitochondrial antiviral signaling protein (MAVS). Live-cell imaging of rgRSV-infected A549 human epithelial cells showed that RSV replication and transcription occurs in proximity to mitochondria. NS1 localization to mitochondria was directly visualized by confocal microscopy using a cell-permeable chemical probe for His6-NS1. Further, NS1 colocalization with MAVS in A549 cells infected with RSV was shown by confocal laser microscopy and immuno-electron microscopy. NS1 protein is present in the mitochondrial fraction and co-immunoprecipitates with MAVS in total cell lysatesof A549 cells transfected with the plasmid pNS1-Flag. By immunoprecipitation with anti-RIG-I antibody, RSV NS1 was shown to associate with MAVS at an early stage of RSV infection, and to disrupt MAVS interaction with RIG-I (retinoic acid inducible gene) and the downstream IFN antiviral and inflammatory response. Together, these results demonstrate that NS1 binds to MAVS and that this binding inhibits the MAVS-RIG-I interaction required for IFN production.
Collapse
Affiliation(s)
- Sandhya Boyapalle
- James A Haley Veteran's Administration Hospital, Tampa, Florida, United States of America
- University of South Florida Nanomedicine Research Center,Tampa, Florida, United States of America
- Department of Internal Medicine, Division of Translational Medicine, Tampa, Florida, United States of America
| | - Terianne Wong
- University of South Florida Nanomedicine Research Center,Tampa, Florida, United States of America
- Department of Internal Medicine, Division of Translational Medicine, Tampa, Florida, United States of America
| | - Julio Garay
- University of South Florida Nanomedicine Research Center,Tampa, Florida, United States of America
- Department of Internal Medicine, Division of Translational Medicine, Tampa, Florida, United States of America
| | - Michael Teng
- Department of Allergy and Immunology, Tampa, Florida, United States of America
| | - Homero San Juan-Vergara
- Department of Internal Medicine, Division of Translational Medicine, Tampa, Florida, United States of America
- Departamento de Medicina, Fundación Universidad del Norte, Barranquilla, Colombia
| | - Subhra Mohapatra
- James A Haley Veteran's Administration Hospital, Tampa, Florida, United States of America
- University of South Florida Nanomedicine Research Center,Tampa, Florida, United States of America
- Department of Internal Medicine, Division of Translational Medicine, Tampa, Florida, United States of America
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, Florida, United States of America
| | - Shyam Mohapatra
- James A Haley Veteran's Administration Hospital, Tampa, Florida, United States of America
- University of South Florida Nanomedicine Research Center,Tampa, Florida, United States of America
- Department of Internal Medicine, Division of Translational Medicine, Tampa, Florida, United States of America
- * E-mail:
| |
Collapse
|
17
|
How can microarrays unlock asthma? J Allergy (Cairo) 2012; 2012:241314. [PMID: 22500180 PMCID: PMC3303677 DOI: 10.1155/2012/241314] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 09/30/2011] [Accepted: 10/12/2011] [Indexed: 01/10/2023] Open
Abstract
Asthma is a complex disease regulated by the interplay of a large number of underlying mechanisms which contribute to the overall pathology. Despite various breakthroughs identifying genes related to asthma, our understanding of the importance of the genetic background remains limited. Although current therapies for asthma are relatively effective, subpopulations of asthmatics do not respond to these regimens. By unlocking the role of these underlying mechanisms, a source of novel and more effective treatments may be identified. In the new age of high-throughput technologies, gene-expression microarrays provide a quick and effective method of identifying novel genes and pathways, which would be impossible to discover using an individual gene screening approach. In this review we follow the history of expression microarray technologies and describe their contributions to advancing our current knowledge and understanding of asthma pathology.
Collapse
|
18
|
Hinzey A, Alexander J, Corry J, Adams KM, Claggett AM, Traylor ZP, Davis IC, Webster Marketon JI. Respiratory syncytial virus represses glucocorticoid receptor-mediated gene activation. Endocrinology 2011; 152:483-94. [PMID: 21190962 PMCID: PMC3037158 DOI: 10.1210/en.2010-0774] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Respiratory syncytial virus (RSV) is a common cause of bronchiolitis in infants. Although antiinflammatory in nature, glucocorticoids have been shown to be ineffective in the treatment of RSV-induced bronchiolitis and wheezing. In addition, the effectiveness of glucocorticoids at inhibiting RSV-induced proinflammatory cytokine production in cell culture has been questioned. In this study, we have investigated the effect of RSV infection on glucocorticoid-induced gene activation in lung epithelium-derived cells. We show that RSV infection inhibits dexamethasone induction of three glucocorticoid receptor (GR)-regulated genes (glucocorticoid-inducible leucine zipper, FK506 binding protein, and MAPK phosphatase 1) in A549, BEAS-2B cells, and primary small airway epithelial cells. UV irradiation of the virus prevents this repression, suggesting that viral replication is required. RSV is known to activate the nuclear factor κB (NFκB) pathway, which is mutually antagonistic towards the GR pathway. However, specific inhibition of NFκB had no effect on the repression of GR-induced genes by RSV infection, indicating that RSV repression of GR is independent of NFκB. RSV infection of A549 cells does not alter GR protein levels or GR nuclear translocation but does reduce GR binding to the promoters of the glucocorticoid responsive genes analyzed in this study. Repression of GR by RSV infection may account for the apparent clinical ineffectiveness of glucocorticoids in RSV bronchiolitis therapy. In addition, this data adds to our previously published data suggesting that GR may be a general target for infectious agents. Identifying the mechanisms through which this suppression occurs may lead to the development of novel therapeutics.
Collapse
Affiliation(s)
- Adam Hinzey
- Division of Pulmonary, Department of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Gene expression differences in lungs of mice during secondary immune responses to respiratory syncytial virus infection. J Virol 2010; 84:9584-94. [PMID: 20592085 DOI: 10.1128/jvi.00302-10] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vaccine-induced immunity has been shown to alter the course of a respiratory syncytial virus (RSV) infection both in murine models and in humans. To elucidate which mechanisms underlie the effect of vaccine-induced immunity on the course of RSV infection, transcription profiles in the lungs of RSV-infected mice were examined by microarray analysis. Three models were used: RSV reinfection as a model for natural immunity, RSV challenge after formalin-inactivated RSV vaccination as a model for vaccine-enhanced disease, and RSV challenge following vaccination with recombinant RSV virus lacking the G gene (DeltaG-RSV) as a model for vaccine-induced immunity. Gene transcription profiles, histopathology, and viral loads were analyzed at 1, 2, and 5 days after RSV challenge. On the first 2 days after challenge, all mice displayed an expression pattern in the lung similar of that found in primary infection, showing a strong innate immune response. On day 5 after RSV reinfection or after challenge following DeltaG-RSV vaccination, the innate immune response was waning. In contrast, in mice with vaccine-enhanced disease, the innate immune response 5 days after RSV challenge was still present even though viral replication was diminished. In addition, only in this group was Th2 gene expression induced. These findings support a hypothesis that vaccine-enhanced disease is mediated by prolonged innate immune responses and Th2 polarization in the absence of viral replication.
Collapse
|
20
|
Identification of gene biomarkers for respiratory syncytial virus infection in a bronchial epithelial cell line. Genomic Med 2009; 2:113-25. [PMID: 19459069 DOI: 10.1007/s11568-009-9080-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 04/14/2009] [Accepted: 04/24/2009] [Indexed: 12/26/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection involves complex virus-host interplay. In this study, we analyzed gene expression in RSV-infected BEAS-2B cells to discover novel signaling pathways and biomarkers. We hybridized RNAs from RSV- or vehicle-treated BEAS-2B to Affymetrix HU133 plus 2.0 microarrays (n = 4). At 4 and 24 h post-infection, 277 and 900 genes (RSV/control ratio >/=2.0 or </=0.5), and 1 and 12 pathways respectively were significantly altered. Twenty-three and 92 genes at 4 and 24 h respectively matched respiratory disease biomarkers with ARG2 flagged at 24 h and SCNN1G, EPB41L4B, CSF1, PTEN, TUBB1 and ESR2 at both time points. Hierachical clustering showed a cluster containing ARG2 and IL8. In human bronchial epithelial cells, RSV upregulated arginase II protein. Knockdown of ARG2 increased RSV-induced IL-8, LDH and histone release. With microarray, we identified novel proximal airway epithelial cell genes that may be tested in the sputum samples as biomarkers of RSV infection.
Collapse
|
21
|
Respiratory syncytial virus activates innate immunity through Toll-like receptor 2. J Virol 2008; 83:1492-500. [PMID: 19019963 DOI: 10.1128/jvi.00671-08] [Citation(s) in RCA: 206] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a common cause of infection that is associated with a range of respiratory illnesses, from common cold-like symptoms to serious lower respiratory tract illnesses such as pneumonia and bronchiolitis. RSV is the single most important cause of serious lower respiratory tract illness in children <1 year of age. Host innate and acquired immune responses activated following RSV infection have been suspected to contribute to RSV disease. Toll-like receptors (TLRs) activate innate and acquired immunity and are candidates for playing key roles in the host immune response to RSV. Leukocytes express TLRs, including TLR2, TLR6, TLR3, TLR4, and TLR7, that can interact with RSV and promote immune responses following infection. Using knockout mice, we have demonstrated that TLR2 and TLR6 signaling in leukocytes can activate innate immunity against RSV by promoting tumor necrosis factor alpha, interleukin-6, CCL2 (monocyte chemoattractant protein 1), and CCL5 (RANTES). As previously noted, TLR4 also contributes to cytokine activation (L. M. Haynes, D. D. Moore, E. A. Kurt-Jones, R. W. Finberg, L. J. Anderson, and R. A. Tripp, J. Virol. 75:10730-10737, 2001, and E. A. Kurt-Jones, L. Popova, L. Kwinn, L. M. Haynes, L. P. Jones, R. A. Tripp, E. E. Walsh, M. W. Freeman, D. T. Golenbock, L. J. Anderson, and R. W. Finberg, Nat. Immunol. 1:398-401, 2000). Furthermore, we demonstrated that signals generated following TLR2 and TLR6 activation were important for controlling viral replication in vivo. Additionally, TLR2 interactions with RSV promoted neutrophil migration and dendritic cell activation within the lung. Collectively, these studies indicate that TLR2 is involved in RSV recognition and subsequent innate immune activation.
Collapse
|
22
|
Mohapatra SS, Boyapalle S. Epidemiologic, experimental, and clinical links between respiratory syncytial virus infection and asthma. Clin Microbiol Rev 2008; 21:495-504. [PMID: 18625684 PMCID: PMC2493089 DOI: 10.1128/cmr.00054-07] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Virtually all children experience respiratory syncytial virus (RSV) infection at least once during the first 2 years of life, but only a few develop bronchiolitis and more severe disease requiring hospitalization, usually in the first 6 months of life. Children who recover from RSV-induced bronchiolitis are at increased risk for the development of recurrent wheeze and asthma in later childhood. Recent studies suggest that there is an association between RSV-induced bronchiolitis and asthma within the first decade of life but that this association is not significant after age 13. Despite the considerable progress made in our understanding of several aspects of respiratory viral infections, further work needs to be done to clarify the molecular mechanisms of early interactions between virus and host cell and the role of host gene products in the infection process. This review provides a critical appraisal of the literature in epidemiology and experimental research which links RSV infection to asthma. Studies to date demonstrate that there is a significant association between RSV infection and childhood asthma and that preventing severe primary RSV infections can decrease the risk of childhood asthma.
Collapse
Affiliation(s)
- Shyam S Mohapatra
- Department of Internal Medicine, Division of Allergy and Immunology, University of South Florida College of Medicine, James A Haley Veterans Hospital, 12901 Bruce B Downs Blvd, Tampa, Florida 33612, USA.
| | | |
Collapse
|
23
|
Amino acid 226 in the hemagglutinin of H4N6 influenza virus determines binding affinity for alpha2,6-linked sialic acid and infectivity levels in primary swine and human respiratory epithelial cells. J Virol 2008; 82:8204-9. [PMID: 18550676 DOI: 10.1128/jvi.00718-08] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Avian lineage H4N6 influenza viruses previously isolated from pigs differ at hemagglutinin amino acids 226 and 228 from H4 subtype viruses isolated from birds. Using a parental H4N6 swine isolate and hemagglutinin mutant viruses (at residues 226 and/or 228), we determined that viruses which contain L226 had a higher affinity for sialic acid alpha2,6 galactose (SAalpha2,6Gal) and a higher infectivity level for primary swine and human respiratory epithelial cells, whereas viruses which contain Q226 had lower SAalpha2,6Gal affinity and lower infectivity levels for both types of cells. Using specific neuraminidases, we found that irrespective of their relative binding preferences, all of the influenza viruses examined utilized SAalpha2,6Gal to infect swine and human cells.
Collapse
|
24
|
Identification of amino acids in the HA of H3 influenza viruses that determine infectivity levels in primary swine respiratory epithelial cells. Virus Res 2008; 133:269-79. [DOI: 10.1016/j.virusres.2008.01.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Revised: 01/25/2008] [Accepted: 01/27/2008] [Indexed: 11/20/2022]
|
25
|
Novershtern N, Itzhaki Z, Manor O, Friedman N, Kaminski N. A functional and regulatory map of asthma. Am J Respir Cell Mol Biol 2007; 38:324-36. [PMID: 17921359 DOI: 10.1165/rcmb.2007-0151oc] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The prevalence and morbidity of asthma, a chronic inflammatory airway disease, is increasing. Animal models provide a meaningful but limited view of the mechanisms of asthma in humans. A systems-level view of asthma that integrates multiple levels of molecular and functional information is needed. For this, we compiled a gene expression compendium from five publicly available mouse microarray datasets and a gene knowledge base of 4,305 gene annotation sets. Using this collection we generated a high-level map of the functional themes that characterize animal models of asthma, dominated by innate and adaptive immune response. We used Module Networks analysis to identify co-regulated gene modules. The resulting modules reflect four distinct responses to treatment, including early response, general induction, repression, and IL-13-dependent response. One module with a persistent induction in response to treatment is mainly composed of genes with suggested roles in asthma, suggesting a similar role for other module members. Analysis of IL-13-dependent response using protein interaction networks highlights a role for TGF-beta1 as a key regulator of asthma. Our analysis demonstrates the discovery potential of systems-level approaches and provides a framework for applying such approaches to asthma.
Collapse
Affiliation(s)
- Noa Novershtern
- School of Computer Science and Engineering, Faculty of Medicine, The Hebrew University, Jerusalem
| | | | | | | | | |
Collapse
|
26
|
Tran KC, He B, Teng MN. Replacement of the respiratory syncytial virus nonstructural proteins NS1 and NS2 by the V protein of parainfluenza virus 5. Virology 2007; 368:73-82. [PMID: 17632199 PMCID: PMC2078599 DOI: 10.1016/j.virol.2007.06.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Revised: 04/09/2007] [Accepted: 06/13/2007] [Indexed: 01/18/2023]
Abstract
Paramyxoviruses have been shown to produce proteins that inhibit interferon production and signaling. For human respiratory syncytial virus (RSV), the nonstructural NS1 and NS2 proteins have been shown to have interferon antagonist activity through an unknown mechanism. To understand further the functions of NS1 and NS2, we generated recombinant RSV in which both NS1 and NS2 were replaced by the PIV5 V protein, which has well-characterized IFN antagonist activities (DeltaNS1/2-V). Expression of V was able to partially inhibit IFN responses in DeltaNS1/2-V-infected cells. In addition, the replication kinetics of DeltaNS1/2-V were intermediate between DeltaNS1/2 and wild-type (rA2) in A549 cells. However, expression of V did not affect the ability of DeltaNS1/2-V to activate IRF3 nuclear translocation and IFNbeta transcription. These data indicate that V was able to replace some of the IFN inhibitory functions of the RSV NS1 and NS2 proteins, but also that NS1 and NS2 have functions in viral replication beyond IFN antagonism.
Collapse
Affiliation(s)
- Kim C. Tran
- Department of Biochemistry and Molecular Biology, Center for Molecular Immunology and Infectious Diseases, Pennsylvania State University, University Park, PA, 16802
| | - Biao He
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Diseases, Pennsylvania State University, University Park, PA, 16802
| | - Michael N. Teng
- Department of Biochemistry and Molecular Biology, Center for Molecular Immunology and Infectious Diseases, Pennsylvania State University, University Park, PA, 16802
- *Corresponding author: Department of Biochemistry and Molecular Biology, Pennsylvania State University, 406 South Frear, University Park, PA 16802, Tel: (814) 863-6996, FAX: (814) 863-7024,
| |
Collapse
|
27
|
Arnold R, König W. Peroxisome proliferator-activated receptor-γ agonists inhibit the replication of respiratory syncytial virus (RSV) in human lung epithelial cells. Virology 2006; 350:335-46. [PMID: 16616290 DOI: 10.1016/j.virol.2006.03.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2005] [Revised: 02/13/2006] [Accepted: 03/09/2006] [Indexed: 11/16/2022]
Abstract
We have previously shown that peroxisome proliferator-activated receptor-gamma (PPARgamma) agonists inhibited the inflammatory response of RSV-infected human lung epithelial cells. In this study, we supply evidence that specific PPARgamma agonists (15d-PGJ2, ciglitazone, troglitazone, Fmoc-Leu) efficiently blocked the RSV-induced cytotoxicity and development of syncytia in tissue culture (A549, HEp-2). All PPARgamma agonists under study markedly inhibited the cell surface expression of the viral G and F protein on RSV-infected A549 cells. This was paralleled by a reduced cellular amount of N protein-encoding mRNA determined by real-time RT-PCR. Concomitantly, a reduced release of infectious progeny virus into the cell supernatants of human lung epithelial cells (A549, normal human bronchial epithelial cells (NHBE)) was observed. Similar results were obtained regardless whether PPARgamma agonists were added prior to RSV infection or thereafter, suggesting that the agonists inhibited viral gene expression and not the primary adhesion or fusion process.
Collapse
Affiliation(s)
- Ralf Arnold
- Institute of Medical Microbiology, Otto-von-Guericke-University, Leipzigerstr. 44, 39120 Magdeburg, Germany.
| | | |
Collapse
|
28
|
The Role of Phosphoinositide 3-Kinase-Akt Signaling in Virus Infection. APOPTOSIS, CELL SIGNALING, AND HUMAN DISEASES 2006. [PMCID: PMC7120950 DOI: 10.1007/978-1-59745-199-4_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
29
|
Verma S, Ziegler K, Ananthula P, Co JKG, Frisque RJ, Yanagihara R, Nerurkar VR. JC virus induces altered patterns of cellular gene expression: interferon-inducible genes as major transcriptional targets. Virology 2005; 345:457-67. [PMID: 16297951 DOI: 10.1016/j.virol.2005.10.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2005] [Revised: 09/19/2005] [Accepted: 10/12/2005] [Indexed: 11/16/2022]
Abstract
Human polyomavirus JC (JCV) infects 80% of the population worldwide. Primary infection, typically occurring during childhood, is asymptomatic in immunocompetent individuals and results in lifelong latency and persistent infection. However, among the severely immunocompromised, JCV may cause a fatal demyelinating disease, progressive multifocal leukoencephalopathy (PML). Virus-host interactions influencing persistence and pathogenicity are not well understood, although significant regulation of JCV activity is thought to occur at the level of transcription. Regulation of the JCV early and late promoters during the lytic cycle is a complex event that requires participation of both viral and cellular factors. We have used cDNA microarray technology to analyze global alterations in gene expression in JCV-permissive primary human fetal glial cells (PHFG). Expression of more than 400 cellular genes was altered, including many that influence cell proliferation, cell communication and interferon (IFN)-mediated host defense responses. Genes in the latter category included signal transducer and activator of transcription 1 (STAT1), interferon stimulating gene 56 (ISG56), myxovirus resistance 1 (MxA), 2'5'-oligoadenylate synthetase (OAS), and cig5. The expression of these genes was further confirmed in JCV-infected PHFG cells and the human glioblastoma cell line U87MG to ensure the specificity of JCV in inducing this strong antiviral response. Results obtained by real-time RT-PCR and Western blot analyses supported the microarray data and provide temporal information related to virus-induced changes in the IFN response pathway. Our data indicate that the induction of an antiviral response may be one of the cellular factors regulating/controlling JCV replication in immunocompetent hosts and therefore constraining the development of PML.
Collapse
Affiliation(s)
- Saguna Verma
- Retrovirology Research Laboratory, Department of Tropical Medicine and Medical Microbiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, 96822, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Alcorn JL, Stark JM, Chiappetta CL, Jenkins G, Colasurdo GN. Effects of RSV infection on pulmonary surfactant protein SP-A in cultured human type II cells: contrasting consequences on SP-A mRNA and protein. Am J Physiol Lung Cell Mol Physiol 2005; 289:L1113-22. [PMID: 16055477 DOI: 10.1152/ajplung.00436.2004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the most important cause of serious lower respiratory illness in infants and children. Surfactant proteins A (SP-A) and D (SP-D) play critical roles in lung defense against RSV infections. Alterations in surfactant protein homeostasis in the lung may result from changes in production, metabolism, or uptake of the protein within the lung. We hypothesized that RSV infection of the type II cell, the primary source of surfactant protein, may alter surfactant protein gene expression. Human type II cells grown in primary culture possess lamellar bodies (a type II cell-specific organelle) and the ability to express surfactant protein mRNA. These cells were infected with RSV (by morphology and antibody binding). Surfactant protein mRNA levels determined by quantitative RT-PCR indicated a marked increase in SP-A mRNA levels (3-fold) 24 h after RSV exposure, whereas SP-D mRNA levels were unaffected. In contrast to mRNA levels, total SP-A protein levels (determined by Western blot analysis) were decreased 40% after RSV infection. The percentage of secreted SP-A was 43% of the total SP-A in the RSV-infected cells, whereas the percentage of secreted SP-A was 61% of the total SP-A in the uninfected cells. These changes in SP-A transcript levels and protein secretion in cultured human cells were recapitulated in RSV-infected mouse lung. Our findings suggest that type II cells are potentially important targets of RSV lower respiratory infection and that alterations in surfactant protein gene expression and SP-A protein homeostasis in the lung may arise via direct effects of RSV.
Collapse
Affiliation(s)
- Joseph L Alcorn
- Dept. of Pediatrics, The University of Texas-Houston Medical School, 6431 Fannin, Suite 3.222, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
31
|
McLaren PJ, Mayne M, Rosser S, Moffatt T, Becker KG, Plummer FA, Fowke KR. Antigen-specific gene expression profiles of peripheral blood mononuclear cells do not reflect those of T-lymphocyte subsets. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2005; 11:977-82. [PMID: 15358662 PMCID: PMC515274 DOI: 10.1128/cdli.11.5.977-982.2004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Advances in microarray technology have allowed for the monitoring of thousands of genes simultaneously. This technology is of particular interest to immunologists studying infectious diseases, because it provides tremendous potential for investigating host-pathogen interactions at the level of immune gene expression. To date, many studies have focused either on cell lines, where the physiological relevance is questionable, or on mixed cell populations, where the contributions of individual subpopulations are unknown. In the present study, we perform an intrasubject comparison of antigen-stimulated immune gene expression profiles between a mixed population of peripheral blood mononuclear cells (PBMC) and the two predominant cell types found in PBMC, CD4+ and CD8+ T lymphocytes. We show that the microarray profiles of CD4+ and CD8+ T lymphocytes differ from each other as well as from that of the mixed cell population. The independence of the gene expression profiles of different cell types is demonstrated with a ubiquitous antigen (Candida albicans) as well as with a disease-specific antigen (human immunodeficiency virus p24). This study has important implications for microarray studies of host immunity and underscores the importance of profiling the expression of specific cell types.
Collapse
Affiliation(s)
- Paul J McLaren
- Department of Medical Microbiology, University of Manitoba, Winnipeg, R3E 0W3, Canada
| | | | | | | | | | | | | |
Collapse
|
32
|
San-Juan-Vergara H, Peeples ME, Lockey RF, Mohapatra SS. Protein kinase C-alpha activity is required for respiratory syncytial virus fusion to human bronchial epithelial cells. J Virol 2004; 78:13717-26. [PMID: 15564481 PMCID: PMC533893 DOI: 10.1128/jvi.78.24.13717-13726.2004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection activates protein kinase C (PKC), but the precise PKC isoform(s) involved and its role(s) remain to be elucidated. On the basis of the activation kinetics of different signaling pathways and the effect of various PKC inhibitors, it was reasoned that PKC activation is important in the early stages of RSV infection, especially RSV fusion and/or replication. Herein, the role of PKC-alpha during the early stages of RSV infection in normal human bronchial epithelial cells is determined. The results show that the blocking of PKC-alpha activation by classical inhibitors, pseudosubstrate peptides, or the overexpression of dominant-negative mutants of PKC-alpha in these cells leads to significantly decreased RSV infection. RSV induces phosphorylation, activation, and cytoplasm-to-membrane translocation of PKC-alpha. Also, PKC-alpha colocalizes with virus particles and is required for RSV fusion to the cell membrane. Thus, PKC-alpha could provide a new pharmacological target for controlling RSV infection.
Collapse
Affiliation(s)
- Homero San-Juan-Vergara
- The Joy McCann Culverhouse Airways Disease Research Center, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | | | | | | |
Collapse
|
33
|
Ramaswamy M, Shi L, Monick MM, Hunninghake GW, Look DC. Specific Inhibition of Type I Interferon Signal Transduction by Respiratory Syncytial Virus. Am J Respir Cell Mol Biol 2004; 30:893-900. [PMID: 14722224 DOI: 10.1165/rcmb.2003-0410oc] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Respiratory viruses often express mechanisms to resist host antiviral systems, but the biochemical basis for evasion of interferon effects by respiratory syncytial virus (RSV) is poorly defined. In this study, we identified RSV effects on interferon (IFN)-dependent signal transduction and gene expression in human airway epithelial cells. Initial experiments demonstrated inhibition of antiviral gene expression induced by IFN-alpha and IFN-beta, but not IFN-gamma, in epithelial cells infected with RSV. Selective viral effects on type I IFN-dependent signaling were confirmed when we observed impaired type I, but not type II, IFN-induced activation of the transcription factor Stat1 in RSV-infected cells. RSV infection of airway epithelial cells resulted in decreased Stat2 expression and function with preservation of upstream signaling events, providing a molecular mechanism for viral inhibition of the type I IFN JAK-STAT pathway. Furthermore, nonspecific pharmacologic inhibition of proteasome function in RSV-infected cells restored Stat2 levels and IFN-dependent activation of Stat1. The results indicate that RSV acts on epithelial cells in the airway to directly modulate the type I IFN JAK-STAT pathway, and this effect is likely mediated though proteasome-dependent degradation of Stat2. Decreased antiviral gene expression in RSV-infected airway epithelial cells may allow RSV replication and establishment of a productive viral infection through subversion of IFN-dependent immunity.
Collapse
Affiliation(s)
- Murali Ramaswamy
- University of Iowa Roy J. and Lucille A. Carver College of Medicine, Department of Internal Medicine, 200 Hawkins Drive, C33-GH, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
34
|
Abstract
Respiratory syncytial virus (RSV) is recognized as the most important cause of serious lower respiratory tract illness in infants and young children worldwide causing repeat infections throughout life with serious complications occurring in the elderly and immune compromised patient. The level of disease pathogenesis associated with RSV infection is balanced between virus elimination and the nature of the immune response to infection. The innate and adaptive immune responses to RSV infection are not fully elucidated; however, significant progress has been made in understanding the virus-host relationship and mechanisms associated with disease pathogenesis. This review summarizes important aspects of these findings, and provides current perspective on processes that may contribute to RSV disease pathogenesis.
Collapse
Affiliation(s)
- Ralph A Tripp
- Division of Viral and Rickettsial Diseases, Viral and Enteric Virus Branch, Centers for Disease Control and Prevention, Atlanta, Georgia.
| |
Collapse
|
35
|
Kong X, San Juan H, Behera A, Peeples ME, Wu J, Lockey RF, Mohapatra SS. ERK-1/2 activity is required for efficient RSV infection. FEBS Lett 2004; 559:33-8. [PMID: 14960303 DOI: 10.1016/s0014-5793(04)00002-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2003] [Revised: 11/26/2003] [Accepted: 12/03/2003] [Indexed: 10/26/2022]
Abstract
Respiratory syncytial virus (RSV) infection up-regulates the expression of genes encoding proinflammatory mediators in bronchial epithelial cells. However, the specific signaling events immediately following RSV exposure are poorly understood. Herein, we report that RSV attachment to A549 cells activates both ERK-1 and ERK-2 pathways within 5 min. Inhibition of ERK pathways significantly decreases RSV infection of these cells compared to controls. These results demonstrate that the activation of the ERK-1/2 is required in RSV-induced early gene expression.
Collapse
Affiliation(s)
- Xiaoyuan Kong
- The Joy McCann Culverhouse Airways Disease Research Center, Division of Allergy and Immunology, Department of Internal Medicine, MDC-19, 12901 Bruce B. Downs Blvd, Tampa, FL 33612, USA
| | | | | | | | | | | | | |
Collapse
|