1
|
Duan Y, Ou Y, Li J, Gan X, Cao J. Serum cytokine levels in children with community-acquired pneumonia caused by different respiratory pathogens. Ital J Pediatr 2025; 51:147. [PMID: 40394688 PMCID: PMC12093880 DOI: 10.1186/s13052-025-02012-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 05/11/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Our study aims to investigate the levels of serum cytokines in children with community-acquired pneumonia (CAP) caused by different respiratory pathogens, and evaluate the predictive value of cytokines levels for severe pneumonia. METHODS A retrospective study was conducted on the clinical data of children hospitalized with CAP. According to the pathogens, patients were divided into the M. pneumoniae group, Adenovirus group, respiratory syncytial virus group, H.influenzae group, and S.pneumoniae group. RESULTS The M. pneumoniae group was higher than RSV group in the level of serum pro-inflammatory cytokines include IL-2, IL-6, IL-17 A, and IFN-γ. But M. pneumoniae group was higher than Adenovirus group only in IL-6. M. pneumoniae group was higher than H. influenzae group and S. pneumoniae group in IL-17 A, IFN-γ. as primary anti-inflammatory cytokine, IL-10 was lower in the M. pneumoniae group compared with Adenovirus and RSV groups. IL-6 was higher in S. pneumoniae group than RSV group. IFN-γ was lower in H. influenzae group than Adenovirus group and RSV group. IL-10 was higher in RSV group than H. influenzae group. IL-6 was higher in Adenovirus group than RSV group. In M. pneumoniae group and H. influenzae group, the levels of IL-6, IL-10, and IFN-γ were significantly higher in the severe pneumonia subgroup compared with the non-severe pneumonia subgroup (P < 0.05). CONCLUSIONS Compared with other groups, M. pneumoniae group was higher in the level of serum proinflammatory cytokines. Additionally, the levels of IL-6, IL-10, and IFN-γ can be used as predictors of severe pneumonia caused by M. pneumoniae and H. influenzae.
Collapse
Affiliation(s)
- Yuanhui Duan
- Department of Medical General Ward Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Yuexu Ou
- Department of Medical General Ward Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Jieling Li
- Department of Medical General Ward Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Xiaoming Gan
- Department of Medical General Ward Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Jie Cao
- Department of Medical General Ward Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China.
| |
Collapse
|
2
|
Xu L, He R, Ye X, Wang Y, Hui S, Li H, Chen H, Huang P. Leveraging transcriptome-wide association studies identifies the relationship between upper respiratory flora and cell type-specific gene expression in severe respiratory disease. PLoS One 2025; 20:e0322864. [PMID: 40343915 PMCID: PMC12063895 DOI: 10.1371/journal.pone.0322864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/30/2025] [Indexed: 05/11/2025] Open
Abstract
ObjectivesThe upper respiratory tract flora may influence host immunity and modulate susceptibility to viral respiratory infections. This study aimed to investigate the associations between upper respiratory tract flora and immune cells in severe ILI, identify specific microbial taxa and immune response pathways contributing to disease severity, and elucidate how flora influences ILI progression by modulating immune cell functions.MethodsHeritability of GWAS summary data was estimated using LDSC (v1.0.1). Gene-level genetic associations were analyzed with MAGMA. scRNA-seq data were integrated with genetic association data using scDRS. FUSION was used to construct cell type-specific expression quantitative trait locus models based on genotypes and scRNA-seq data from the onek1k project, which were combined with flora abundance-related GWAS data for a transcriptome-wide association study.ResultsFrom the LDSC analysis, data from 1195 severe ILI-associated GWASs with upper respiratory flora(h2 > 0.1) were included in subsequent analysis. TWAS identified 19 significant association pairs (Padj < 0.05), and 1226 differentially expressed genes between mild and severe ILI patients (Padj < 0.05 and | log2FC|>0.25). Functional enrichment analyses using GO, KEGG, and Reactome databases revealed that immune cells,such as CD4 + T effector memory cells, cDCs, NK cells, were enriched in multiple biological processes or pathways.ConclusionsThis study identified associations between severe ILI-related upper respiratory tract flora and cell type-specific gene expression, potentially explaining how differential flora influences ILI progression. CD16 + monocytes exhibited the most differentially expressed genes, followed by proliferating cells and cDCs, highlighting the significant role of immune cell-enriched pathways in ILI progression.
Collapse
Affiliation(s)
- Lei Xu
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Ran He
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Xiangyu Ye
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Yifan Wang
- Department of Infectious Disease, Jurong Hospital Affiliated to Jiangsu University, Jurong, Jiangsu, China
| | - Shirong Hui
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Haochang Li
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Hongbo Chen
- Department of Infectious Disease, Jurong Hospital Affiliated to Jiangsu University, Jurong, Jiangsu, China
| | - Peng Huang
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Reagin KL, Oliva KE, Hansen MR, Slade CD, Watford WT, Klonowski KD. Regulation of respiratory CD8+ T-cell immunity by suppressive monocyte-like dendritic cells (MCs). JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:977-994. [PMID: 40163680 PMCID: PMC12123217 DOI: 10.1093/jimmun/vkae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/12/2024] [Indexed: 04/02/2025]
Abstract
Active immune suppression can mediate the balance between protective cellular immunity and harmful immunopathology. This suppression can occur locally, at an infection site, or in regional draining lymph nodes (dLNs). Immune regulation is of particular importance in sites such as the lung where aberrant immunopathology can result in loss of tissue function and respiratory failure. We have recently identified a novel population of CD11b+CD103+CCR2+ monocyte-like dendritic cells (MCs) which directly suppress CD8+ T-cell proliferation in vitro. Respiratory infection of mice with RNA viruses recruits these MCs either exclusively to the dLN (after vesicular stomatitis virus infection) or both the dLN and site of viral replication (after influenza infection). Here we show that depletion of MCs from the dLN of mice using CCR2-DTR bone marrow chimeras results in enhanced respiratory CD8+ T-cell responses and lung tissue-resident memory cell (TRM) formation which correlated with enhanced antiviral responses upon heterologous VSV challenge. Conversely, depletion of MCs from both the dLN and respiratory tract following influenza infection results in enhanced respiratory CD8+ T-cell responses coupled with fatal immunopathology. Together, these data suggest that suppressive MCs govern key aspects of respiratory CD8+ T-cell immunity, thereby balancing immunity and adverse pathology in the context of viral infection.
Collapse
Affiliation(s)
- Katie L Reagin
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Kimberly E Oliva
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Matthew R Hansen
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Chris D Slade
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Wendy T Watford
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Kimberly D Klonowski
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| |
Collapse
|
4
|
Sutanto H, Pradana FR, Adytia GJ, Ansharullah BA, Waitupu A, Bramantono B, Fetarayani D. Memory T Cells in Respiratory Virus Infections: Protective Potential and Persistent Vulnerabilities. Med Sci (Basel) 2025; 13:48. [PMID: 40407543 PMCID: PMC12101432 DOI: 10.3390/medsci13020048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/19/2025] [Accepted: 04/25/2025] [Indexed: 05/26/2025] Open
Abstract
Respiratory virus infections, such as those caused by influenza viruses, respiratory syncytial virus (RSV), and coronaviruses, pose a significant global health burden. While the immune system's adaptive components, including memory T cells, are critical for recognizing and combating these pathogens, recurrent infections and variable disease outcomes persist. Memory T cells are a key element of long-term immunity, capable of responding swiftly upon re-exposure to pathogens. They play diverse roles, including cross-reactivity to conserved viral epitopes and modulation of inflammatory responses. However, the protective efficacy of these cells is influenced by several factors, including viral evolution, host age, and immune system dynamics. This review explores the dichotomy of memory T cells in respiratory virus infections: their potential to confer robust protection and the limitations that allow for breakthrough infections. Understanding the underlying mechanisms governing the formation, maintenance, and functional deployment of memory T cells in respiratory mucosa is critical for improving immunological interventions. We highlight recent advances in vaccine strategies aimed at bolstering T cell-mediated immunity and discuss the challenges posed by viral immune evasion. Addressing these gaps in knowledge is pivotal for designing effective therapeutics and vaccines to mitigate the global burden of respiratory viruses.
Collapse
Affiliation(s)
- Henry Sutanto
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Febrian Ramadhan Pradana
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Galih Januar Adytia
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Bagus Aditya Ansharullah
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Alief Waitupu
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Bramantono Bramantono
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
- Division of Tropical and Infectious Diseases, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Deasy Fetarayani
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| |
Collapse
|
5
|
Sims AC, Schäfer A, Okuda K, Leist SR, Kocher JF, Cockrell AS, Hawkins PE, Furusho M, Jensen KL, Kyle JE, Burnum-Johnson KE, Stratton KG, Lamar NC, Niccora CD, Weitz KK, Smith RD, Metz TO, Waters KM, Boucher RC, Montgomery SA, Baric RS, Sheahan TP. Dysregulation of lung epithelial cell homeostasis and immunity contributes to Middle East respiratory syndrome coronavirus disease severity. mSphere 2025; 10:e0095124. [PMID: 39882872 PMCID: PMC11853001 DOI: 10.1128/msphere.00951-24] [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: 12/10/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
Coronaviruses (CoV) emerge suddenly from animal reservoirs to cause novel diseases in new hosts. Discovered in 2012, the Middle East respiratory syndrome coronavirus (MERS-CoV) is endemic in camels in the Middle East and is continually causing local outbreaks and epidemics. While all three newly emerging human CoVs from the past 20 years (SARS-CoV, SARS-CoV-2, and MERS-CoV) cause respiratory disease, each CoV has unique host interactions that drive differential pathogeneses. To better understand the virus and host interactions driving lethal MERS-CoV infection, we performed a longitudinal multi-omics analysis of sublethal and lethal MERS-CoV infection in mice. Significant differences were observed in body weight loss, virus titers, and acute lung injury among lethal and sub-lethal virus doses. Virus-induced apoptosis of type I and II alveolar epithelial cells suggests that loss or dysregulation of these key cell populations was a major driver of severe disease. Omics analysis suggested differential pathogenesis was multi-factorial with clear differences among innate and adaptive immune pathways as well as those that regulate lung epithelial homeostasis. Infection of mice lacking functional T and B cells showed that adaptive immunity was important in controlling viral replication but also increased pathogenesis. In summary, we provide a high-resolution host response atlas for MERS-CoV infection and disease severity. Multi-omics studies of viral pathogenesis offer a unique opportunity to not only better understand the molecular mechanisms of disease but also to identify genes and pathways that can be exploited for therapeutic intervention all of which is important for our future pandemic preparedness.IMPORTANCEEmerging coronaviruses like SARS-CoV, SARS-CoV-2, and MERS-CoV cause a range of disease outcomes in humans from an asymptomatic, moderate, and severe respiratory disease that can progress to death but the factors causing these disparate outcomes remain unclear. Understanding host responses to mild and life-threatening infections provides insight into virus-host networks within and across organ systems that contribute to disease outcomes. We used multi-omics approaches to comprehensively define the host response to moderate and severe MERS-CoV infection. Severe respiratory disease was associated with dysregulation of the immune response. Key lung epithelial cell populations that are essential for lung function get infected and die. Mice lacking key immune cell populations experienced greater virus replication but decreased disease severity implicating the immune system in both protective and pathogenic roles in response to MERS-CoV. These data could be utilized to design new therapeutic strategies targeting specific pathways that contribute to severe disease.
Collapse
Affiliation(s)
- Amy C. Sims
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kenichi Okuda
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sarah R. Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jacob F. Kocher
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Adam S. Cockrell
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Padraig E. Hawkins
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Minako Furusho
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kara L. Jensen
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jennifer E. Kyle
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | | | - Kelly G. Stratton
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Natalie C. Lamar
- AI & Data Analytics Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Carrie D. Niccora
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Karl K. Weitz
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Richard D. Smith
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Thomas O. Metz
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Katrina M. Waters
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Richard C. Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stephanie A. Montgomery
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Timothy P. Sheahan
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
6
|
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
|
7
|
Ko KH, Bae HS, Park JW, Lee JS, Park S, Heo J, Park H, Choi J, Bae E, Na W, Park SH, Seong BL, Han SH, Kim DH, Cha SB. A vaccine platform targeting lung-resident memory CD4 + T-cells provides protection against heterosubtypic influenza infections in mice and ferrets. Nat Commun 2024; 15:10368. [PMID: 39609429 PMCID: PMC11604757 DOI: 10.1038/s41467-024-54620-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
Lung tissue-resident memory T (TRM) cells induced by influenza vaccination are crucial for heterosubtypic immunity upon re-exposure to the influenza virus, enabling rapid and robust responses upon reactivation. To enhance the efficacy of influenza vaccines, we induce the generation of lung TRM cells following intranasal vaccination with a commercial influenza vaccine adjuvanted with NexaVant (NVT), a TLR3 agonist-based adjuvant. We demonstrate that intranasal immunization with the NVT-adjuvanted vaccine provides improved protection against influenza virus infections by inducing the generation of CD4+ TRM cells in the lungs in a type I interferon-dependent manner. These pulmonary CD4+ TRM cells provide potent mucosal immunity and cross-protection against heterosubtypic infections in both mouse and ferret models. This vaccine platform has the potential to significantly improve conventional intramuscular influenza vaccines by providing broader protection.
Collapse
Affiliation(s)
- Kwang Hyun Ko
- R&D Center, NA Vaccine Institute, Seoul, 05854, Republic of Korea
- Interdisciplinary Program in Genetic Engineering, College of Natural Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyun Shik Bae
- R&D Center, NA Vaccine Institute, Seoul, 05854, Republic of Korea
| | - Jeong Woo Park
- Interdisciplinary Program in Genetic Engineering, College of Natural Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jin-Sun Lee
- Interdisciplinary Program in Genetic Engineering, College of Natural Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Somin Park
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jun Heo
- Vaccine R&D Team, Central Institute, Il-Yang Pharmaceutical, Yongin, 17096, Republic of Korea
| | - Hyunsoo Park
- Vaccine R&D Team, Central Institute, Il-Yang Pharmaceutical, Yongin, 17096, Republic of Korea
| | - Jaeseok Choi
- College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Eunseo Bae
- College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Woonsung Na
- College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Seong-Hyun Park
- Graduate Program in Biomaterials Science and Engineering, College of Life Science and Biotechnology, Yonsei University, Seoul, 08826, Republic of Korea
| | - Baik-Lin Seong
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 08826, Republic of Korea
| | - Seung Hyun Han
- Interdisciplinary Program in Genetic Engineering, College of Natural Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dong-Ho Kim
- R&D Center, NA Vaccine Institute, Seoul, 05854, Republic of Korea.
| | - Seung Bin Cha
- R&D Center, NA Vaccine Institute, Seoul, 05854, Republic of Korea.
| |
Collapse
|
8
|
Fuchs J, Hübner J, Schmidt A, Irrgang P, Maier C, Vieira Antão A, Oltmanns F, Thirion C, Lapuente D, Tenbusch M. Evaluation of adenoviral vector Ad19a encoding RSV-F as novel vaccine against respiratory syncytial virus. NPJ Vaccines 2024; 9:205. [PMID: 39472590 PMCID: PMC11522487 DOI: 10.1038/s41541-024-01001-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of severe lower respiratory tract infections in infants and toddlers. Since natural infections do not induce persistent immunity, there is the need of vaccines providing long-term protection. Here, we evaluated a new adenoviral vector (rAd) vaccine based on the rare serotype rAd19a and compared the immunogenicity and efficacy to the highly immunogenic rAd5. Given as an intranasal boost in DNA primed mice, both vectors encoding the F protein provided efficient protection against a subsequent RSV infection. However, intramuscular immunization with rAd19a vectors provoked vaccine-enhanced disease after RSV infection compared to non-vaccinated animals. While mucosal IgA antibodies and tissue-resident memory T-cells in intranasally vaccinated mice rapidly control RSV replication, a strong anamnestic systemic T-cell response in absence of local immunity might be the reason for immune-mediated enhanced disease. Our study highlighted the potential benefits of developing effective mucosal against respiratory pathogens.
Collapse
Affiliation(s)
- Jana Fuchs
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Julian Hübner
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Anna Schmidt
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Pascal Irrgang
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Clara Maier
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Ana Vieira Antão
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Friederike Oltmanns
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | | | - Dennis Lapuente
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany.
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054, Erlangen, Germany.
| |
Collapse
|
9
|
Paik NY, Neethling J, Anwar M, Gupta P, Sanborn MA, Shen Z, Bandara T, Hyun J, Naiche LA, Kitajewski JK, Rehman J, Shin JW, Mehta D, Pajcini KV. Notch transcriptional target tmtc1 maintains vascular homeostasis. Cell Mol Life Sci 2024; 81:370. [PMID: 39190102 PMCID: PMC11349727 DOI: 10.1007/s00018-024-05407-9] [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: 02/17/2024] [Revised: 07/31/2024] [Accepted: 08/11/2024] [Indexed: 08/28/2024]
Abstract
Proper lung function requires the maintenance of a tight endothelial barrier while simultaneously permitting the exchange of macromolecules and fluids to underlying tissue. Disruption of this barrier results in an increased vascular permeability in the lungs, leading to acute lung injury. In this study, we set out to determine whether transcriptional targets of Notch signaling function to preserve vascular integrity. We tested the in vivo requirement for Notch transcriptional signaling in maintaining the pulmonary endothelial barrier by using two complementary endothelial-specific Notch loss-of-function murine transgenic models. Notch signaling was blocked using endothelial-specific activation of an inhibitor of Notch transcriptional activation, Dominant Negative Mastermindlike (DNMAML; CDH5CreERT2), or endothelial-specific loss of Notch1 (Notch1f/f; CDH5CreERT2). Both Notch mutants increased vascular permeability with pan-Notch inhibition by DNMAML showing a more severe phenotype in the lungs and in purified endothelial cells. RNA sequencing of primary lung endothelial cells (ECs) identified novel Notch targets, one of which was transmembrane O-mannosyltransferase targeting cadherins 1 (tmtc1). We show that tmtc1 interacts with vascular endothelial cadherin (VE-cadherin) and regulates VE-cadherin egress from the endoplasmic reticulum through direct interaction. Our findings demonstrate that Notch signaling maintains endothelial adherens junctions and vascular homeostasis by a transcriptional mechanism that drives expression of critical factors important for processing and transport of VE-cadherin.
Collapse
Affiliation(s)
- Na Yoon Paik
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - Jacob Neethling
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - Mumtaz Anwar
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - Prerak Gupta
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - Mark A Sanborn
- Department of Biochemistry, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Zekun Shen
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - Thilinie Bandara
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - James Hyun
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - L A Naiche
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL, 60612, USA
| | - Jan K Kitajewski
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL, 60612, USA
| | - Jalees Rehman
- Department of Biochemistry, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Jae-Won Shin
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - Dolly Mehta
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - Kostandin V Pajcini
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA.
| |
Collapse
|
10
|
Heil L, Jewell S, Lines JL, Garvy BA. The Altered Neonatal CD8 + T Cell Immunodominance Hierarchy during Influenza Virus Infection Impacts Peptide Vaccination. Viruses 2024; 16:1271. [PMID: 39205245 PMCID: PMC11359775 DOI: 10.3390/v16081271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Neonates are more susceptible to influenza virus infection than adults, resulting in increased morbidity and mortality and delayed clearance of the virus. Generating effective CD8+ T cell responses may be important for improving vaccination outcomes in vulnerable populations, but neonatal T cells frequently respond differently than adult cells. We sought to understand CD8+ T cell specificity and immunodominance during neonatal influenza infection and how any differences from the adult hierarchy might impact peptide vaccine effectiveness. Neonatal C57BL/6 mice displayed an altered CD8+ T cell immunodominance hierarchy during influenza infection, preferentially responding to an epitope in the influenza protein PA rather than the co-dominant adult response to NP and PA. Heterosubtypic infections in mice first infected as pups also displayed altered immunodominance and reduced protection compared to mice first infected as adults. Adoptive transfer of influenza-infected bone-marrow-derived dendritic cells promoted an NP-specific CD8+ T cell response in influenza-virus-infected pups and increased viral clearance. Finally, pups responded to PA (224-233), but not NP (366-374) during peptide vaccination. PA (224-233)-vaccinated mice were not protected during viral challenge. Epitope usage should be considered when designing vaccines that target T cells when the intended patient population includes infants and adults.
Collapse
Affiliation(s)
- Luke Heil
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA; (L.H.); (S.J.); (J.L.L.)
| | - Samantha Jewell
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA; (L.H.); (S.J.); (J.L.L.)
- Department of Physical and Life Sciences, Nevada State University, Henderson, NV 89002, USA
| | - J. Louise Lines
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA; (L.H.); (S.J.); (J.L.L.)
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Beth A. Garvy
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA; (L.H.); (S.J.); (J.L.L.)
- Division of Infectious Diseases, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
11
|
Yuan D, Yang J, Wu W, Amier Y, Li X, Wan W, Huang Y, Li J, Yu X. The immune factors have complex causal regulation effects on kidney stone disease: a mendelian randomization study. BMC Immunol 2024; 25:34. [PMID: 38877395 PMCID: PMC11177369 DOI: 10.1186/s12865-024-00627-x] [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: 03/18/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
PURPOSE Previous studies have reported the potential impact of immune cells on kidney stone disease (KSD), but definitive causal relationships have yet to be established. The purpose of this paper is to elucidate the potential causal association between immune cells and KSD by Mendelian randomization (MR) analysis. METHODS In our study, a thorough two-sample Mendelian randomization (MR) analysis was performed by us to determine the potential causal relationship between immune cell traits and kidney stone disease. We included a total of four immune traits (median fluorescence intensity (MFI), relative cellular (RC), absolute cellular (AC), and morphological parameters (MP)), which are publicly available data. GWAS summary data related to KSD (9713 cases and 366,693 controls) were obtained from the FinnGen consortium. The primary MR analysis method was Inverse variance weighted. Cochran's Q test, MR Egger, and MR-Pleiotropy RESidual Sum and Outlier (MR-PRESSO) were used to assess the stability of the results. RESULTS After FDR correction, the CD8 on HLA DR + CD8br (OR = 0.95, 95% CI = 0.93-0.98, p-value = 7.20 × 10- 4, q-value = 0.088) was determined to be distinctly associated with KSD, and we also found other 25 suggestive associations between immune cells and KSD, of which 13 associations were suggested as protective factors and 12 associations were suggested as risk factors. There was no horizontal pleiotropy or significant heterogeneity in our MR analysis, as determined by the p-value results of our Cochrane Q-test, MR Egger's intercept test, and MR-PRESSO, which were all > 0.05. CONCLUSIONS Our study has explored the potential causal connection between immune cells and KSD by Mendelian randomization analysis, thus providing some insights for future clinical studies.
Collapse
Affiliation(s)
- Dongfeng Yuan
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Junyi Yang
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weisong Wu
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yirixiatijiang Amier
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xianmiu Li
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenlong Wan
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yisheng Huang
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiabo Li
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Yu
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Liberalization Ave, No. 1095, Wuhan, 430030, China.
| |
Collapse
|
12
|
Jue H, Dan-fei C, Fang-fang L, Ke-pin Y, Jia-ye X, Hui-ting Z, Xiao-bo X, Jian C. Evaluating the link between immune characteristics and attention deficit hyperactivity disorder through a bi-directional Mendelian randomization study. Front Immunol 2024; 15:1367418. [PMID: 38903512 PMCID: PMC11188446 DOI: 10.3389/fimmu.2024.1367418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/15/2024] [Indexed: 06/22/2024] Open
Abstract
Context Despite the recognition of attention deficit hyperactivity disorder (ADHD) as a multifaceted neurodevelopmental disorder, its core causes are still ambiguous. The objective of this study was to explore if the traits of circulating immune cells contribute causally to susceptibility to ADHD. Methods By employing a unified GWAS summary data covering 731 immune traits from the GWAS Catalog (accession numbers from GCST0001391 to GCST0002121), our analysis focused on the flow cytometry of lymphocyte clusters, encompassing 3,757 Sardinians, to identify genetically expected immune cells. Furthermore, we obtained summarized GWAS statistics from the Psychiatric Genomics Consortium to evaluate the genetic forecasting of ADHD. The studies employed ADHD2019 (20,183 cases and 35,191 controls from the 2019 GWAS ADHD dataset) and ADHD2022 (38,691 cases and 275,986 controls from the 2022 GWAS ADHD dataset). Through the examination of genome-wide association signals, we identified shared genetic variances between circulating immune cells and ADHD, employing the comprehensive ADHD2022 dataset. We primarily utilized inverse variance weighted (IVW) and weighted median methods in our Mendelian randomization research and sensitivity assessments to evaluate diversity and pleiotropy. Results After adjusting for false discovery rate (FDR), three distinct immunophenotypes were identified as associated with the risk of ADHD: CD33 in Im MDSC (OR=1.03, CI: 1.01~1.04, P=3.04×10-5, PFDR =0.015), CD8br NKT %T cell (OR=1.08, 95%CI: 1.04~1.12, P=9.33×10-5, PFDR =0.023), and CD8br NKT %lymphocyte (OR=1.08, 95%CI: 1.03~1.12, P=3.59×10-4, PFDR =0.066). Furthermore, ADHD showed no statistical effects on immunophenotypes. It's worth noting that 20 phenotypes exist where ADHD's appearance could diminish 85% of immune cells, including FSC-A in myeloid DC (β= -0.278, 95% CI: 0.616~0.931, P=0.008), CD3 in CD45RA- CD4+ (β= -0.233, 95% CI: 0.654~0.960, P=0.017), CD62L- monocyte AC (β=0.227, 95% CI: 0.038~1.518, P=0.019), CD33 in CD33br HLA DR+ CD14dim (β= -0.331, 95% CI: 0.543~0.950, P=0.020), and CD25 in CD39+ resting Treg (β=0.226, 95% CI: 1.522, P=0.022), and FSC-A in monocytes (β= -0.255, 95% CI: 0.621~0.967, P=0.234), among others. Conclusion Studies indicate that the immune system's response influences the emergence of ADHD. The findings greatly improve our understanding of the interplay between immune responses and ADHD risk, aiding in the development of treatment strategies from an immunological perspective.
Collapse
Affiliation(s)
- Hu Jue
- First Clinical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chen Dan-fei
- First Clinical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Fang-fang
- First Clinical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Ke-pin
- First Clinical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xu Jia-ye
- First Clinical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhang Hui-ting
- Department of Pediatrics, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuan Xiao-bo
- Department of Pediatrics, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Chen Jian
- First Clinical School, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
13
|
Dillard JA, Taft-Benz SA, Knight AC, Anderson EJ, Pressey KD, Parotti B, Martinez SA, Diaz JL, Sarkar S, Madden EA, De la Cruz G, Adams LE, Dinnon KH, Leist SR, Martinez DR, Schäfer A, Powers JM, Yount BL, Castillo IN, Morales NL, Burdick J, Evangelista MKD, Ralph LM, Pankow NC, Linnertz CL, Lakshmanane P, Montgomery SA, Ferris MT, Baric RS, Baxter VK, Heise MT. Adjuvant-dependent impact of inactivated SARS-CoV-2 vaccines during heterologous infection by a SARS-related coronavirus. Nat Commun 2024; 15:3738. [PMID: 38702297 PMCID: PMC11068739 DOI: 10.1038/s41467-024-47450-x] [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: 10/06/2023] [Accepted: 04/02/2024] [Indexed: 05/06/2024] Open
Abstract
Whole virus-based inactivated SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide have been critical to the COVID-19 pandemic response. Although these vaccines are protective against homologous coronavirus infection, the emergence of novel variants and the presence of large zoonotic reservoirs harboring novel heterologous coronaviruses provide significant opportunities for vaccine breakthrough, which raises the risk of adverse outcomes like vaccine-associated enhanced respiratory disease. Here, we use a female mouse model of coronavirus disease to evaluate inactivated vaccine performance against either homologous challenge with SARS-CoV-2 or heterologous challenge with a bat-derived coronavirus that represents a potential emerging disease threat. We show that inactivated SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide can cause enhanced respiratory disease during heterologous infection, while use of an alternative adjuvant does not drive disease and promotes heterologous viral clearance. In this work, we highlight the impact of adjuvant selection on inactivated vaccine safety and efficacy against heterologous coronavirus infection.
Collapse
Affiliation(s)
- Jacob A Dillard
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sharon A Taft-Benz
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Audrey C Knight
- Department of Pathology & Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elizabeth J Anderson
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katia D Pressey
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Breantié Parotti
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sabian A Martinez
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jennifer L Diaz
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sanjay Sarkar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily A Madden
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabriela De la Cruz
- Pathology Services Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lily E Adams
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kenneth H Dinnon
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah R Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David R Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John M Powers
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Boyd L Yount
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Izabella N Castillo
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Noah L Morales
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jane Burdick
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Lauren M Ralph
- Pathology Services Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nicholas C Pankow
- Pathology Services Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Colton L Linnertz
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Premkumar Lakshmanane
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephanie A Montgomery
- Department of Pathology & Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Dallas Tissue Research, Farmers Branch, TX, USA
| | - Martin T Ferris
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ralph S Baric
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Victoria K Baxter
- Department of Pathology & Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Texas Biomedical Research Institute, San Antonio, TX, USA.
| | - Mark T Heise
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
14
|
Heise M, Dillard J, Taft-Benz S, Knight A, Anderson E, Pressey K, Parotti B, Martinez S, Diaz J, Sarkar S, Madden E, De la Cruz G, Adams L, Dinnon K, Leist S, Martinez D, Schaefer A, Powers J, Yount B, Castillo I, Morales N, Burdick J, Evangelista MK, Ralph L, Pankow N, Linnertz C, Lakshmanane P, Montgomery S, Ferris M, Baric R, Baxter V. Adjuvant-dependent effects on the safety and efficacy of inactivated SARS-CoV-2 vaccines during heterologous infection by a SARS-related coronavirus. RESEARCH SQUARE 2023:rs.3.rs-3401539. [PMID: 37961507 PMCID: PMC10635311 DOI: 10.21203/rs.3.rs-3401539/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Inactivated whole virus SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide (Alum) are among the most widely used COVID-19 vaccines globally and have been critical to the COVID-19 pandemic response. Although these vaccines are protective against homologous virus infection in healthy recipients, the emergence of novel SARS-CoV-2 variants and the presence of large zoonotic reservoirs provide significant opportunities for vaccine breakthrough, which raises the risk of adverse outcomes including vaccine-associated enhanced respiratory disease (VAERD). To evaluate this possibility, we tested the performance of an inactivated SARS-CoV-2 vaccine (iCoV2) in combination with Alum against either homologous or heterologous coronavirus challenge in a mouse model of coronavirus-induced pulmonary disease. Consistent with human results, iCoV2 + Alum protected against homologous challenge. However, challenge with a heterologous SARS-related coronavirus, Rs-SHC014-CoV (SHC014), up to at least 10 months post-vaccination, resulted in VAERD in iCoV2 + Alum-vaccinated animals, characterized by pulmonary eosinophilic infiltrates, enhanced pulmonary pathology, delayed viral clearance, and decreased pulmonary function. In contrast, vaccination with iCoV2 in combination with an alternative adjuvant (RIBI) did not induce VAERD and promoted enhanced SHC014 clearance. Further characterization of iCoV2 + Alum-induced immunity suggested that CD4+ T cells were a major driver of VAERD, and these responses were partially reversed by re-boosting with recombinant Spike protein + RIBI adjuvant. These results highlight potential risks associated with vaccine breakthrough in recipients of Alum-adjuvanted inactivated vaccines and provide important insights into factors affecting both the safety and efficacy of coronavirus vaccines in the face of heterologous virus infections.
Collapse
Affiliation(s)
- Mark Heise
- University of North Carolina at Chapel Hill
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Boyd Yount
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill
| | | | | | | | | | | | | | | | - Prem Lakshmanane
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC
| | | | | | | | - Victoria Baxter
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| |
Collapse
|
15
|
Yu Y, Wang J, Wu MX. Microneedle-Mediated Immunization Promotes Lung CD8+ T-Cell Immunity. J Invest Dermatol 2023; 143:1983-1992.e3. [PMID: 37044258 PMCID: PMC10524108 DOI: 10.1016/j.jid.2023.03.1672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/09/2023] [Accepted: 03/23/2023] [Indexed: 04/14/2023]
Abstract
Microneedle array has proven more efficient in stimulating humoral immunity than intramuscular vaccination. However, its effectiveness in inducing pulmonary CD8+ T cells remains elusive, which is essential to the frontline defense against pulmonary viral infections such as influenza and COVID-19 viruses. The current investigation reveals that superior CD8+ T-cell responses are elicited by immunization with a microneedle array over intradermal or intramuscular immunization using the model antigen ovalbumin, irrespective of whether or not the antigen is provided in the lung. Mechanistically, microneedle array-mediated immunization targeted the epidermal layer and stimulated predominantly Langerhans cells, resulting in increased expression of α4β1 adhesion molecules on the CD8+ T-cell surface, which may play a role in T-cell homing to the lung, whereas CD8+ T cells induced by intramuscular immunization did not express the adhesion molecule sufficiently. CD8+ T cells with a lung-homing propensity were also seen after intradermal vaccination, yet to a much lesser extent. Accordingly, microneedle array immunization provided stronger protection against influenza viral infection than intradermal or intramuscular immunization. The observations offer insights into a strong cross-talk between epidermal immunization and lung immunity and are valuable for designing and delivering vaccines against respiratory viral infections.
Collapse
Affiliation(s)
- Yang Yu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA
| | - Ji Wang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA; The first affiliated Hospital, Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
16
|
Agac A, Kolbe SM, Ludlow M, Osterhaus ADME, Meineke R, Rimmelzwaan GF. Host Responses to Respiratory Syncytial Virus Infection. Viruses 2023; 15:1999. [PMID: 37896776 PMCID: PMC10611157 DOI: 10.3390/v15101999] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/29/2023] Open
Abstract
Respiratory syncytial virus (RSV) infections are a constant public health problem, especially in infants and older adults. Virtually all children will have been infected with RSV by the age of two, and reinfections are common throughout life. Since antigenic variation, which is frequently observed among other respiratory viruses such as SARS-CoV-2 or influenza viruses, can only be observed for RSV to a limited extent, reinfections may result from short-term or incomplete immunity. After decades of research, two RSV vaccines were approved to prevent lower respiratory tract infections in older adults. Recently, the FDA approved a vaccine for active vaccination of pregnant women to prevent severe RSV disease in infants during their first RSV season. This review focuses on the host response to RSV infections mediated by epithelial cells as the first physical barrier, followed by responses of the innate and adaptive immune systems. We address possible RSV-mediated immunomodulatory and pathogenic mechanisms during infections and discuss the current vaccine candidates and alternative treatment options.
Collapse
Affiliation(s)
| | | | | | | | | | - Guus F. Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (A.A.); (S.M.K.); (M.L.); (A.D.M.E.O.); (R.M.)
| |
Collapse
|
17
|
Miles MA, Liong S, Liong F, Coward-Smith M, Trollope GS, Oseghale O, Erlich JR, Brooks RD, Logan JM, Hickey S, Wang H, Bozinovski S, O’Leary JJ, Brooks DA, Selemidis S. TLR7 promotes chronic airway disease in RSV-infected mice. Front Immunol 2023; 14:1240552. [PMID: 37795093 PMCID: PMC10545951 DOI: 10.3389/fimmu.2023.1240552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/28/2023] [Indexed: 10/06/2023] Open
Abstract
Respiratory syncytial virus (RSV) commonly infects the upper respiratory tract (URT) of humans, manifesting with mild cold or flu-like symptoms. However, in infants and the elderly, severe disease of the lower respiratory tract (LRT) often occurs and can develop into chronic airway disease. A better understanding of how an acute RSV infection transitions to a LRT chronic inflammatory disease is critically important to improve patient care and long-term health outcomes. To model acute and chronic phases of the disease, we infected wild-type C57BL/6 and toll-like receptor 7 knockout (TLR7 KO) mice with RSV and temporally assessed nasal, airway and lung inflammation for up to 42 days post-infection. We show that TLR7 reduced viral titers in the URT during acute infection but promoted pronounced pathogenic and chronic airway inflammation and hyperreactivity in the LRT. This study defines a hitherto unappreciated molecular mechanism of lower respiratory pathogenesis to RSV, highlighting the potential of TLR7 modulation to constrain RSV pathology to the URT.
Collapse
Affiliation(s)
- Mark A. Miles
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Stella Liong
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Felicia Liong
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Madison Coward-Smith
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Gemma S. Trollope
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Osezua Oseghale
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Jonathan R. Erlich
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Robert D. Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Jessica M. Logan
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Shane Hickey
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Hao Wang
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Steven Bozinovski
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - John J. O’Leary
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
- Sir Patrick Dun’s Laboratory, Central Pathology Laboratory, St James’s Hospital, Dublin, Ireland
| | - Doug A. Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
18
|
Kosanovich JL, Eichinger KM, Lipp MA, Gidwani SV, Brahmbhatt D, Yondola MA, Perkins TN, Empey KM. Exacerbated lung inflammation following secondary RSV exposure is CD4+ T cell-dependent and is not mitigated in infant BALB/c mice born to PreF-vaccinated dams. Front Immunol 2023; 14:1206026. [PMID: 37646035 PMCID: PMC10461110 DOI: 10.3389/fimmu.2023.1206026] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/25/2023] [Indexed: 09/01/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of childhood hospitalizations due to bronchiolitis in children under 5 years of age. Moreover, severe RSV disease requiring hospitalization is associated with the subsequent development of wheezing and asthma. Due to the young age in which viral protection is needed and risk of vaccine enhanced disease following direct infant vaccination, current approaches aim to protect young children through maternal immunization strategies that boost neutralizing maternal antibody (matAb) levels. However, there is a scarcity of studies investigating the influence of maternal immunization on secondary immune responses to RSV in the offspring or whether the subsequent development of wheezing and asthma is mitigated. Toward this goal, our lab developed a murine model of maternal RSV vaccination and repeat RSV exposure to evaluate the changes in immune response and development of exacerbated lung inflammation on secondary RSV exposure in mice born to immunized dams. Despite complete protection following primary RSV exposure, offspring born to pre-fusion F (PreF)-vaccinated dams had exaggerated secondary ILC2 and Th2 responses, characterized by enhanced production of IL-4, IL-5, and IL-13. These enhanced type 2 cellular responses were associated with exaggerated airway eosinophilia and mucus hyperproduction upon re-exposure to RSV. Importantly, depletion of CD4+ T cells led to complete amelioration of the observed type 2 pathology on secondary RSV exposure. These unanticipated results highlight the need for additional studies that look beyond primary protection to better understand how maternal immunization shapes subsequent immune responses to repeat RSV exposure.
Collapse
Affiliation(s)
- Jessica L. Kosanovich
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Katherine M. Eichinger
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Madeline A. Lipp
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | | | | | | | - Timothy N. Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Kerry M. Empey
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
19
|
Pulkina A, Vasilyev K, Muzhikyan A, Sergeeva M, Romanovskaya-Romanko E, Shurygina AP, Shuklina M, Vasin A, Stukova M, Egorov A. IgGκ Signal Peptide Enhances the Efficacy of an Influenza Vector Vaccine against Respiratory Syncytial Virus Infection in Mice. Int J Mol Sci 2023; 24:11445. [PMID: 37511205 PMCID: PMC10380829 DOI: 10.3390/ijms241411445] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Intranasal vaccination using influenza vectors is a promising approach to developing vaccines against respiratory pathogens due to the activation of the mucosa-associated immune response. However, there is no clear evidence of a vector design that could be considered preferable. To find the optimal structure of an influenza vector with a modified NS genomic segment, we constructed four vector expressing identical transgene sequences inherited from the F protein of the respiratory syncytial virus (RSV). Two vectors were designed aiming at transgene accumulation in the cytosol. Another two were supplemented with an IgGκ signal peptide prior to the transgene for its extracellular delivery. Surprisingly, adding the IgGκ substantially enhanced the T-cell immune response to the CD8 epitope of the transgene. Moreover, this strategy allowed us to obtain a better protection of mice from the RSV challenge after a single intranasal immunization. Protection was achieved without antibodies, mediated by a balanced T-cell immune response including the formation of the RSV specific effector CD8+ IFNγ+/IL10+-producing cells and the accumulation of Treg cells preventing immunopathology in the lungs of infected mice. In addition to the presented method for optimizing the influenza vector, our results highlight the possibility of achieving protection against RSV through a respiratory-associated T-cell immune response alone.
Collapse
Affiliation(s)
- Anastasia Pulkina
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Kirill Vasilyev
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Arman Muzhikyan
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Mariia Sergeeva
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Ekaterina Romanovskaya-Romanko
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Anna-Polina Shurygina
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Marina Shuklina
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Andrey Vasin
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Marina Stukova
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Andrej Egorov
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| |
Collapse
|
20
|
De C, Pickles RJ, Yao W, Liao B, Boone A, Choi M, Battaglia DM, Askin FB, Whitmire JK, Silvestri G, Garcia JV, Wahl A. Human T cells efficiently control RSV infection. JCI Insight 2023; 8:e168110. [PMID: 37159271 PMCID: PMC10393221 DOI: 10.1172/jci.insight.168110] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/26/2023] [Indexed: 05/10/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection causes significant morbidity and mortality in infants, immunocompromised individuals, and older individuals. There is an urgent need for effective antivirals and vaccines for high-risk individuals. We used 2 complementary in vivo models to analyze RSV-associated human lung pathology and human immune correlates of protection. RSV infection resulted in widespread human lung epithelial damage, a proinflammatory innate immune response, and elicited a natural adaptive human immune response that conferred protective immunity. We demonstrated a key role for human T cells in controlling RSV infection. Specifically, primed human CD8+ T cells or CD4+ T cells effectively and independently control RSV replication in human lung tissue in the absence of an RSV-specific antibody response. These preclinical data support the development of RSV vaccines, which also elicit effective T cell responses to improve RSV vaccine efficacy.
Collapse
Affiliation(s)
- Chandrav De
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Raymond J. Pickles
- Department of Microbiology and Immunology, and
- Marsico Lung Institute, University of North Carolina (UNC) at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wenbo Yao
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Baolin Liao
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
- Department of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Allison Boone
- Department of Microbiology and Immunology, and
- Marsico Lung Institute, University of North Carolina (UNC) at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mingyu Choi
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Diana M. Battaglia
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | | | - Jason K. Whitmire
- Department of Microbiology and Immunology, and
- Department of Genetics, and
- Lineberger Comprehensive Cancer Center, UNC at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Guido Silvestri
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - J. Victor Garcia
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Angela Wahl
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| |
Collapse
|
21
|
Dillard JA, Martinez SA, Dearing JJ, Montgomery SA, Baxter AK. Animal Models for the Study of SARS-CoV-2-Induced Respiratory Disease and Pathology. Comp Med 2023; 73:72-90. [PMID: 36229170 PMCID: PMC9948904 DOI: 10.30802/aalas-cm-22-000089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Emergence of the betacoronavirus SARS-CoV-2 has resulted in a historic pandemic, with millions of deaths worldwide. An unprecedented effort has been made by the medical, scientific, and public health communities to rapidly develop and implement vaccines and therapeutics to prevent and reduce hospitalizations and deaths. Although SARS-CoV-2 infection can lead to disease in many organ systems, the respiratory system is its main target, with pneumonia and acute respiratory distress syndrome as the hallmark features of severe disease. The large number of patients who have contracted COVID-19 infections since 2019 has permitted a detailed characterization of the clinical and pathologic features of the disease in humans. However, continued progress in the development of effective preventatives and therapies requires a deeper understanding of the pathogenesis of infection. Studies using animal models are necessary to complement in vitro findings and human clinical data. Multiple animal species have been evaluated as potential models for studying the respiratory disease caused by SARSCoV-2 infection. Knowing the similarities and differences between animal and human responses to infection is critical for effective translation of animal data into human medicine. This review provides a detailed summary of the respiratory disease and associated pathology induced by SARS-CoV-2 infection in humans and compares them with the disease that develops in 3 commonly used models: NHP, hamsters, and mice. The effective use of animals to study SARS-CoV-2-induced respiratory disease will enhance our understanding of SARS-CoV-2 pathogenesis, allow the development of novel preventatives and therapeutics, and aid in the preparation for the next emerging virus with pandemic potential.
Collapse
Key Words
- ace2, angiotensin-converting enzyme 2
- agm, african green monkey
- ali, acute lung injury
- ards, acute respiratory distress syndrome
- balf, bronchoalveolar lavage fluid
- cards, covid-19-associated acute respiratory distress syndrome
- dad, diffuse alveolar damage
- dpi, days postinfection
- ggo, ground glass opacities
- s, spike glycoprotein
Collapse
Affiliation(s)
- Jacob A Dillard
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sabian A Martinez
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Justin J Dearing
- Biological and Biomedical Sciences Program, Office of Graduate Education, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Stephanie A Montgomery
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Andvictoria K Baxter
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina;,
| |
Collapse
|
22
|
Kim KH, Pyo H, Lee H, Oh D, Noh JM, Ahn YC, Kim CG, Yoon HI, Lee J, Park S, Jung HA, Sun JM, Lee SH, Ahn JS, Park K, Ku BM, Shin EC, Ahn MJ. Association of T Cell Senescence with Radiation Pneumonitis in Patients with Non-small Cell Lung Cancer. Int J Radiat Oncol Biol Phys 2023; 115:464-475. [PMID: 35896144 DOI: 10.1016/j.ijrobp.2022.07.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/07/2022] [Accepted: 07/13/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE Associations between immunosenescence and radiation pneumonitis (RP) are largely unknown. We aimed to identify a peripheral blood T cell senescence biomarker to predict RP in patients with non-small cell lung cancer (NSCLC). METHODS AND MATERIALS Patients with locally advanced NSCLC who received definitive concurrent chemoradiotherapy (dCRT) were prospectively registered (cohort 1, n=23; cohort 2, n=31). Peripheral blood was collected at baseline, during dCRT, and at 1 month post-dCRT. Patients were dichotomized to grade ≥2 (G2+) RP and grade 0-1 (G0-1) RP. Flow cytometry was performed to assess phenotypes and functional properties of T cell subsets. RP incidence was estimated via competing risk analysis. RESULTS Five and six patients exhibited G2+ RP following dCRT in cohorts 1 and 2, respectively. Patients with G2+ RP exhibited a more aged T cell pool and higher frequencies of senescent CD57+CD28-CD8+ T cells than patients with G0-1 RP at baseline, during dCRT, and at 1 month post-dCRT. These senescent cells exhibited increased granzyme B, IFN-γ, and TNF-α production. Higher baseline frequency of CD57+CD28-CD8+ T cells was an independent predictor of G2+ RP (hazard ratio, 8.42; 95% confidence interval, 2.58-27.45; P<0.001). Recursive partitioning analysis revealed three distinct risk groups stratified by baseline CD57+CD28-CD8+ T cell frequency and lung V20 Gy, with 1-year cumulative G2+ RP incidences of 50.0%, 16.7%, and 0% for high-, intermediate-, and low-risk groups, respectively (P=0.002). CONCLUSIONS Higher baseline frequencies of CD57+CD28-CD8+ T cells correlated with increased G2+ RP risks. Our results suggest the need for further investigation of the role of T cell senescence on radiation-induced organ damage.
Collapse
Affiliation(s)
- Kyung Hwan Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hongryull Pyo
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hoyoung Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Dongryul Oh
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jae Myoung Noh
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yong Chan Ahn
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Chang Gon Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hong In Yoon
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jiyun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea; Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun-Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Keunchil Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Bo Mi Ku
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
23
|
Rodríguez-Guilarte L, Ramírez MA, Andrade CA, Kalergis AM. LAG-3 Contribution to T Cell Downmodulation during Acute Respiratory Viral Infections. Viruses 2023; 15:147. [PMID: 36680187 PMCID: PMC9865459 DOI: 10.3390/v15010147] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/05/2023] Open
Abstract
LAG-3 is a type I transmembrane protein expressed on immune cells, such as activated T cells, and binds to MHC class II with high affinity. LAG-3 is an inhibitory receptor, and its multiple biological activities on T cell activation and effector functions play a regulatory role in the immune response. Immunotherapies directed at immune checkpoints, including LAG-3, have become a promising strategy for controlling malignant tumors and chronic viral diseases. Several studies have suggested an association between the expression of LAG-3 with an inadequate immune response during respiratory viral infections and the susceptibility to reinfections, which might be a consequence of the inhibition of T cell effector functions. However, important information relative to therapeutic potential during acute viral lower respiratory tract infections and the mechanism of action of the LAG-3 checkpoint remains to be characterized. In this article, we discuss the contribution of LAG-3 to the impairment of T cells during viral respiratory infections. Understanding the host immune response to respiratory infections is crucial for developing effective vaccines and therapies.
Collapse
Affiliation(s)
- Linmar Rodríguez-Guilarte
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Mario A. Ramírez
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Catalina A. Andrade
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Alexis M. Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| |
Collapse
|
24
|
Li N, Shi T, Zhang M, He Y, Feng J, Mei Z, Su X, Jie Z. PLZF promotes the development of asthma tolerance via affecting memory phenotypes of immune cells. Int Immunopharmacol 2023; 114:109559. [PMID: 36525795 DOI: 10.1016/j.intimp.2022.109559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Clarifying the pathogenesis of asthma and/or identifying the specific pathway underlying oral asthma tolerance (OT) would be of great significance. In our previous study, promyelocytic leukemia zinc finger (PLZF), which reportedly regulates memory phenotypes, was found to promote ovalbumin (OVA)-induced OT. Therefore, this study aimed to explore the regulatory effects of PLZF on memory phenotypes in asthma and OT mouse models. We found that Zbtb16 (encoding PLZF) and PLZF+ cells were highly increased in OT lungs compared with asthmatic lungs. PLZF was co-expressed with GATA3, and IL-4+PLZF+ cells were significantly lower in OT lungs than in asthmatic lungs. Notably, memory cells were decreased in OT mice, and these mice had PLZF+ cells that expressed lower levels of CD44 than those of asthmatic mice. When Zbtb16 was overexpressed in splenic lymphocytes, the number of CD44+ cells decreased. There were increased memory cells in splenic lymphocytes after treatment with the supernatant of OVA-treated airway epithelial cells; however, this was reversed by Zbtb16 overexpression. Early respiratory syncytial virus infection increased memory cells and reduced PLZF+ cells in the OT mice. Collectively, these results indicate that PLZF may reduce the proportion of memory cells, thereby, promoting the establishment of OT.
Collapse
Affiliation(s)
- Na Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Department of Medicine, Respiratory, Emergency and Intensive Care Medicine, The Affiliated Dushu Lake Hospital of Soochow University, Suzhou, China
| | - Tianyun Shi
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Meng Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yanchao He
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jingjing Feng
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Zhoufang Mei
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Xiao Su
- Unit of Respiratory Infection and Immunity, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
| | - Zhijun Jie
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
25
|
Huang L, Liu MQ, Wan CQ, Cheng NN, Su YB, Zheng YP, Peng XL, Yu JM, Fu YH, He JS. The protective immunity induced by intranasally inoculated serotype 63 chimpanzee adenovirus vector expressing human respiratory syncytial virus prefusion fusion glycoprotein in BALB/c mice. Front Microbiol 2022; 13:1041338. [PMID: 36466668 PMCID: PMC9716990 DOI: 10.3389/fmicb.2022.1041338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/21/2022] [Indexed: 12/23/2023] Open
Abstract
Human respiratory syncytial virus (RSV) is a ubiquitous pediatric pathogen causing serious lower respiratory tract disease worldwide. No licensed vaccine is currently available. In this work, the coding gene for mDS-Dav1, the full-length and prefusion conformation RSV fusion glycoprotein (F), was designed by introducing the stabilized prefusion F (preF) mutations from DS-Cav1 into the encoding gene of wild-type RSV (wtRSV) F protein. The recombinant adenovirus encoding mDS-Cav1, rChAd63-mDS-Cav1, was constructed based on serotype 63 chimpanzee adenovirus vector and characterized in vitro. After immunizing mice via intranasal route, the rChAd63-mDS-Cav1 induced enhanced neutralizing antibody and F-specific CD8+ T cell responses as well as good immune protection against RSV challenge with the absence of enhanced RSV disease (ERD) in BALB/c mice. The results indicate that rChAd63-mDS-Cav1 is a promising mucosal vaccine candidate against RSV infection and warrants further development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yuan-Hui Fu
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Jin-Sheng He
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, China
| |
Collapse
|
26
|
Wang Y, Zheng J, Wang X, Yang P, Zhao D. Alveolar macrophages and airway hyperresponsiveness associated with respiratory syncytial virus infection. Front Immunol 2022; 13:1012048. [PMID: 36341376 PMCID: PMC9630648 DOI: 10.3389/fimmu.2022.1012048] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a ubiquitous pathogen of viral bronchiolitis and pneumonia in children younger than 2 years of age, which is closely associated with recurrent wheezing and airway hyperresponsiveness (AHR). Alveolar macrophages (AMs) located on the surface of the alveoli cavity are the important innate immune barrier in the respiratory tract. AMs are recognized as recruited airspace macrophages (RecAMs) and resident airspace macrophages (RAMs) based on their origins and roaming traits. AMs are polarized in the case of RSV infection, forming two macrophage phenotypes termed as M1-like and M2-like macrophages. Both M1 macrophages and M2 macrophages are involved in the modulation of inflammatory responses, among which M1 macrophages are capable of pro-inflammatory responses and M2 macrophages are capable of anti-proinflammatory responses and repair damaged tissues in the acute and convalescent phases of RSV infection. Polarized AMs affect disease progression through the alteration of immune cell surface phenotypes as well as participate in the regulation of T lymphocyte differentiation and the type of inflammatory response, which are closely associated with long-term AHR. In recent years, some progress have been made in the regulatory mechanism of AM polarization caused by RSV infection, which participates in acute respiratory inflammatory response and mediating AHR in infants. Here we summarized the role of RSV-infection-mediated AM polarization associated with AHR in infants.
Collapse
Affiliation(s)
- Yuxin Wang
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junwen Zheng
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xia Wang
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Pu Yang
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Children’s Digital Health and Data Center of Wuhan University, Wuhan, China
- *Correspondence: Dongchi Zhao, ; Pu Yang,
| | - Dongchi Zhao
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Children’s Digital Health and Data Center of Wuhan University, Wuhan, China
- *Correspondence: Dongchi Zhao, ; Pu Yang,
| |
Collapse
|
27
|
Costa Silva RCM, Bandeira-Melo C, Paula Neto HA, Vale AM, Travassos LH. COVID-19 diverse outcomes: Aggravated reinfection, type I interferons and antibodies. Med Hypotheses 2022; 167:110943. [PMID: 36105250 PMCID: PMC9461281 DOI: 10.1016/j.mehy.2022.110943] [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: 02/17/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 12/04/2022]
Abstract
SARS-CoV-2 infection intrigued medicine with diverse outcomes ranging from asymptomatic to severe acute respiratory syndrome (SARS) and death. After more than two years of pandemic, reports of reinfection concern researchers and physicists. Here, we will discuss potential mechanisms that can explain reinfections, including the aggravated ones. The major topics of this hypothesis paper are the disbalance between interferon and antibodies responses, HLA heterogeneity among the affected population, and increased proportion of cytotoxic CD4+ T cells polarization in relation to T follicular cells (Tfh) subtypes. These features affect antibody levels and hamper the humoral immunity necessary to prevent or minimize the viral burden in the case of reinfections.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratório de Imunoreceptores e Sinalização, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Christianne Bandeira-Melo
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heitor Afonso Paula Neto
- Laboratório de Alvos Moleculares, Faculdade de Farmácia, Departamento de Biotecnologia Farmacêutica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - André Macedo Vale
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Holanda Travassos
- Laboratório de Imunoreceptores e Sinalização, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
28
|
Wang L, Cao Z, Wang Z, Guo J, Wen J. Reactive oxygen species associated immunoregulation post influenza virus infection. Front Immunol 2022; 13:927593. [PMID: 35967412 PMCID: PMC9373727 DOI: 10.3389/fimmu.2022.927593] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
An appropriate level of reactive oxygen species (ROS) is necessary for cell proliferation, signaling transduction, and apoptosis due to their highly reactive character. ROS are generated through multiple metabolic pathways under a fine-tuned control between oxidant and antioxidant signaling. A growing number of evidence has proved their highly relevant role in modulating inflammation during influenza virus infection. As a network of biological process for protecting organism from invasion of pathogens, immune system can react and fight back through either innate immune system or adaptive immune system, or both. Herein, we provide a review about the mechanisms of ROS generation when encounter influenza virus infection, and how the imbalanced level of ROS influences the replication of virus. We also summarize the pathways used by both the innate and adaptive immune system to sense and attack the invaded virus and abnormal levels of ROS. We further review the limitation of current strategies and discuss the direction of future work.
Collapse
Affiliation(s)
- Lan Wang
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States,UCLA Acquired Immune Deficiency Syndrome (AIDS) Institute, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Zheng Cao
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Zi Wang
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Jimin Guo
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States,UCLA Acquired Immune Deficiency Syndrome (AIDS) Institute, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Jing Wen
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States,UCLA Acquired Immune Deficiency Syndrome (AIDS) Institute, University of California Los Angeles (UCLA), Los Angeles, CA, United States,*Correspondence: Jing Wen,
| |
Collapse
|
29
|
Maier C, Fuchs J, Irrgang P, Wißing MH, Beyerlein J, Tenbusch M, Lapuente D. Mucosal immunization with an adenoviral vector vaccine confers superior protection against RSV compared to natural immunity. Front Immunol 2022; 13:920256. [PMID: 36003372 PMCID: PMC9394428 DOI: 10.3389/fimmu.2022.920256] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/07/2022] [Indexed: 01/09/2023] Open
Abstract
Respiratory syncytial virus (RSV) infections are the leading cause of severe respiratory illness in early infancy. Although the majority of children and adults mount immune responses against RSV, recurrent infections are frequent throughout life. Humoral and cellular responses contribute to an effective immunity but also their localization at respiratory mucosae is increasingly recognized as an important factor. In the present study, we evaluate a mucosal vaccine based on an adenoviral vector encoding for the RSV fusion protein (Ad-F), and we investigate two genetic adjuvant candidates that encode for Interleukin (IL)-1β and IFN-β promoter stimulator I (IPS-1), respectively. While vaccination with Ad-F alone was immunogenic, the inclusion of Ad-IL-1β increased F-specific mucosal immunoglobulin A (IgA) and tissue-resident memory T cells (TRM). Consequently, immunization with Ad-F led to some control of virus replication upon RSV infection, but Ad-F+Ad-IL-1β was the most effective vaccine strategy in limiting viral load and weight loss. Subsequently, we compared the Ad-F+Ad-IL-1β-induced immunity with that provoked by a primary RSV infection. Systemic F-specific antibody responses were higher in immunized than in previously infected mice. However, the primary infection provoked glycoprotein G-specific antibodies as well eventually leading to similar neutralization titers in both groups. In contrast, mucosal antibody levels were low after infection, whereas mucosal immunization raised robust F-specific responses including IgA. Similarly, vaccination generated F-specific TRM more efficiently compared to a primary RSV infection. Although the primary infection resulted in matrix protein 2 (M2)-specific T cells as well, they did not reach levels of F-specific immunity in the vaccinated group. Moreover, the infection-induced T cell response was less biased towards TRM compared to vaccine-induced immunity. Finally, our vaccine candidate provided superior protection against RSV infection compared to a primary infection as indicated by reduced weight loss, virus replication, and tissue damage. In conclusion, our mucosal vaccine candidate Ad-F+Ad-IL-1β elicits stronger mucosal immune responses and a more effective protection against RSV infection than natural immunity generated by a previous infection. Harnessing mucosal immune responses by next-generation vaccines is therefore a promising option to establish effective RSV immunity and thereby tackle a major cause of infant hospitalization.
Collapse
Affiliation(s)
- Clara Maier
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Jana Fuchs
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Pascal Irrgang
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Jasmin Beyerlein
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany,*Correspondence: Matthias Tenbusch, ; Dennis Lapuente,
| | - Dennis Lapuente
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany,*Correspondence: Matthias Tenbusch, ; Dennis Lapuente,
| |
Collapse
|
30
|
Hirai T, Yoshioka Y. Considerations of CD8+ T Cells for Optimized Vaccine Strategies Against Respiratory Viruses. Front Immunol 2022; 13:918611. [PMID: 35774782 PMCID: PMC9237416 DOI: 10.3389/fimmu.2022.918611] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
The primary goal of vaccines that protect against respiratory viruses appears to be the induction of neutralizing antibodies for a long period. Although this goal need not be changed, recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have drawn strong attention to another arm of acquired immunity, CD8+ T cells, which are also called killer T cells. Recent evidence accumulated during the coronavirus disease 2019 (COVID-19) pandemic has revealed that even variants of SARS-CoV-2 that escaped from neutralizing-antibodies that were induced by either infection or vaccination could not escape from CD8+ T cell-mediated immunity. In addition, although traditional vaccine platforms, such as inactivated virus and subunit vaccines, are less efficient in inducing CD8+ T cells, newly introduced platforms for SARS-CoV-2, namely, mRNA and adenoviral vector vaccines, can induce strong CD8+ T cell-mediated immunity in addition to inducing neutralizing antibodies. However, CD8+ T cells function locally and need to be at the site of infection to control it. To fully utilize the protective performance of CD8+ T cells, it would be insufficient to induce only memory cells circulating in blood, using injectable vaccines; mucosal immunization could be required to set up CD8+ T cells for the optimal protection. CD8+ T cells might also contribute to the pathology of the infection, change their function with age and respond differently to booster vaccines in comparison with antibodies. Herein, we overview cutting-edge ideas on CD8+ T cell-mediated immunity that can enable the rational design of vaccines for respiratory viruses.
Collapse
Affiliation(s)
- Toshiro Hirai
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- *Correspondence: Toshiro Hirai,
| | - Yasuo Yoshioka
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- The Research Foundation for Microbial Diseases of Osaka University, Suita, Japan
| |
Collapse
|
31
|
Pathogenesis of pneumonia and acute lung injury. Clin Sci (Lond) 2022; 136:747-769. [PMID: 35621124 DOI: 10.1042/cs20210879] [Citation(s) in RCA: 157] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/29/2022] [Accepted: 05/09/2022] [Indexed: 12/15/2022]
Abstract
Pneumonia and its sequelae, acute lung injury, present unique challenges for pulmonary and critical care healthcare professionals, and these challenges have recently garnered global attention due to the ongoing Sars-CoV-2 pandemic. One limitation to translational investigation of acute lung injury, including its most severe manifestation (acute respiratory distress syndrome, ARDS) has been heterogeneity resulting from the clinical and physiologic diagnosis that represents a wide variety of etiologies. Recent efforts have improved our understanding and approach to heterogeneity by defining sub-phenotypes of ARDS although significant gaps in knowledge remain. Improving our mechanistic understanding of acute lung injury and its most common cause, infectious pneumonia, can advance our approach to precision targeted clinical interventions. Here, we review the pathogenesis of pneumonia and acute lung injury, including how respiratory infections and lung injury disrupt lung homoeostasis, and provide an overview of respiratory microbial pathogenesis, the lung microbiome, and interventions that have been demonstrated to improve outcomes-or not-in human clinical trials.
Collapse
|
32
|
Blunck BN, Angelo LS, Henke D, Avadhanula V, Cusick M, Ferlic-Stark L, Zechiedrich L, Gilbert BE, Piedra PA. Adult Memory T Cell Responses to the Respiratory Syncytial Virus Fusion Protein During a Single RSV Season (2018-2019). Front Immunol 2022; 13:823652. [PMID: 35422803 PMCID: PMC9002099 DOI: 10.3389/fimmu.2022.823652] [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: 11/27/2021] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
Respiratory Syncytial Virus (RSV) is ubiquitous and re-infection with both subtypes (RSV/A and RSV/B) is common. The fusion (F) protein of RSV is antigenically conserved, induces neutralizing antibodies, and is a primary target of vaccine development. Insight into the breadth and durability of RSV-specific adaptive immune response, particularly to the F protein, may shed light on susceptibility to re-infection. We prospectively enrolled healthy adult subjects (n = 19) and collected serum and peripheral blood mononuclear cells (PBMCs) during the 2018–2019 RSV season. Previously, we described their RSV-specific antibody responses and identified three distinct antibody kinetic profiles associated with infection status: uninfected (n = 12), acutely infected (n = 4), and recently infected (n = 3). In this study, we measured the longevity of RSV-specific memory T cell responses to the F protein following natural RSV infection. We stimulated PBMCs with overlapping 15-mer peptide libraries spanning the F protein derived from either RSV/A or RSV/B and found that memory T cell responses mimic the antibody responses for all three groups. The uninfected group had stable, robust memory T cell responses and polyfunctionality. The acutely infected group had reduced polyfunctionality of memory T cell response at enrollment compared to the uninfected group, but these returned to comparable levels by end-of-season. The recently infected group, who were unable to maintain high levels of RSV-specific antibody following infection, similarly had decreased memory T cell responses and polyfunctionality during the RSV season. We observed subtype-specific differences in memory T cell responses and polyfunctionality, with RSV/A stimulating stronger memory T cell responses with higher polyfunctionality even though RSV/B was the dominant subtype in circulation. A subset of individuals demonstrated an overall deficiency in the generation of a durable RSV-specific adaptive immune response. Because memory T cell polyfunctionality may be associated with protection against re-infection, this latter group would likely be at greater risk of re-infection. Overall, these results expand our understanding of the longevity of the adaptive immune response to the RSV fusion protein and should be considered in future vaccine development efforts.
Collapse
Affiliation(s)
- Brittani N Blunck
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Laura S Angelo
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - David Henke
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Vasanthi Avadhanula
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Matthew Cusick
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Laura Ferlic-Stark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Lynn Zechiedrich
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, United States.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, United States
| | - Brian E Gilbert
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
33
|
Leonardi AJ, Argyropoulos CP, Hamdy A, Proenca RB. Understanding the Effects of Age and T-Cell Differentiation on COVID-19 Severity: Implicating a Fas/FasL-mediated Feed-Forward Controller of T-Cell Differentiation. Front Immunol 2022; 13:853606. [PMID: 35309371 PMCID: PMC8927653 DOI: 10.3389/fimmu.2022.853606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/11/2022] [Indexed: 01/14/2023] Open
Affiliation(s)
- Anthony J Leonardi
- Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Christos P Argyropoulos
- Department of Internal Medicine, Division of Nephrology, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Adam Hamdy
- Independent Researcher, Port Louis, Mauritius
| | - Rui B Proenca
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
34
|
Restriction of viral replication, rather than T cell immunopathology, drives lethality in MNV CR6-infected STAT1-deficient mice. J Virol 2022; 96:e0206521. [PMID: 35107369 DOI: 10.1128/jvi.02065-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recent evidence indicates that viral components of the microbiota can contribute to intestinal homeostasis and protection from local inflammatory or infectious insults. However, host-derived mechanisms that regulate the virome remain largely unknown. Here, we use colonization with the model commensal murine norovirus (MNV CR6) to interrogate host-directed mechanisms of viral regulation, and show that STAT1 is a central coordinator of both viral replication and antiviral T cell responses. In addition to restricting CR6 replication to the intestinal tract, we show that STAT1 regulates antiviral CD4+ and CD8+ T cell responses, and prevents systemic viral-induced tissue damage and disease. Despite altered T cell responses that resemble those that mediate lethal immunopathology in systemic viral infections in STAT1-deficient mice, depletion of adaptive immune cells and their associated effector functions had no effect on CR6-induced disease. However, therapeutic administration of an antiviral compound limited viral replication, preventing viral-induced tissue damage and death without impacting the generation of inflammatory antiviral T cell responses. Collectively, our data show that STAT1 restricts MNV CR6 replication within the intestinal mucosa, and that uncontrolled viral replication mediates disease rather than the concomitant development of dysregulated antiviral T cell responses in STAT1-deficient mice. Importance The intestinal microbiota is a collection of bacteria, archaea, fungi and viruses that colonize the mammalian gut. Co-evolution of the host and microbiota has required development of immunological tolerance to prevent ongoing inflammatory responses against intestinal microbes. Breakdown of tolerance to bacterial components of the microbiota can contribute to immune activation and inflammatory disease. However, the mechanisms that are necessary to maintain tolerance to viral components of the microbiome, and the consequences of loss of tolerance, are less well understood. Here, we show that STAT1 is integral for preventing escape of a commensal-like virus, murine norovirus CR6 (MNV CR6) from the gut, and that in the absence of STAT1, mice succumb to infection-induced disease. In contrast to other systemic viral infections, mortality of STAT1-deficient mice is not driven by immune-mediated pathology. Our data demonstrates the importance of host-mediated geographical restriction of commensal-like viruses.
Collapse
|
35
|
Saeland E, van der Fits L, Bolder R, Heemskerk-van der Meer M, Drijver J, van Polanen Y, Vaneman C, Tettero L, Serroyen J, Schuitemaker H, Callendret B, Langedijk JPM, Zahn RC. Immunogenicity and protective efficacy of adenoviral and subunit RSV vaccines based on stabilized prefusion F protein in pre-clinical models. Vaccine 2021; 40:934-944. [PMID: 34973849 DOI: 10.1016/j.vaccine.2021.12.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/29/2021] [Accepted: 12/16/2021] [Indexed: 11/29/2022]
Abstract
Respiratory Syncytial Virus (RSV) remains a leading cause of severe respiratory disease for which no licensed vaccine is available. We have previously described the derivation of an RSV Fusion protein (F) stabilized in its prefusion conformation (preF) as vaccine immunogen and demonstrated superior immunogenicity in naive mice of preF versus wild type RSV F protein, both as protein and when expressed from an Ad26 vaccine vector. Here we address the question if there are qualitative differences between the two vaccine platforms for induction of protective immunity. In naïve mice, both Ad26.RSV.preF and preF protein induced humoral responses, whereas cellular responses were only elicited by Ad26.RSV.preF. In RSV pre-exposed mice, a single dose of either vaccine induced cellular responses and strong humoral responses. Ad26-induced RSV-specific cellular immune responses were detected systemically and locally in the lungs. Both vaccines showed protective efficacy in the cotton rat model, but Ad26.RSV.preF conferred protection at lower virus neutralizing titers in comparison to RSV preF protein. Factors that may contribute to the protective capacity of Ad26.RSV.preF elicited immunity are the induced IgG2a antibodies that are able to engage Fcγ receptors mediating Antibody Dependent Cellular Cytotoxicity (ADCC), and the induction of systemic and lung resident RSV specific CD8 + T cells. These data demonstrate qualitative improvement of immune responses elicited by an adenoviral vector based vaccine encoding the RSV preF antigen compared to the subunit vaccine in small animal models which may inform RSV vaccine development.
Collapse
Affiliation(s)
| | | | - Renske Bolder
- Janssen Vaccines & Prevention, Leiden, the Netherlands
| | | | - Joke Drijver
- Janssen Vaccines & Prevention, Leiden, the Netherlands
| | | | | | | | - Jan Serroyen
- Janssen Vaccines & Prevention, Leiden, the Netherlands
| | | | | | | | - Roland C Zahn
- Janssen Vaccines & Prevention, Leiden, the Netherlands
| |
Collapse
|
36
|
Osuna-Gómez R, Arqueros C, Galano C, Mulet M, Zamora C, Barnadas A, Vidal S. Effector Mechanisms of CD8+ HLA-DR+ T Cells in Breast Cancer Patients Who Respond to Neoadjuvant Chemotherapy. Cancers (Basel) 2021; 13:6167. [PMID: 34944786 PMCID: PMC8699550 DOI: 10.3390/cancers13246167] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/01/2021] [Accepted: 12/04/2021] [Indexed: 01/09/2023] Open
Abstract
Cytotoxic T lymphocyte (CTLs) activation is an independent predictor of response to neoadjuvant chemotherapy (NACT) in breast cancer (BC) patients. Here, we go deeper into the function of CD8+ HLA-DR+ T cells from NACT treated HER2 negative BC patients. Flow cytometry analysis revealed that CD8+ HLA-DR+ T cell percentage was increased in NACT responder (R) compared to non-responder (NR) patients. R patients with ER-/PR- hormone receptors had the highest CD8+ HLA-DR+ T cell frequencies, while no differences were found when patients were classified according to cancer stage or menopause status. Interestingly, the cytotoxicity and production of anti-tumor cytokines were enhanced when CD8+ HLA-DR+ T cells from healthy donors were cultured with plasma from R, but not from NR patients. The induced anti-tumor profile of CD8+ HLA-DR+ T cells was associated with plasmatic IL-12 and IFN-γ levels, increased cytokines in R patients. IL-12 or IFN-γ neutralization decreased cytotoxic activity and TNF-α production by cultured CD8+ HLA-DR+ T cells in R plasma presence. All these data suggest that an effective response to NACT in BC patients is associated with increased IL-12 or IFN-γ levels involved in the induction of cytotoxic and pro-inflammatory mechanisms in CD8+ HLA-DR+ T cells.
Collapse
Affiliation(s)
- Rubén Osuna-Gómez
- Inflammatory Diseases, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (R.O.-G.); (C.G.); (M.M.); (C.Z.)
| | - Cristina Arqueros
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.A.); (A.B.)
- Centro de Investigación Biomedica en Red Cancer (CIBERONC), 28029 Madrid, Spain
| | - Carla Galano
- Inflammatory Diseases, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (R.O.-G.); (C.G.); (M.M.); (C.Z.)
| | - Maria Mulet
- Inflammatory Diseases, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (R.O.-G.); (C.G.); (M.M.); (C.Z.)
| | - Carlos Zamora
- Inflammatory Diseases, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (R.O.-G.); (C.G.); (M.M.); (C.Z.)
| | - Agustí Barnadas
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.A.); (A.B.)
- Centro de Investigación Biomedica en Red Cancer (CIBERONC), 28029 Madrid, Spain
- School of Medicine, Universitat Autònoma Barcelona, 08193 Bellaterra, Spain
| | - Silvia Vidal
- Inflammatory Diseases, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (R.O.-G.); (C.G.); (M.M.); (C.Z.)
| |
Collapse
|
37
|
de Freitas DDN, Marinho Franceschina C, Muller D, Hilario GT, Gassen RB, Fazolo T, de Lima Kaminski V, Bogo Chies JA, Maito F, Antunes KH, Zanin RF, Rodrigues LC, Duarte de Souza AP. RvD1 treatment during primary infection modulates memory response increasing viral load during respiratory viral reinfection. Immunobiology 2021; 226:152151. [PMID: 34742024 DOI: 10.1016/j.imbio.2021.152151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 10/22/2021] [Accepted: 10/23/2021] [Indexed: 11/25/2022]
Abstract
Resolvin D1 (RvD1), which is biosynthesized from essential long-chain fatty acids, is involved in anti-inflammatory activity and modulation of T cell response. Memory CD8+ T cells are important for controlling tumor growth and viral infections. Exacerbated inflammation has been described as impairing memory CD8+ T cell differentiation. This study aimed to verify the effects of RvD1 on memory CD8+ T cells in vitro and in vivo in a respiratory virus infection model. Peripheral blood mononuclear cells were treated at different time points with RvD1 and stimulated with anti-CD3/anti-CD28 antibodies. Pre-treatment with RvD1 increases the expansion of memory CD8+ T cells. The IL-12 level, a cytokine described to control memory CD8+ T cells, was reduced with RvD1 pre-treatment. When the mTOR axis was inhibited, the IL-12 levels were restored. In a respiratory virus infection model, Balb/c mice were treated with RvD1 before infection or after 7 days after infection. RvD1 treatment after infection increased the frequency of memory CD8+ T cells in the lung expressing II4, II10, and Ifng. During reinfection, RvD1-treated and RSV-infected mice present a high viral load in the lung and lower antibody response in the serum. Our results show that RvD1 modulates the expansion and phenotype of memory CD8+ T cells but contributed to a non-protective response after RSV reinfection.
Collapse
Affiliation(s)
- Deise do Nascimento de Freitas
- Laboratory of Clinical and Experimental Immunology, School of Health and Life Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Caroline Marinho Franceschina
- Laboratory of Clinical and Experimental Immunology, School of Health and Life Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Douglas Muller
- Federal University of Health Sciences of Porto Alegre (UFSCPA), Porto Alegre, RS, Brazil
| | - Gabriel T Hilario
- Federal University of Health Sciences of Porto Alegre (UFSCPA), Porto Alegre, RS, Brazil
| | - Rodrigo B Gassen
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tiago Fazolo
- Federal University of Health Sciences of Porto Alegre (UFSCPA), Porto Alegre, RS, Brazil
| | - Valéria de Lima Kaminski
- Applied Immunology Laboratory, Postgraduate Program in Biotechnology, Institute of Science and Technology - ICT, Federal University of São Paulo - UNIFESP, Brazil
| | - José Artur Bogo Chies
- Laboratory of Immunogenetics and Immunobiology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fábio Maito
- Laboratory of Histology, School of Health and Life Science, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90610-000, RS, Brazil
| | - Krist Helen Antunes
- Laboratory of Clinical and Experimental Immunology, School of Health and Life Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil; Infant Center, School of Medicine PUCRS, Porto Alegre, RS, Brazil
| | - Rafael F Zanin
- Department of Health and Human Development, La Salle University, Canoas, RS, Brazil
| | - Luiz Carlos Rodrigues
- Federal University of Health Sciences of Porto Alegre (UFSCPA), Porto Alegre, RS, Brazil
| | - Ana Paula Duarte de Souza
- Laboratory of Clinical and Experimental Immunology, School of Health and Life Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil; Infant Center, School of Medicine PUCRS, Porto Alegre, RS, Brazil.
| |
Collapse
|
38
|
Martinez DR, Schäfer A, Gobeil S, Li D, De la Cruz G, Parks R, Lu X, Barr M, Stalls V, Janowska K, Beaudoin E, Manne K, Mansouri K, Edwards RJ, Cronin K, Yount B, Anasti K, Montgomery SA, Tang J, Golding H, Shen S, Zhou T, Kwong PD, Graham BS, Mascola JR, Montefiori DC, Alam SM, Sempowski GD, Khurana S, Wiehe K, Saunders KO, Acharya P, Haynes BF, Baric RS. A broadly cross-reactive antibody neutralizes and protects against sarbecovirus challenge in mice. Sci Transl Med 2021; 14:eabj7125. [PMID: 34726473 DOI: 10.1126/scitranslmed.abj7125] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- David R Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sophie Gobeil
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Dapeng Li
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Gabriela De la Cruz
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Xiaozhi Lu
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Maggie Barr
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Victoria Stalls
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Katarzyna Janowska
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Esther Beaudoin
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kartik Manne
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Katayoun Mansouri
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Robert J Edwards
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kenneth Cronin
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Boyd Yount
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kara Anasti
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Stephanie A Montgomery
- Department of Laboratory Medicine and Pathology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Juanjie Tang
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, Maryland, USA, 20871
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, Maryland, USA, 20871
| | - Shaunna Shen
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - David C Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - S Munir Alam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Gregory D Sempowski
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, Maryland, USA, 20871
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Priyamvada Acharya
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
39
|
Li Z, Wu J, Zhou J, Yuan B, Chen J, Wu W, Mo L, Qu Z, Zhou F, Dong Y, Huang K, Liu Z, Wang T, Symmes D, Gu J, Sho E, Zhang J, Chen R, Xu Y. A Vimentin-Targeting Oral Compound with Host-Directed Antiviral and Anti-Inflammatory Actions Addresses Multiple Features of COVID-19 and Related Diseases. mBio 2021; 12:e0254221. [PMID: 34634931 PMCID: PMC8510534 DOI: 10.1128/mbio.02542-21] [Citation(s) in RCA: 15] [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: 08/25/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022] Open
Abstract
Damage in COVID-19 results from both the SARS-CoV-2 virus and its triggered overactive host immune responses. Therapeutic agents that focus solely on reducing viral load or hyperinflammation fail to provide satisfying outcomes in all cases. Although viral and cellular factors have been extensively profiled to identify potential anti-COVID-19 targets, new drugs with significant efficacy remain to be developed. Here, we report the potent preclinical efficacy of ALD-R491, a vimentin-targeting small molecule compound, in treating COVID-19 through its host-directed antiviral and anti-inflammatory actions. We found that by altering the physical properties of vimentin filaments, ALD-491 affected general cellular processes as well as specific cellular functions relevant to SARS-CoV-2 infection. Specifically, ALD-R491 reduced endocytosis, endosomal trafficking, and exosomal release, thus impeding the entry and egress of the virus; increased the microcidal capacity of macrophages, thus facilitating the pathogen clearance; and enhanced the activity of regulatory T cells, therefore suppressing the overactive immune responses. In cultured cells, ALD-R491 potently inhibited the SARS-CoV-2 spike protein and human ACE2-mediated pseudoviral infection. In aged mice with ongoing, productive SARS-CoV-2 infection, ALD-R491 reduced disease symptoms as well as lung damage. In rats, ALD-R491 also reduced bleomycin-induced lung injury and fibrosis. Our results indicate a unique mechanism and significant therapeutic potential for ALD-R491 against COVID-19. We anticipate that ALD-R491, an oral, fast-acting, and non-cytotoxic agent targeting the cellular protein with multipart actions, will be convenient, safe, and broadly effective, regardless of viral mutations, for patients with early- or late-stage disease, post-COVID-19 complications, and other related diseases. IMPORTANCE With the Delta variant currently fueling a resurgence of new infections in the fully vaccinated population, developing an effective therapeutic drug is especially critical and urgent in fighting COVID-19. In contrast to the many efforts to repurpose existing drugs or address only one aspect of COVID-19, we are developing a novel agent with first-in-class mechanisms of action that address both the viral infection and the overactive immune system in the pathogenesis of the disease. Unlike virus-directed therapeutics that may lose efficacy due to viral mutations, and immunosuppressants that require ideal timing to be effective, this agent, with its unique host-directed antiviral and anti-inflammatory actions, can work against all variants of the virus, be effective during all stages of the disease, and even resolve post-disease damage and complications. Further development of the compound will provide an important tool in the fight against COVID-19 and its complications, as well as future outbreaks of new viruses.
Collapse
Affiliation(s)
- Zhizhen Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, Jiangsu, China
| | - Jianping Wu
- Laboratory Animal Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Luoda Biosciences, Inc., Chuzhou, Anhui, China
| | - Ji Zhou
- Institute of Biology and Medical Sciences, Medical School of Soochow University, Suzhou, Jiangsu, China
| | - Baoshi Yuan
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, Jiangsu, China
| | - Jiqiao Chen
- KCI Biotech (Suzhou) Inc., Suzhou, Jiangsu, China
| | - Wanchen Wu
- Joinn Laboratories (Suzhou), Co., Ltd., Suzhou, Jiangsu, China
| | - Lian Mo
- Aluda Pharmaceuticals, Inc., Menlo Park, California, USA
| | - Zhipeng Qu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, Jiangsu, China
| | - Fei Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, Jiangsu, China
| | - Yingying Dong
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, Jiangsu, China
| | - Kai Huang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, Jiangsu, China
| | - Zhiwei Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, Jiangsu, China
| | - Tao Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, Jiangsu, China
| | - Deebie Symmes
- Aluda Pharmaceuticals, Inc., Menlo Park, California, USA
| | - Jingliang Gu
- Joinn Laboratories (Suzhou), Co., Ltd., Suzhou, Jiangsu, China
| | - Eiketsu Sho
- KCI Biotech (Suzhou) Inc., Suzhou, Jiangsu, China
| | - Jingping Zhang
- Institute of Biology and Medical Sciences, Medical School of Soochow University, Suzhou, Jiangsu, China
| | - Ruihuan Chen
- Luoda Biosciences, Inc., Chuzhou, Anhui, China
- Aluda Pharmaceuticals, Inc., Menlo Park, California, USA
| | - Ying Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
40
|
Lee YS, Hong SH, Park HJ, Lee HY, Hwang JY, Kim SY, Park JW, Choi KS, Seong JK, Park SI, Lee SM, Hwang KA, Yun JW, Nam JH. Peptides Derived From S and N Proteins of Severe Acute Respiratory Syndrome Coronavirus 2 Induce T Cell Responses: A Proof of Concept for T Cell Vaccines. Front Microbiol 2021; 12:732450. [PMID: 34630356 PMCID: PMC8498111 DOI: 10.3389/fmicb.2021.732450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants that escape vaccine-induced neutralizing antibodies has indicated the importance of T cell responses against this virus. In this study, we highlight the SARS-CoV-2 epitopes that induce potent T cell responses and discuss whether T cell responses alone are adequate to confer protection against SARS-CoV-2 and describe the administration of 20 peptides with an RNA adjuvant in mice. The peptides have been synthesized based on SARS-CoV-2 spike and nucleocapsid protein sequences. Our study demonstrates that immunization with these peptides significantly increases the proportion of effector memory T cell population and interferon-γ (IFN-γ)-, interleukin-4 (IL-4)-, tumor necrosis factor-α (TNF-α)-, and granzyme B-producing T cells. Of these 20 peptides, four induce the generation of IFN-γ-producing T cells, elicit CD8+ T cell (CTL) responses in a dose-dependent manner, and induce cytotoxic T lymphocytes that eliminate peptide-pulsed target cells in vivo. Although it is not statistically significant, these peptide vaccines reduce viral titers in infected hamsters and alleviate pulmonary pathology in SARS-CoV-2-infected human ACE2 transgenic mice. These findings may aid the design of effective SARS-CoV-2 peptide vaccines, while providing insights into the role of T cells in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yu-Sun Lee
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, South Korea
| | - So-Hee Hong
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, South Korea
| | - Hyo-Jung Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, South Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul, South Korea
| | - Ji-Yeon Hwang
- Preclinical Research Center, Seoul National University Bundang Hospital, Seoul, South Korea
| | - Seo Yeon Kim
- Preclinical Research Center, Seoul National University Bundang Hospital, Seoul, South Korea
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, South Korea
| | - Kang-Seuk Choi
- BK21 Program for Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea.,Laboratory of Avian Diseases, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea.,Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea.,Interdisciplinary Program for Bioinformatics, Program for Cancer Biology and BIO-MAX/N-Bio Institute, Seoul National University, Seoul, South Korea
| | - Sang-In Park
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Sang-Myeong Lee
- College of Veterinary Medicine, Chungbuk National University, Cheongju, South Korea
| | - Kyung-Ah Hwang
- Department of Research and Development, SML Genetree, Seoul, South Korea
| | - Jun-Won Yun
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, South Korea.,BK21 PLUS Program, The Catholic University of Korea, Bucheon, South Korea
| | - Jae-Hwan Nam
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, South Korea.,BK21 PLUS Program, The Catholic University of Korea, Bucheon, South Korea
| |
Collapse
|
41
|
Taoufik Y, de Goër de Herve MG, Corgnac S, Durrbach A, Mami-Chouaib F. When Immunity Kills: The Lessons of SARS-CoV-2 Outbreak. Front Immunol 2021; 12:692598. [PMID: 34630382 PMCID: PMC8497820 DOI: 10.3389/fimmu.2021.692598] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 09/02/2021] [Indexed: 01/08/2023] Open
Abstract
Since its emergence at the end of 2019, SARS-CoV-2 has spread worldwide at a very rapid pace. While most infected individuals have an asymptomatic or mild disease, a minority, mainly the elderly, develop a severe disease that may lead to a fatal acute respiratory distress syndrome (ARDS). ARDS results from a highly inflammatory immunopathology process that includes systemic manifestations and massive alveolar damages that impair gas exchange. The present review summarizes our current knowledge in the rapidly evolving field of SARS-CoV-2 immunopathology, emphasizing the role of specific T cell responses. Indeed, accumulating evidence suggest that while T-cell response directed against SARS-CoV-2 likely plays a crucial role in virus clearance, it may also participate in the immunopathology process that leads to ARDS.
Collapse
Affiliation(s)
- Yassine Taoufik
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR) 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France.,Department of Hematology and Immunology, Assistance Publique - Hôpitaux de Paris, Université Paris-Saclay, le Kremlin-Bicêtre, France
| | - Marie-Ghislaine de Goër de Herve
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR) 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France.,Department of Hematology and Immunology, Assistance Publique - Hôpitaux de Paris, Université Paris-Saclay, le Kremlin-Bicêtre, France.,Department of Nephrology, Assistance Publique - Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Créteil, France
| | - Stéphanie Corgnac
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR) 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Antoine Durrbach
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR) 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France.,Department of Nephrology, Assistance Publique - Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Créteil, France
| | - Fathia Mami-Chouaib
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR) 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
42
|
Zhou P, Chen J, He J, Zheng T, Yunis J, Makota V, Alexandre YO, Gong F, Zhang X, Xie W, Li Y, Shao M, Zhu Y, Sinclair JE, Miao M, Chen Y, Short KR, Mueller SN, Sun X, Yu D, Li Z. Low-dose IL-2 therapy invigorates CD8+ T cells for viral control in systemic lupus erythematosus. PLoS Pathog 2021; 17:e1009858. [PMID: 34618873 PMCID: PMC8525737 DOI: 10.1371/journal.ppat.1009858] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 10/19/2021] [Accepted: 07/30/2021] [Indexed: 01/10/2023] Open
Abstract
Autoimmune diseases are often treated by glucocorticoids and immunosuppressive drugs that could increase the risk for infection, which in turn deteriorate disease and cause mortality. Low-dose IL-2 (Ld-IL2) therapy emerges as a new treatment for a wide range of autoimmune diseases. To examine its influence on infection, we retrospectively studied 665 patients with systemic lupus erythematosus (SLE) including about one third receiving Ld-IL2 therapy, where Ld-IL2 therapy was found beneficial in reducing the incidence of infections. In line with this clinical observation, IL-2 treatment accelerated viral clearance in mice infected with influenza A virus or lymphocytic choriomeningitis virus (LCMV). Noticeably, despite enhancing anti-viral immunity in LCMV infection, IL-2 treatment exacerbated CD8+ T cell-mediated immunopathology. In summary, Ld-IL2 therapy reduced the risk of infections in SLE patients and enhanced the control of viral infection, but caution should be taken to avoid potential CD8+ T cell-mediated immunopathology.
Collapse
Affiliation(s)
- Pengcheng Zhou
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Jiali Chen
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Jing He
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Ting Zheng
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Joseph Yunis
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Victor Makota
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Yannick O. Alexandre
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Fang Gong
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xia Zhang
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Wuxiang Xie
- Peking University Clinical Research Institute, Peking University Health Science Center, Beijing, China
| | - Yuhui Li
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Miao Shao
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yanshan Zhu
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Jane E. Sinclair
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Miao Miao
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Kirsty R. Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Scott N. Mueller
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Di Yu
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
43
|
Looman KIM, Cecil CAM, Grosserichter‐Wagener C, Kiefte‐de Jong JC, van Zelm MC, Moll HA. Associations between T cells and attention problems in the general pediatric population: The Generation R study. JCPP ADVANCES 2021; 1:e12038. [PMID: 37431441 PMCID: PMC10242894 DOI: 10.1002/jcv2.12038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 08/23/2021] [Indexed: 11/08/2022] Open
Abstract
Objective The pathogenesis of attention-deficit/hyperactivity disorder (ADHD) is currently unclear. We hypothesized that chronic immune activation, as indexed by T and B cells, plays a role in the pathophysiology of attention problems. Therefore, we examined T and B cell subsets in a general pediatric population with information on attention problems. Methods We included 756 10-year-old children from the Generation R population-based cohort. Eleven-color flow cytometry was performed on peripheral blood samples to determine T and B cell subsets. The Child Behavior Checklist rated by parents was used to measure attention problems. Data were analyzed using linear regression analyses, adjusting for maternal and child covariates and co-occurring childhood psychopathology. Results For T helper 1 (Th1) cells, one standard deviation (SD) increase was associated with 5.3% (95%CI 0.3; 10.5) higher attention problem scores. Furthermore, 1SD increase in CD8+ T cells was associated with 7.5% (95%CI 2.4; 12.7) higher attention problem scores. Within total CD8+ T cells, 1SD increase in naive or central memory cells was associated with 6.9% (95%CI 2.0; 12.1) and 6.4% (95%CI 1.5; 11.6) higher attention problem scores, respectively. No associations between Th2, Treg or B memory cells and attention problem scores were observed. Conclusion Higher Th1 and cytotoxic T cell numbers are associated with higher attention problem scores independent of co-occurring psychopathology. This might indicate a possible role of a pro-inflammatory immune profile in childhood attention problems.
Collapse
Affiliation(s)
- Kirsten I. M. Looman
- Generation R Study GroupErasmus MCUniversity Medical CenterRotterdamThe Netherlands
- Department of PediatricsSophia Children's HospitalErasmus MCUniversity Medical CenterRotterdamThe Netherlands
| | - Charlotte A. M. Cecil
- Department of Child and Adolescent Psychiatry/PsychologyErasmus MCUniversity Medical CenterRotterdamThe Netherlands
- Department of EpidemiologyErasmus MCUniversity Medical CenterRotterdamThe Netherlands
- Molecular EpidemiologyDepartment of Biomedical Data SciencesLeiden University Medical CenterLeidenThe Netherlands
| | | | - Jessica C. Kiefte‐de Jong
- Department of EpidemiologyErasmus MCUniversity Medical CenterRotterdamThe Netherlands
- Department of Public Health and Primary Care/LUMC Campus The HagueLeiden University Medical CenterLeidenThe Netherlands
| | - Menno C. van Zelm
- Department of Immunology and PathologyCentral Clinical SchoolMonash University and Alfred HospitalMelbourneVictoriaAustralia
| | - Henriëtte A. Moll
- Department of PediatricsSophia Children's HospitalErasmus MCUniversity Medical CenterRotterdamThe Netherlands
| |
Collapse
|
44
|
Knight AC, Montgomery SA, Fletcher CA, Baxter VK. Mouse Models for the Study of SARS-CoV-2 Infection. Comp Med 2021; 71:383-397. [PMID: 34610856 PMCID: PMC8594264 DOI: 10.30802/aalas-cm-21-000031] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/19/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023]
Abstract
Mice are an invaluable resource for studying virus-induced disease. They are a small, genetically modifiable animal for which a large arsenal of genetic and immunologic tools is available for evaluation of pathogenesis and potential vaccines and therapeutics. SARS-CoV-2, the betacoronavirus responsible for the COVID-19 pandemic, does not naturally replicate in wild-type mice, due to structural differences between human and mouse ACE2, the primary receptor for SARS-CoV-2 entry into cells. However, several mouse strains have been developed that allow for SARS-CoV-2 replication and clinical disease. Two broad strategies have primarily been deployed for developing mouse strains susceptible to COVID-19-like disease: adding in the human ACE2 gene and adapting the virus to the mouse ACE2 receptor. Both approaches result in mice that develop several of the clinical and pathologic hallmarks of COVID-19, including acute respiratory distress syndrome and acute lung injury. In this review, we describe key acute pulmonary and extrapulmonary pathologic changes seen in COVID-19 patients that mouse models of SARS-CoV-2 infection ideally replicate, the essential development of mouse models for the study of Severe Acute Respiratory Syndrome and Middle Eastern Respiratory Syndrome and the basis of many of the models of COVID-19, and key clinical and pathologic features of currently available mouse models of SARS-CoV-2 infection.
Collapse
Key Words
- aav, adeno-associated virus
- ace2, angiotensin-converting enzyme 2
- ali, acute lung injury
- ards, acute respiratory distress syndrome
- covid-19, coronavirus disease 19
- dad, diffuse alveolar damage
- dpi, days postinfection
- dpp4, dipeptidyl peptidase 4
- hace2, human angiotensin-converting enzyme 2
- mace2, mouse angiotensin-converting enzyme 2
- mers, middle eastern respiratory syndrome
- mers-cov, middle eastern respiratory syndrome coronavirus
- sars, severe acute respiratory syndrome
- sars-cov, severe acute respiratory syndrome coronavirus
- sars-cov-2, severe acute respiratory syndrome coronavirus 2
Collapse
Affiliation(s)
- Audrey C Knight
- Department of Pathology and Laboratory Medicine
- Institute for Global Health and Infectious Diseases, and
| | - Stephanie A Montgomery
- Department of Pathology and Laboratory Medicine
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Craig A Fletcher
- Department of Pathology and Laboratory Medicine
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Victoria K Baxter
- Department of Pathology and Laboratory Medicine
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
45
|
Geng Q, Tai W, Baxter VK, Shi J, Wan Y, Zhang X, Montgomery SA, Taft-Benz SA, Anderson EJ, Knight AC, Dinnon KH, Leist SR, Baric RS, Shang J, Hong SW, Drelich A, Tseng CTK, Jenkins M, Heise M, Du L, Li F. Novel virus-like nanoparticle vaccine effectively protects animal model from SARS-CoV-2 infection. PLoS Pathog 2021; 17:e1009897. [PMID: 34492082 PMCID: PMC8448314 DOI: 10.1371/journal.ppat.1009897] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 09/17/2021] [Accepted: 08/16/2021] [Indexed: 11/18/2022] Open
Abstract
The key to battling the COVID-19 pandemic and its potential aftermath is to develop a variety of vaccines that are efficacious and safe, elicit lasting immunity, and cover a range of SARS-CoV-2 variants. Recombinant viral receptor-binding domains (RBDs) are safe vaccine candidates but often have limited efficacy due to the lack of virus-like immunogen display pattern. Here we have developed a novel virus-like nanoparticle (VLP) vaccine that displays 120 copies of SARS-CoV-2 RBD on its surface. This VLP-RBD vaccine mimics virus-based vaccines in immunogen display, which boosts its efficacy, while maintaining the safety of protein-based subunit vaccines. Compared to the RBD vaccine, the VLP-RBD vaccine induced five times more neutralizing antibodies in mice that efficiently blocked SARS-CoV-2 from attaching to its host receptor and potently neutralized the cell entry of variant SARS-CoV-2 strains, SARS-CoV-1, and SARS-CoV-1-related bat coronavirus. These neutralizing immune responses induced by the VLP-RBD vaccine did not wane during the two-month study period. Furthermore, the VLP-RBD vaccine effectively protected mice from SARS-CoV-2 challenge, dramatically reducing the development of clinical signs and pathological changes in immunized mice. The VLP-RBD vaccine provides one potentially effective solution to controlling the spread of SARS-CoV-2.
Collapse
Affiliation(s)
- Qibin Geng
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota, United States of America
- Center for Coronavirus Research, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Wanbo Tai
- Laboratory of Viral Immunology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Victoria K. Baxter
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Division of Comparative Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Juan Shi
- Laboratory of Viral Immunology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Yushun Wan
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota, United States of America
- Center for Coronavirus Research, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Xiujuan Zhang
- Laboratory of Viral Immunology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Stephanie A. Montgomery
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Sharon A. Taft-Benz
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Elizabeth J. Anderson
- Division of Comparative Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Audrey C. Knight
- Division of Comparative Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Kenneth H. Dinnon
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Sarah R. Leist
- Rapidly Emerging Antiviral Drug Development Initiative, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Ralph S. Baric
- Division of Comparative Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Rapidly Emerging Antiviral Drug Development Initiative, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Jian Shang
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota, United States of America
- Center for Coronavirus Research, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Sung-Wook Hong
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Aleksandra Drelich
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Chien-Te K. Tseng
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Marc Jenkins
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Mark Heise
- Division of Comparative Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Rapidly Emerging Antiviral Drug Development Initiative, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Lanying Du
- Laboratory of Viral Immunology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Fang Li
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota, United States of America
- Center for Coronavirus Research, University of Minnesota, Saint Paul, Minnesota, United States of America
| |
Collapse
|
46
|
Martinez DR, Schäfer A, Leist SR, De la Cruz G, West A, Atochina-Vasserman EN, Lindesmith LC, Pardi N, Parks R, Barr M, Li D, Yount B, Saunders KO, Weissman D, Haynes BF, Montgomery SA, Baric RS. Chimeric spike mRNA vaccines protect against Sarbecovirus challenge in mice. Science 2021; 373:991-998. [PMID: 34214046 PMCID: PMC8899822 DOI: 10.1126/science.abi4506] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022]
Abstract
The emergence of severe acute respiratory syndrome coronavirus (SARS-CoV) in 2003 and SARS-CoV-2 in 2019 highlights the need to develop universal vaccination strategies against the broader Sarbecovirus subgenus. Using chimeric spike designs, we demonstrate protection against challenge from SARS-CoV, SARS-CoV-2, SARS-CoV-2 B.1.351, bat CoV (Bt-CoV) RsSHC014, and a heterologous Bt-CoV WIV-1 in vulnerable aged mice. Chimeric spike messenger RNAs (mRNAs) induced high levels of broadly protective neutralizing antibodies against high-risk Sarbecoviruses. By contrast, SARS-CoV-2 mRNA vaccination not only showed a marked reduction in neutralizing titers against heterologous Sarbecoviruses, but SARS-CoV and WIV-1 challenge in mice resulted in breakthrough infections. Chimeric spike mRNA vaccines efficiently neutralized D614G, mink cluster five, and the UK B.1.1.7 and South African B.1.351 variants of concern. Thus, multiplexed-chimeric spikes can prevent SARS-like zoonotic coronavirus infections with pandemic potential.
Collapse
Affiliation(s)
- David R Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah R Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabriela De la Cruz
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Ande West
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elena N Atochina-Vasserman
- Infectious Disease Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Lisa C Lindesmith
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Norbert Pardi
- Infectious Disease Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Maggie Barr
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Dapeng Li
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Boyd Yount
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Drew Weissman
- Infectious Disease Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Stephanie A Montgomery
- Department of Laboratory Medicine and Pathology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
47
|
Martinez DR, Schäfer A, Leist SR, Li D, Gully K, Yount B, Feng JY, Bunyan E, Porter DP, Cihlar T, Montgomery SA, Haynes BF, Baric RS, Nussenzweig MC, Sheahan TP. Prevention and therapy of SARS-CoV-2 and the B.1.351 variant in mice. Cell Rep 2021; 36:109450. [PMID: 34289384 PMCID: PMC8270748 DOI: 10.1016/j.celrep.2021.109450] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/22/2021] [Accepted: 07/02/2021] [Indexed: 01/08/2023] Open
Abstract
Improving clinical care for individuals infected with SARS-CoV-2 variants is a global health priority. Small-molecule antivirals like remdesivir (RDV) and biologics such as human monoclonal antibodies (mAbs) have demonstrated therapeutic efficacy against SARS-CoV-2, the causative agent of coronavirus disease 2019 (COVID-19). It is not known whether combination RDV/mAb will improve outcomes over single-agent therapies or whether antibody therapies will remain efficacious against variants. Here, we show that a combination of two mAbs in clinical trials, C144 and C135, have potent antiviral effects against even when initiated 48 h after infection and have therapeutic efficacy in vivo against the B.1.351 variant of concern (VOC). Combining RDV and antibodies provided a modest improvement in outcomes compared with single agents. These data support the continued use of RDV to treat SARS-CoV-2 infections and the continued clinical development of the C144 and C135 antibody combination to treat patients infected with SARS-CoV-2 variants.
Collapse
Affiliation(s)
- David R Martinez
- Department of Epidemiology, READDI Initiative, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Alexandra Schäfer
- Department of Epidemiology, READDI Initiative, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah R Leist
- Department of Epidemiology, READDI Initiative, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dapeng Li
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA
| | - Kendra Gully
- Department of Epidemiology, READDI Initiative, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Boyd Yount
- Department of Epidemiology, READDI Initiative, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joy Y Feng
- Gilead Sciences, Inc., Foster City, CA, USA
| | | | | | | | - Stephanie A Montgomery
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA
| | - Ralph S Baric
- Department of Epidemiology, READDI Initiative, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michel C Nussenzweig
- The Rockefeller University, New York, NY, USA; The Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Timothy P Sheahan
- Department of Epidemiology, READDI Initiative, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
48
|
Stephens LM, Varga SM. Considerations for a Respiratory Syncytial Virus Vaccine Targeting an Elderly Population. Vaccines (Basel) 2021; 9:vaccines9060624. [PMID: 34207770 PMCID: PMC8228432 DOI: 10.3390/vaccines9060624] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/22/2022] Open
Abstract
Respiratory syncytial virus (RSV) is most commonly associated with acute lower respiratory tract infections in infants and children. However, RSV also causes a high disease burden in the elderly that is often under recognized. Adults >65 years of age account for an estimated 80,000 RSV-associated hospitalizations and 14,000 deaths in the United States annually. RSV infection in aged individuals can result in more severe disease symptoms including pneumonia and bronchiolitis. Given the large disease burden caused by RSV in the aged, this population remains an important target for vaccine development. Aging results in lowered immune responsiveness characterized by impairments in both innate and adaptive immunity. This immune senescence poses a challenge when developing a vaccine targeting elderly individuals. An RSV vaccine tailored towards an elderly population will need to maximize the immune response elicited in order to overcome age-related defects in the immune system. In this article, we review the hurdles that must be overcome to successfully develop an RSV vaccine for use in the elderly, and discuss the vaccine candidates currently being tested in this highly susceptible population.
Collapse
Affiliation(s)
- Laura M. Stephens
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA;
| | - Steven M. Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA;
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
- Correspondence:
| |
Collapse
|
49
|
Weerts EAWS, Matthijs MGR, Bonhof J, van Haarlem DA, Dwars RM, Gröne A, Verheije MH, Jansen CA. The contribution of the immune response to enhanced colibacillosis upon preceding viral respiratory infection in broiler chicken in a dual infection model. Vet Immunol Immunopathol 2021; 238:110276. [PMID: 34126552 DOI: 10.1016/j.vetimm.2021.110276] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 05/07/2021] [Accepted: 05/25/2021] [Indexed: 01/10/2023]
Abstract
Colibacillosis in chickens caused by avian pathogenic Escherichia coli (APEC) is known to be aggravated by preceding infections with infectious bronchitis virus (IBV), Newcastle disease virus (NDV) and avian metapneumovirus (aMPV). The mechanism behind these virus-induced predispositions for secondary bacterial infections is poorly understood. Here we set out to investigate the immunopathogenesis of enhanced respiratory colibacillosis after preceding infections with these three viruses. Broilers were inoculated intratracheally with APEC six days after oculonasal and intratracheal inoculation with IBV, NDV, aMPV or buffered saline. After euthanasia at 1 and 8 days post infection (dpi) with APEC, birds were macroscopically examined and tissue samples were taken from the trachea, lungs and air sacs. In none of the groups differences in body weight were observed during the course of infection. Macroscopic lesion scoring revealed most severe tissue changes after NDV-APEC and IBV-APEC infection. Histologically, persistent tracheitis was detected in all virus-APEC groups, but not after APEC-only infection. In the lungs, mostly APEC-associated transient pneumonia was observed. Severe and persistent airsacculitis was present after NDV-APEC and IBV-APEC infection. Bacterial antigen was detected by immunohistochemistry only at 1 dpi APEC, predominantly in NDV-APEC- and IBV-APEC-infected lungs. Higher numbers of CD4+ and CD8+ lymphocytes persisted over time in NDV-APEC- and IBV-APEC-infected tracheas, as did CD4+ lymphocytes in NBV-APEC- and IBV-APEC-infected air sacs. KUL01+ cells, which include monocytes and macrophages, and TCRγδ+ lymphocytes were observed mostly in lung tissue in all infected groups with transient higher numbers of KUL01+ cells over time and higher numbers of TCRγδ+ lymphocytes mainly at 8 dpi. qPCR analysis revealed mostly trends of transient higher levels of IL-6 and IFNγ mRNA in lung tissue after IBV-APEC and also NDV-APEC infection and persistent higher levels of IL-6 mRNA after aMPV-APEC infection. In spleens, transient higher levels of IL-17 mRNA and more persistent higher levels of IL-6 mRNA were observed after all co-infections. No changes in IL-10 mRNA expression were seen. These results demonstrate a major impact of dual infections with respiratory viruses and APEC, compared to a single infection with APEC, on the chicken respiratory tract and suggest that immunopathogenesis contributes to lesion persistence.
Collapse
Affiliation(s)
- Erik A W S Weerts
- Division of Pathology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands.
| | - Mieke G R Matthijs
- Division of Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 7, 3584 CL Utrecht, the Netherlands
| | - Josette Bonhof
- Division of Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Daphne A van Haarlem
- Division of Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - R Marius Dwars
- Division of Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 7, 3584 CL Utrecht, the Netherlands
| | - Andrea Gröne
- Division of Pathology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - M Hélène Verheije
- Division of Pathology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Christine A Jansen
- Division of Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| |
Collapse
|
50
|
Martinez DR, Schäfer A, Leist SR, De la Cruz G, West A, Atochina-Vasserman EN, Lindesmith LC, Pardi N, Parks R, Barr M, Li D, Yount B, Saunders KO, Weissman D, Haynes BF, Montgomery SA, Baric RS. Chimeric spike mRNA vaccines protect against Sarbecoviru s challenge in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.03.11.434872. [PMID: 33758837 PMCID: PMC7986996 DOI: 10.1101/2021.03.11.434872] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The emergence of SARS-CoV in 2003 and SARS-CoV-2 in 2019 highlights the need to develop universal vaccination strategies against the broader Sarbecovirus subgenus. Using chimeric spike designs, we demonstrate protection against challenge from SARS-CoV, SARS-CoV-2, SARS-CoV-2 B.1.351, bat CoV (Bt-CoV) RsSHC014, and a heterologous Bt-CoV WIV-1 in vulnerable aged mice. Chimeric spike mRNAs induced high levels of broadly protective neutralizing antibodies against high-risk Sarbecoviruses. In contrast, SARS-CoV-2 mRNA vaccination not only showed a marked reduction in neutralizing titers against heterologous Sarbecoviruses, but SARS-CoV and WIV-1 challenge in mice resulted in breakthrough infection. Chimeric spike mRNA vaccines efficiently neutralized D614G, UK B.1.1.7., mink cluster five, and the South African B.1.351 variant of concern. Thus, multiplexed-chimeric spikes can prevent SARS-like zoonotic coronavirus infections with pandemic potential.
Collapse
Affiliation(s)
- David R. Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah R. Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabriela De la Cruz
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Ande West
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elena N. Atochina-Vasserman
- Infectious Disease Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Lisa C. Lindesmith
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Norbert Pardi
- Infectious Disease Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Maggie Barr
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Dapeng Li
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Boyd Yount
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kevin O. Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Drew Weissman
- Infectious Disease Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Barton F. Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Stephanie A. Montgomery
- Department of Laboratory Medicine and Pathology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|