51
|
Scarpa F, Sernicola L, Farcomeni S, Ciccozzi A, Sanna D, Casu M, Vitale M, Cicenia A, Giovanetti M, Romano C, Branda F, Ciccozzi M, Borsetti A. Phylodynamic and Evolution of the Hemagglutinin (HA) and Neuraminidase (NA) Genes of Influenza A(H1N1) pdm09 Viruses Circulating in the 2009 and 2023 Seasons in Italy. Pathogens 2024; 13:334. [PMID: 38668289 PMCID: PMC11054071 DOI: 10.3390/pathogens13040334] [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: 03/29/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024] Open
Abstract
The influenza A(H1N1) pdm09 virus, which emerged in 2009, has been circulating seasonally since then. In this study, we conducted a comprehensive genome-based investigation to gain a detailed understanding of the genetic and evolutionary characteristics of the hemagglutinin (HA) and neuraminidase (NA) surface proteins of A/H1N1pdm09 strains circulating in Italy over a fourteen-year period from 2009 to 2023 in relation to global strains. Phylogenetic analysis revealed rapid transmission and diversification of viral variants during the early pandemic that clustered in clade 6B.1. In contrast, limited genetic diversity was observed during the 2023 season, probably due to the genetic drift, which provides the virus with a constant adaptability to the host; furthermore, all isolates were split into two main groups representing two clades, i.e., 6B.1A.5a.2a and its descendant 6B.1A.5a.2a.1. The HA gene showed a faster rate of evolution compared to the NA gene. Using FUBAR, we identified positively selected sites 41 and 177 for HA and 248, 286, and 455 for NA in 2009, as well as sites 22, 123, and 513 for HA and 339 for NA in 2023, all of which may be important sites related to the host immune response. Changes in glycosylation acquisition/loss at prominent sites, i.e., 177 in HA and 248 in NA, should be considered as a predictive tool for early warning signs of emerging pandemics, and for vaccine and drug development.
Collapse
Affiliation(s)
- Fabio Scarpa
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (D.S.)
| | - Leonardo Sernicola
- National HIV/AIDS Research Center (CNAIDS), Istituto Superiore di Sanità, 00162 Rome, Italy; (L.S.); (S.F.)
| | - Stefania Farcomeni
- National HIV/AIDS Research Center (CNAIDS), Istituto Superiore di Sanità, 00162 Rome, Italy; (L.S.); (S.F.)
| | - Alessandra Ciccozzi
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (D.S.)
| | - Daria Sanna
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (D.S.)
| | - Marco Casu
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy;
| | - Marco Vitale
- Laboratorio di Biologia Molecolare—Fondazione Università Niccolò Cusano, 00166 Rome, Italy; (M.V.); (A.C.)
| | - Alessia Cicenia
- Laboratorio di Biologia Molecolare—Fondazione Università Niccolò Cusano, 00166 Rome, Italy; (M.V.); (A.C.)
| | - Marta Giovanetti
- Department of Sciences and Technologies for Sustainable Development and One Health, Universita Campus Bio-Medico di Roma, 00128 Rome, Italy;
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-009, MG, Brazil
- Climate Amplified Diseases and Epidemics (CLIMADE), Brasilia 70070-130, DF, Brazil
| | - Chiara Romano
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, 00128 Rome, Italy; (C.R.); (F.B.); (M.C.)
| | - Francesco Branda
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, 00128 Rome, Italy; (C.R.); (F.B.); (M.C.)
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, 00128 Rome, Italy; (C.R.); (F.B.); (M.C.)
| | - Alessandra Borsetti
- National HIV/AIDS Research Center (CNAIDS), Istituto Superiore di Sanità, 00162 Rome, Italy; (L.S.); (S.F.)
| |
Collapse
|
52
|
Wu W, Arunagiri V, Do-Umehara HC, Chen C, Gu S, Biswas I, Ridge KM, Budinger GRS, Liu S, Liu J. Miz1 represses type I interferon production and limits viral clearance during influenza A virus infection. Sci Signal 2024; 17:eadg7867. [PMID: 38593156 PMCID: PMC11182629 DOI: 10.1126/scisignal.adg7867] [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: 01/20/2023] [Accepted: 03/25/2024] [Indexed: 04/11/2024]
Abstract
Type I interferons (IFNs) are critical for the antiviral immune response, and fine-tuning type I IFN production is critical to effectively clearing viruses without causing harmful immunopathology. We showed that the transcription factor Miz1 epigenetically repressed the expression of genes encoding type I IFNs in mouse lung epithelial cells by recruiting histone deacetylase 1 (HDAC1) to the promoters of Ifna and Ifnb. Loss of function of Miz1 resulted in augmented production of these type I IFNs during influenza A virus (IAV) infection, leading to improved viral clearance in vitro and in vivo. IAV infection induced Miz1 accumulation by promoting the cullin-4B (CUL4B)-mediated ubiquitylation and degradation of the E3 ubiquitin ligase Mule (Mcl-1 ubiquitin ligase E3; also known as Huwe1 or Arf-BP1), which targets Miz1 for degradation. As a result, Miz1 accumulation limited type I IFN production and favored viral replication. This study reveals a previously unrecognized function of Miz1 in regulating antiviral defense and a potential mechanism for influenza viruses to evade host immune defense.
Collapse
Affiliation(s)
- Wenjiao Wu
- Department of Surgery, College of Medicine; Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Department of Pharmacy, Guangdong Second Provincial General Hospital, 466 Middle Xingang Road, Guangzhou, 510317, Guangdong, China
| | - Vinothini Arunagiri
- Department of Surgery, College of Medicine; Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Hanh Chi Do-Umehara
- Department of Surgery, College of Medicine; Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Cong Chen
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Shuyin Gu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Indrani Biswas
- Department of Surgery, College of Medicine; Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Karen M. Ridge
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - G. R. Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou 510515, China
| | - Jing Liu
- Department of Surgery, College of Medicine; Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
53
|
Lou J, Liang W, Cao L, Hu I, Zhao S, Chen Z, Chan RWY, Cheung PPH, Zheng H, Liu C, Li Q, Chong MKC, Zhang Y, Yeoh EK, Chan PKS, Zee BCY, Mok CKP, Wang MH. Predictive evolutionary modelling for influenza virus by site-based dynamics of mutations. Nat Commun 2024; 15:2546. [PMID: 38514647 PMCID: PMC10958014 DOI: 10.1038/s41467-024-46918-0] [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: 12/06/2023] [Accepted: 03/12/2024] [Indexed: 03/23/2024] Open
Abstract
Influenza virus continuously evolves to escape human adaptive immunity and generates seasonal epidemics. Therefore, influenza vaccine strains need to be updated annually for the upcoming flu season to ensure vaccine effectiveness. We develop a computational approach, beth-1, to forecast virus evolution and select representative virus for influenza vaccine. The method involves modelling site-wise mutation fitness. Informed by virus genome and population sero-positivity, we calibrate transition time of mutations and project the fitness landscape to future time, based on which beth-1 selects the optimal vaccine strain. In season-to-season prediction in historical data for the influenza A pH1N1 and H3N2 viruses, beth-1 demonstrates superior genetic matching compared to existing approaches. In prospective validations, the model shows superior or non-inferior genetic matching and neutralization against circulating virus in mice immunization experiments compared to the current vaccine. The method offers a promising and ready-to-use tool to facilitate vaccine strain selection for the influenza virus through capturing heterogeneous evolutionary dynamics over genome space-time and linking molecular variants to population immune response.
Collapse
Affiliation(s)
- Jingzhi Lou
- JC School of Public Health and Primary Care (JCSPHPC), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Beth Bioinformatics Co. Ltd, Hong Kong SAR, China
| | - Weiwen Liang
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lirong Cao
- JC School of Public Health and Primary Care (JCSPHPC), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Inchi Hu
- Department of Statistics, George Mason University, Fairfax, VA, USA
| | - Shi Zhao
- JC School of Public Health and Primary Care (JCSPHPC), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - Zigui Chen
- Department of Microbiology, CUHK, Hong Kong SAR, China
| | - Renee Wan Yi Chan
- Department of Paediatrics, CUHK, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence, CUHK, Hong Kong SAR, China
| | | | - Hong Zheng
- JC School of Public Health and Primary Care (JCSPHPC), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Caiqi Liu
- JC School of Public Health and Primary Care (JCSPHPC), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Qi Li
- JC School of Public Health and Primary Care (JCSPHPC), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Marc Ka Chun Chong
- JC School of Public Health and Primary Care (JCSPHPC), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Yexian Zhang
- Beth Bioinformatics Co. Ltd, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Eng-Kiong Yeoh
- JC School of Public Health and Primary Care (JCSPHPC), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Centre for Health Systems and Policy Research, CUHK, Hong Kong SAR, China
| | - Paul Kay-Sheung Chan
- Department of Microbiology, CUHK, Hong Kong SAR, China
- Stanley Ho Centre for Emerging Infectious Diseases, CUHK, Hong Kong SAR, China
| | - Benny Chung Ying Zee
- JC School of Public Health and Primary Care (JCSPHPC), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Chris Ka Pun Mok
- JC School of Public Health and Primary Care (JCSPHPC), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China.
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, CUHK, Hong Kong SAR, China.
| | - Maggie Haitian Wang
- JC School of Public Health and Primary Care (JCSPHPC), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China.
- CUHK Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
54
|
Pavia G, Scarpa F, Ciccozzi A, Romano C, Branda F, Quirino A, Marascio N, Matera G, Sanna D, Ciccozzi M. Changing and Evolution of Influenza Virus: Is It a Trivial Flu? Chemotherapy 2024; 69:185-193. [PMID: 38508151 DOI: 10.1159/000538382] [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: 02/24/2024] [Accepted: 03/09/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Influenza viruses are etiological agents which cause contagious respiratory, seasonal epidemics and, for influenza A subtypes, pandemics. The clinical picture of influenza has undergone continuous change over the years, due to intrinsic viral evolution as well as "reassortment" of its genomic segments. The history of influenza highlights its ability to adapt and to rapidly evolve, without specific circumstances. This reflects the complexity of this pathology and poses the fundamental question about its assumption as a "common illness" and its impact on public health. SUMMARY The global influenza epidemics and pandemics claimed millions of deaths, leaving an indelible mark on public health and showing the need for a better comprehension of the influenza virus. The clear understanding of genetic variations during the influenza seasonal epidemics is a crucial point for developing effective strategies for prevention, treatment, and vaccine design. The recent advance in next-generation sequencing approaches, model systems to virus culture, and bioinformatics pipeline played a key role in the rapid characterization of circulating influenza strains. In particular, the increase in computational power allowed the performance of complex tasks in healthcare settings through machine learning algorithms, which analyze different variables, such as medical and laboratory outputs, to optimize medical research and improve public health systems. The early detection of emerging and reemerging pathogens is a matter of importance to prevent future pandemics. KEY MESSAGES The perception of influenza as a "trivial flu" or a more serious public health concern is a subject of ongoing debate, reflecting the multifaceted nature of this infectious disease. The variability in the severity of influenza sheds light on the unpredictability of the viral characteristics, coupled with the challenges in accurately predicting circulating strains. This adds complexity to the public health burden of influenza and highlights the need for targeted interventions.
Collapse
Affiliation(s)
- Grazia Pavia
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, Catanzaro, Italy
| | - Fabio Scarpa
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | - Chiara Romano
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Francesco Branda
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Angela Quirino
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, Catanzaro, Italy
| | - Nadia Marascio
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, Catanzaro, Italy
| | - Giovanni Matera
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, Catanzaro, Italy
| | - Daria Sanna
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
55
|
Chen C, Yang M, Wang Y, Jiang D, Du Y, Cao K, Zhang X, Wu X, Chen M, You Y, Zhou W, Qi J, Yan R, Zhu C, Yang S. Intensity and drivers of subtypes interference between seasonal influenza viruses in mainland China: A modeling study. iScience 2024; 27:109323. [PMID: 38487011 PMCID: PMC10937832 DOI: 10.1016/j.isci.2024.109323] [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: 09/01/2023] [Revised: 01/18/2024] [Accepted: 02/20/2024] [Indexed: 03/17/2024] Open
Abstract
Subtype interference has a significant impact on the epidemiological patterns of seasonal influenza viruses (SIVs). We used attributable risk percent [the absolute value of the ratio of the effective reproduction number (Rₑ) of different subtypes minus one] to quantify interference intensity between A/H1N1 and A/H3N2, as well as B/Victoria and B/Yamagata. The interference intensity between A/H1N1 and A/H3N2 was higher in southern China 0.26 (IQR: 0.11-0.46) than in northern China 0.17 (IQR: 0.07-0.24). Similarly, interference intensity between B/Victoria and B/Yamagata was also higher in southern China 0.14 (IQR: 0.07-0.24) than in norther China 0.10 (IQR: 0.04-0.18). High relative humidity significantly increased subtype interference, with the highest relative risk reaching 20.59 (95% CI: 6.12-69.33) in southern China. Southern China exhibited higher levels of subtype interference, particularly between A/H1N1 and A/H3N2. Higher relative humidity has a more pronounced promoting effect on subtype interference.
Collapse
Affiliation(s)
- Can Chen
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mengya Yang
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yu Wang
- College of Computer Science and Technology, Zhejiang University, Hangzhou 310058, China
| | - Daixi Jiang
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yuxia Du
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Kexin Cao
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaobao Zhang
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoyue Wu
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mengsha Chen
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yue You
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wenkai Zhou
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jiaxing Qi
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Rui Yan
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Changtai Zhu
- Department of Transfusion Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Shigui Yang
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
56
|
Chen L, Hua J, He X. Bioinformatics analysis identifies a key gene HLA_DPA1 in severe influenza-associated immune infiltration. BMC Genomics 2024; 25:257. [PMID: 38454348 PMCID: PMC10918912 DOI: 10.1186/s12864-024-10184-7] [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: 07/07/2023] [Accepted: 03/04/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Severe influenza is a serious global health issue that leads to prolonged hospitalization and mortality on a significant scale. The pathogenesis of this infectious disease is poorly understood. Therefore, this study aimed to identify the key genes associated with severe influenza patients necessitating invasive mechanical ventilation. METHODS The current study utilized two publicly accessible gene expression profiles (GSE111368 and GSE21802) from the Gene Expression Omnibus database. The research focused on identifying the genes exhibiting differential expression between severe and non-severe influenza patients. We employed three machine learning algorithms, namely the Least Absolute Shrinkage and Selection Operator regression model, Random Forest, and Support Vector Machine-Recursive Feature Elimination, to detect potential key genes. The key gene was further selected based on the diagnostic performance of the target genes substantiated in the dataset GSE101702. A single-sample gene set enrichment analysis algorithm was applied to evaluate the participation of immune cell infiltration and their associations with key genes. RESULTS A total of 44 differentially expressed genes were recognized; among them, we focused on 10 common genes, namely PCOLCE2, HLA_DPA1, LOC653061, TDRD9, MPO, HLA_DQA1, MAOA, S100P, RAP1GAP, and CA1. To ensure the robustness of our findings, we employed overlapping LASSO regression, Random Forest, and SVM-RFE algorithms. By utilizing these algorithms, we were able to pinpoint the aforementioned 10 genes as potential biomarkers for distinguishing between both cases of influenza (severe and non-severe). However, the gene HLA_DPA1 has been recognized as a crucial factor in the pathological condition of severe influenza. Notably, the validation dataset revealed that this gene exhibited the highest area under the receiver operating characteristic curve, with a value of 0.891. The use of single-sample gene set enrichment analysis has provided valuable insights into the immune responses of patients afflicted with severe influenza that have further revealed a categorical correlation between the expression of HLA_DPA1 and lymphocytes. CONCLUSION The findings indicated that the HLA_DPA1 gene may play a crucial role in the immune-pathological condition of severe influenza and could serve as a promising therapeutic target for patients infected with severe influenza.
Collapse
Affiliation(s)
- Liang Chen
- Department of Infectious Diseases, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical College of Nanjing University, No 188, Lingshan North Road, Qixia District, Nanjing, 210046, China.
| | - Jie Hua
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaopu He
- Department of Geriatric Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
57
|
Hasan J, Bok S. Plasmonic Fluorescence Sensors in Diagnosis of Infectious Diseases. BIOSENSORS 2024; 14:130. [PMID: 38534237 DOI: 10.3390/bios14030130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024]
Abstract
The increasing demand for rapid, cost-effective, and reliable diagnostic tools in personalized and point-of-care medicine is driving scientists to enhance existing technology platforms and develop new methods for detecting and measuring clinically significant biomarkers. Humanity is confronted with growing risks from emerging and recurring infectious diseases, including the influenza virus, dengue virus (DENV), human immunodeficiency virus (HIV), Ebola virus, tuberculosis, cholera, and, most notably, SARS coronavirus-2 (SARS-CoV-2; COVID-19), among others. Timely diagnosis of infections and effective disease control have always been of paramount importance. Plasmonic-based biosensing holds the potential to address the threat posed by infectious diseases by enabling prompt disease monitoring. In recent years, numerous plasmonic platforms have risen to the challenge of offering on-site strategies to complement traditional diagnostic methods like polymerase chain reaction (PCR) and enzyme-linked immunosorbent assays (ELISA). Disease detection can be accomplished through the utilization of diverse plasmonic phenomena, such as propagating surface plasmon resonance (SPR), localized SPR (LSPR), surface-enhanced Raman scattering (SERS), surface-enhanced fluorescence (SEF), surface-enhanced infrared absorption spectroscopy, and plasmonic fluorescence sensors. This review focuses on diagnostic methods employing plasmonic fluorescence sensors, highlighting their pivotal role in swift disease detection with remarkable sensitivity. It underscores the necessity for continued research to expand the scope and capabilities of plasmonic fluorescence sensors in the field of diagnostics.
Collapse
Affiliation(s)
- Juiena Hasan
- Department of Electrical and Computer Engineering, Ritchie School of Engineering and Computer Science, University of Denver, Denver, CO 80208, USA
| | - Sangho Bok
- Department of Electrical and Computer Engineering, Ritchie School of Engineering and Computer Science, University of Denver, Denver, CO 80208, USA
| |
Collapse
|
58
|
Chakraborty C, Bhattacharya M, Lee SS. Regulatory role of miRNAs in the human immune and inflammatory response during the infection of SARS-CoV-2 and other respiratory viruses: A comprehensive review. Rev Med Virol 2024; 34:e2526. [PMID: 38446531 DOI: 10.1002/rmv.2526] [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: 01/16/2024] [Revised: 02/11/2024] [Accepted: 02/22/2024] [Indexed: 03/07/2024]
Abstract
miRNAs are single-stranded ncRNAs that act as regulators of different human body processes. Several miRNAs have been noted to control the human immune and inflammatory response during severe acute respiratory infection syndrome (SARS-CoV-2) infection. Similarly, many miRNAs were upregulated and downregulated during different respiratory virus infections. Here, an attempt has been made to capture the regulatory role of miRNAs in the human immune and inflammatory response during the infection of SARS-CoV-2 and other respiratory viruses. Firstly, the role of miRNAs has been depicted in the human immune and inflammatory response during the infection of SARS-CoV-2. In this direction, several significant points have been discussed about SARS-CoV-2 infection, such as the role of miRNAs in human innate immune response; miRNAs and its regulation of granulocytes; the role of miRNAs in macrophage activation and polarisation; miRNAs and neutrophil extracellular trap formation; miRNA-related inflammatory response; and miRNAs association in adaptive immunity. Secondly, the miRNAs landscape has been depicted during human respiratory virus infections such as human coronavirus, respiratory syncytial virus, influenza virus, rhinovirus, and human metapneumovirus. The article will provide more understanding of the miRNA-controlled mechanism of the immune and inflammatory response during COVID-19, which will help more therapeutics discoveries to fight against the future pandemic.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, India
| | | | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopaedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Gangwon-do, Republic of Korea
| |
Collapse
|
59
|
Malik S, Asghar M, Waheed Y. Outlining recent updates on influenza therapeutics and vaccines: A comprehensive review. Vaccine X 2024; 17:100452. [PMID: 38328274 PMCID: PMC10848012 DOI: 10.1016/j.jvacx.2024.100452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/27/2023] [Accepted: 01/29/2024] [Indexed: 02/09/2024] Open
Abstract
Influenza virus has presented a considerable healthcare challenge during the past years, particularly in vulnerable groups with compromised immune systems. Therapeutics and vaccination have always been in research annals since the spread of influenza. Efforts have been going on to develop an antiviral therapeutic approach that could assist in better disease management and reduce the overall disease complexity, resistance development, and fatality rates. On the other hand, vaccination presents a chance for effective, long-term, cost-benefit, and preventive response against the morbidity and mortality associated with the influenza. However, the issues of resistance development, strain mutation, antigenic variability, and inability to cure wide-spectrum and large-scale strains of the virus by available vaccines remain there. The article gathers the updated data for the therapeutics and available influenza vaccines, their mechanism of action, shortcomings, and trials under clinical experimentation. A methodological approach has been adopted to identify the prospective therapeutics and available vaccines approved and within the clinical trials against the influenza virus. Review contains influenza therapeutics, including traditional and novel antiviral drugs and inhibitor therapies against influenza virus as well as research trials based on newer drug combinations and latest technologies such as nanotechnology and organic and plant-based natural products. Most recent development of influenza vaccine has been discussed including some updates on traditional vaccination protocols and discussion on next-generation and upgraded novel technologies. This review will help the readers to understand the righteous approach for dealing with influenza virus infection and for deducing futuristic approaches for novel therapeutic and vaccine trials against Influenza.
Collapse
Affiliation(s)
- Shiza Malik
- Bridging Health Foundation, Rawalpindi, Punjab 46000, Pakistan
| | - Muhammad Asghar
- Department of Biology, Lund University, Sweden
- Department of Healthcare Biotechnology, Atta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Yasir Waheed
- Office of Research, Innovation, and Commercialization (ORIC), Shaheed Zulfiqar Ali Bhutto Medical University (SZABMU), Islamabad 44000, Pakistan
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos 1401, Lebanon
| |
Collapse
|
60
|
Yeon Kim S, Lee TY, Hwang YH, Kim D, Kim YJ, Won H. Booster doses of an inactivated F genotype mumps vaccine enhance immunogenicity in mice. Vaccine X 2024; 17:100437. [PMID: 38317857 PMCID: PMC10839135 DOI: 10.1016/j.jvacx.2024.100437] [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: 11/07/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 02/07/2024] Open
Abstract
The mumps virus (MuV) causes a highly contagious human disease characterized by swelling of the parotid glands. Although the administration of an attenuated Jeryl Lynn (JL) MuV vaccine shows efficacy in reducing the incidence of MuV infection, sporadic mumps outbreaks still occur in vaccinated populations. We have previously established that an inactivated F genotype mumps vaccine has a higher neutralizing antibody titer against diverse circulating mumps viruses in mice. Here, we aimed to develop a vaccination strategy to enhance the immune response for MuV and assess the effects of heterologous vaccination compared with homologous approaches. We administered an inactivated F genotype mumps vaccine booster following a homologous prime-boost regime and compared its efficacy with three doses of homologous JL vaccine in mice. We demonstrated robust stimulation of neutralizing antibodies and cellular immune response of interferon-γ-secreting cytotoxic T cells following administration of an inactivated F genotype mumps vaccine booster after a homologous prime-boost regime with JL. Compared with the homologous prime-boost regime, this heterologous prime-boost regime showed protective efficacy against the F genotype of MuV. These findings suggest that the heterologous vaccination strategy based on the administration of an inactivated F genotype mumps vaccine provides more effective cross-protection against circulating wild-type mumps viruses than homologous vaccination.
Collapse
Affiliation(s)
- Seo Yeon Kim
- Division of Infectious Diseases Vaccine Research, National Institute of Health, Korea Centers for Disease Control and Prevention Agency, CheongJu, Chungcheongbuk-do, Republic of Korea
| | - Tae-Young Lee
- Division of Infectious Diseases Vaccine Research, National Institute of Health, Korea Centers for Disease Control and Prevention Agency, CheongJu, Chungcheongbuk-do, Republic of Korea
| | - Yun-Ho Hwang
- Division of Infectious Diseases Vaccine Research, National Institute of Health, Korea Centers for Disease Control and Prevention Agency, CheongJu, Chungcheongbuk-do, Republic of Korea
| | - Dokeun Kim
- Division of Infectious Diseases Vaccine Research, National Institute of Health, Korea Centers for Disease Control and Prevention Agency, CheongJu, Chungcheongbuk-do, Republic of Korea
| | - You-Jin Kim
- Division of Infectious Diseases Vaccine Research, National Institute of Health, Korea Centers for Disease Control and Prevention Agency, CheongJu, Chungcheongbuk-do, Republic of Korea
| | - Hyeran Won
- Division of Infectious Diseases Vaccine Research, National Institute of Health, Korea Centers for Disease Control and Prevention Agency, CheongJu, Chungcheongbuk-do, Republic of Korea
| |
Collapse
|
61
|
Liu W, Zhang X, Wang D, Yu X, Guo S, Teng F. Reduced IgG2 with thrombocytopenia predicts mortality in patients with influenza pneumonia. Heart Lung 2024; 64:24-30. [PMID: 37984100 DOI: 10.1016/j.hrtlng.2023.11.005] [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: 04/07/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Thrombocytopenia is a common disorder during influenza that is related to high mortality. OBJECTIVES A prospective study was performed to investigate the association of immunoglobulin subclass changes accompanying incident thrombocytopenia with clinical outcomes in patients with severe influenza. METHODS 96 influenza patients were recruited and divided into two groups, patients with thrombocytopenia (n = 30) and patients without thrombocytopenia (n = 66). Plasma microarrays were used for quantitative analysis of immunoglobulins. The endpoint was 28-day mortality. Continuous platelet count, d-dimer, level of each Ig subclass and other variables were compared between the two groups. Kaplan-Meier curve was taken to analyze the 28-day survival rate of the two groups and Cox regression analysis was performed to identify variables independently associated with 28-day mortality. RESULTS Patients with thrombocytopenia had significantly high values of d-dimer at admission and when platelet lowest with high SOFA score. Their IgA2, IgG2, and IgG4 values were also lower than those without thrombocytopenia. Patients without thrombocytopenia had a higher 28-day survival rate than those in the thrombocytopenia group. In the multivariate Cox regression model, age (HR = 1.036, 95%CI = 1.011-1.062), IgG2 (HR = 0.990, 95%CI = 0.982-0.998), platelet minimum within 28 days (HR = 0.991, 95%CI = 0.982-0.999) and d-dimer when platelet lowest (HR = 1.091, 95%CI = 1.047-1.137) were independently related to 28-day mortality. CONCLUSION Decreased IgG2 may be associated with thrombocytopenia. A coexistence of thrombocytopenia, IgG2 reduction and d-dimer elevation may improve the accuracy of mortality prediction in patients with influenza pneumonia.
Collapse
Affiliation(s)
- Wenxin Liu
- Emergency Medicine Center, & Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing 100020, China
| | - Xiaomei Zhang
- Department of State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Dan Wang
- Department of State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiaobo Yu
- Department of State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Shubin Guo
- Emergency Medicine Center, & Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing 100020, China.
| | - Fei Teng
- Emergency Medicine Center, & Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing 100020, China.
| |
Collapse
|
62
|
Zahedipour F, Zahedipour F, Zamani P, Jaafari MR, Sahebkar A. Harnessing CRISPR technology for viral therapeutics and vaccines: from preclinical studies to clinical applications. Virus Res 2024; 341:199314. [PMID: 38211734 PMCID: PMC10825633 DOI: 10.1016/j.virusres.2024.199314] [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: 12/09/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
The CRISPR/Cas system, identified as a type of bacterial adaptive immune system, have attracted significant attention due to its remarkable ability to precisely detect and eliminate foreign genetic material and nucleic acids. Expanding upon these inherent capabilities, recent investigations have unveiled the potential of reprogrammed CRISPR/Cas 9, 12, and 13 systems for treating viral infections associated with human diseases, specifically targeting DNA and RNA viruses, respectively. Of particular interest is the RNA virus responsible for the recent global outbreak of coronavirus disease 2019 (COVID-19), which presents a substantial public health risk, coupled with limited efficacy of current prophylactic and therapeutic techniques. In this regard, the utilization of CRISPR/Cas technology offers a promising gene editing approach to overcome the limitations of conventional methods in managing viral infections. This comprehensive review provides an overview of the latest CRISPR/Cas-based therapeutic and vaccine strategies employed to combat human viral infections. Additionally, we discuss significant challenges and offer insights into the future prospects of this cutting-edge gene editing technology.
Collapse
Affiliation(s)
- Farzaneh Zahedipour
- Microbiology Department, Medical Sciences Branch, Islamic Azad University (IAU), Tehran, Iran
| | - Fatemeh Zahedipour
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
63
|
Chen D, Zhang T, Chen S, Ru X, Shao Q, Ye Q, Cheng D. The effect of nonpharmaceutical interventions on influenza virus transmission. Front Public Health 2024; 12:1336077. [PMID: 38389947 PMCID: PMC10881707 DOI: 10.3389/fpubh.2024.1336077] [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: 11/10/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Background The use of nonpharmaceutical interventions (NPIs) during severe acute respiratory syndrome 2019 (COVID-19) outbreaks may influence the spread of influenza viruses. This study aimed to evaluate the impact of NPIs against SARS-CoV-2 on the epidemiological features of the influenza season in China. Methods We conducted a retrospective observational study analyzing influenza monitoring data obtained from the China National Influenza Center between 2011 and 2023. We compared the changes in influenza-positive patients in the pre-COVID-19 epidemic, during the COVID-19 epidemic, and post-COVID-19 epidemic phases to evaluate the effect of NPIs on influenza virus transmission. Results NPIs targeting COVID-19 significantly suppressed influenza activity in China from 2019 to 2022. In the seventh week after the implementation of the NPIs, the number of influenza-positive patients decreased by 97.46% in southern regions of China and 90.31% in northern regions of China. However, the lifting of these policies in December 2022 led to an unprecedented surge in influenza-positive cases in autumn and winter from 2022 to 2023. The percentage of positive influenza cases increased by 206.41% (p < 0.001), with high positivity rates reported in both the northern and southern regions of China. Conclusion Our findings suggest that NPIs against SARS-CoV-2 are effective at controlling influenza epidemics but may compromise individuals' immunity to the virus.
Collapse
Affiliation(s)
- Danlei Chen
- School of Medical Technology and Informatlon Engineering, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Laboratory Medicine, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Ting Zhang
- School of Medical Technology and Informatlon Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Simiao Chen
- Department of Laboratory Medicine, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Xuanwen Ru
- Department of Laboratory Medicine, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Qingyi Shao
- School of Medical Technology and Informatlon Engineering, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Laboratory Medicine, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Qing Ye
- Department of Laboratory Medicine, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Dongqing Cheng
- School of Medical Technology and Informatlon Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
64
|
He Y, Zhou J, Gao H, Liu C, Zhan P, Liu X. Broad-spectrum antiviral strategy: Host-targeting antivirals against emerging and re-emerging viruses. Eur J Med Chem 2024; 265:116069. [PMID: 38160620 DOI: 10.1016/j.ejmech.2023.116069] [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: 10/03/2023] [Revised: 12/06/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024]
Abstract
Viral infections are amongst the most prevalent diseases that pose a significant threat to human health. Targeting viral proteins or host factors represents two primary strategies for the development of antiviral drugs. In contrast to virus-targeting antivirals (VTAs), host-targeting antivirals (HTAs) offer advantages in terms of overcoming drug resistance and effectively combating a wide range of viruses, including newly emerging ones. Therefore, targeting host factors emerges as an extremely promising strategy with the potential to address critical challenges faced by VTAs. In recent years, extensive research has been conducted on the discovery and development of HTAs, leading to the approval of maraviroc, a chemokine receptor type 5 (CCR5) antagonist used for the treatment of HIV-1 infected individuals, with several other potential treatments in various stages of development for different viral infections. This review systematically summarizes advancements made in medicinal chemistry regarding various host targets and classifies them into four distinct catagories based on their involvement in the viral life cycle: virus attachment and entry, biosynthesis, nuclear import and export, and viral release.
Collapse
Affiliation(s)
- Yong He
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China
| | - Jiahui Zhou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China
| | - Huizhan Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China
| | - Chuanfeng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China.
| |
Collapse
|
65
|
Wu YH, Chang WT, Hsu CL, Lu YF, Wang JT, Tzeng SJ. FcγRIIB modulates splenic germinal center response against immune subversion during acute influenza A virus infection. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2024; 57:64-75. [PMID: 38087748 DOI: 10.1016/j.jmii.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/02/2023] [Accepted: 11/28/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND B cells are essential for providing humoral protection against acute influenza A virus (IAV) infection. FcγRIIB, a regulator of antibody (Ab) production, influences immune responses during pathogen infections, but its specific impact on humoral protection and B cell-mediated responses against IAV remains unclear. METHODS To investigate FcγRIIB's role in host defense and B cell function during acute IAV infection, we generated mice with systemic FcγRIIB deficiency, functional impairment, and B cell-specific FcγRIIB deletion. We infected these mice with PR8 (H1N1) or Hkx31 (H3N2) IAVs and evaluated body weight preservation, survival rates, Ab production, viral neutralization, Ab affinity maturation, and germinal center B cell development. RESULTS Mice lacking FcγRIIB or with impaired function showed improved protection, preserved body weight, and increased survival rates during IAV infection. Notably, mice with haploinsufficient FcγRIIB function displayed protective effects. Selective deficiency of FcγRIIB in B cells led to enhanced Ab production, resulting in elevated IAV-specific Abs in the serum with superior viral neutralizing potency. However, the impact on the affinity maturation index of virus-specific Abs was modest. Accordingly, FcγRIIB-deficient B cells maintained normal germinal center B cell development during IAV infection, whereas wild-type mice exhibited delayed differentiation. CONCLUSION Our research underscores the pivotal role of FcγRIIB in host defense and B cell-mediated immunity during acute IAV infection. Additionally, our discoveries hold implications for antiviral treatments, particularly during the initial stages of IAV infection, aimed at enhancing the host's humoral immune response.
Collapse
Affiliation(s)
- Yu-Hsuan Wu
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taiwan.
| | - Wan-Ting Chang
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taiwan.
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taiwan.
| | - Yan-Fong Lu
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taiwan; Department of Obstetrics and Gynecology, Shin-Kong Wu Ho-Su Memorial Hospital, Taiwan.
| | - Jann-Tay Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taiwan.
| | - Shiang-Jong Tzeng
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taiwan.
| |
Collapse
|
66
|
Zhao Z, Han S, Zhang Q, Wang Y, Yue K, Abbas S, He H. Impaired influenza A virus replication by the host restriction factor SAMHD1 which inhibited by PA-mediated dephosphorylation of the host transcription factor IRF3. Virol J 2024; 21:33. [PMID: 38287375 PMCID: PMC10826253 DOI: 10.1186/s12985-024-02295-0] [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: 09/28/2023] [Accepted: 01/11/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Influenza A virus (IAV) can cause severe and life-threatening illness in humans and animals. Therefore, it is important to search for host antiviral proteins and elucidate their antiviral mechanisms for the development of potential treatments. As a part of human innate immunity, host restriction factors can inhibit the replication of viruses, among which SAM and HD domain containing deoxynucleoside triphosphate triphosphohydrolase 1 (SAMHD1) can restrict the replication of viruses, such as HIV and enterovirus EV71. Viruses also developed countermeasures in the arms race with their hosts. There are few reports about whether SAMHD1 has a restriction effect on IAV. METHODS To investigate the impact of IAV infection on SAMHD1 expression in A549 cells, we infected A549 cells with a varying multiplicity of infection (MOI) of IAV and collected cell samples at different time points for WB and RT-qPCR analysis to detect viral protein and SAMHD1 levels. The virus replication level in the cell culture supernatant was determined using TCID50 assay. Luciferase assay was used to reveal that H5N1 virus polymerase acidic protein (PA) affected the activity of the SAMHD1 promoter. To assess the antiviral capacity of SAMHD1, we generated a knockdown and overexpressed cell line for detecting H5N1 replication. RESULTS In this study, we observed that SAMHD1 can restrict the intracellular replication of H5N1 and that the H5N1 viral protein PA can downregulate the expression of SAMHD1 by affecting SAMHD1 transcriptional promoter activity. We also found that SAMHD1's ability to restrict H5N1 is related to phosphorylation at 592-tyrosine. CONCLUSIONS In conclusion, we found that SAMHD1 may affect the replication of IAVs as a host restriction factor and be countered by PA. Furthermore, SAMHD1 may be a potential target for developing antiviral drugs.
Collapse
Affiliation(s)
- Zhilei Zhao
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuyi Han
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Qingxun Zhang
- Beijing Milu Ecological Research Center, Beijing, 100076, China
| | - Ye Wang
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Kening Yue
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Salbia Abbas
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Hongxuan He
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
67
|
Rolta R, Salaria D, Fadare OA, Fadare RY, Masih GD, Prakash A, Medhi B. Identification of novel inhibitor phytoconstituents for Influenza A H3N2: an in silico approach. J Biomol Struct Dyn 2024:1-10. [PMID: 38247233 DOI: 10.1080/07391102.2024.2305313] [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: 08/23/2023] [Accepted: 12/26/2023] [Indexed: 01/23/2024]
Abstract
Influenza A virus subtype H3N2 is a highly infectious respiratory virus that is responsible for global seasonal flu epidemics. The current study was designed to investigate the antiviral activity of 150 phytocompounds of North Western Himalayas medicinal plants by molecular docking. Two target proteins of hemagglutinin of influenza virus A (PDB ID 4WE8) and Influenza virus H3N2 nucleoprotein - R416A mutant (PDB ID 7NT8) are selected for this study. Molecular docking was done by AutoDock vina tool, toxicity and drug-likeness prediction was done by protox II and Moleinspiration. MD simulation of best protein-ligand complexes was done by using Gromacs, version 2021.5. Molecular docking and toxicity data revealed that clicoemodin and rumexocide showed the best binding with both target proteins 4WEB & 7NT8. Clicoemodin showed the -7.5 KJ/mol binding energy with 4WE8 and 7NT8. Similarly, rumexoside showed the -7.6 KJ/mol binding energy with 4WE8 and -7.6 KJ/mol with 7NT8. Furthermore, Molecular dynamic simulation and MMPBSA binding free energy validated the stability of protein-ligand complexes. The current study suggested that clicoemodin and rumexocide are the promising inhibitors of H3N2 proteins hemagglutinin of influenza virus A and Influenza virus H3N2 nucleoprotein - R416A mutant, though there is further in vitro and in vivo validation is required.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rajan Rolta
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Deeksha Salaria
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Olatomide A Fadare
- Organic Chemistry Research Lab, Obafemi Awolowo University, Ile-Ife Osun, Nigeria
| | - Racheal Y Fadare
- Department of Physical and Chemical Sciences, Elizade University, Ilara-Mokin, Nigeria
| | - Gladson David Masih
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ajay Prakash
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
68
|
de Jong SPJ, Felix Garza ZC, Gibson JC, van Leeuwen S, de Vries RP, Boons GJ, van Hoesel M, de Haan K, van Groeningen LE, Hulme KD, van Willigen HDG, Wynberg E, de Bree GJ, Matser A, Bakker M, van der Hoek L, Prins M, Kootstra NA, Eggink D, Nichols BE, Han AX, de Jong MD, Russell CA. Determinants of epidemic size and the impacts of lulls in seasonal influenza virus circulation. Nat Commun 2024; 15:591. [PMID: 38238318 PMCID: PMC10796432 DOI: 10.1038/s41467-023-44668-z] [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: 02/22/2022] [Accepted: 12/21/2023] [Indexed: 01/22/2024] Open
Abstract
During the COVID-19 pandemic, levels of seasonal influenza virus circulation were unprecedentedly low, leading to concerns that a lack of exposure to influenza viruses, combined with waning antibody titres, could result in larger and/or more severe post-pandemic seasonal influenza epidemics. However, in most countries the first post-pandemic influenza season was not unusually large and/or severe. Here, based on an analysis of historical influenza virus epidemic patterns from 2002 to 2019, we show that historic lulls in influenza virus circulation had relatively minor impacts on subsequent epidemic size and that epidemic size was more substantially impacted by season-specific effects unrelated to the magnitude of circulation in prior seasons. From measurements of antibody levels from serum samples collected each year from 2017 to 2021, we show that the rate of waning of antibody titres against influenza virus during the pandemic was smaller than assumed in predictive models. Taken together, these results partially explain why the re-emergence of seasonal influenza virus epidemics was less dramatic than anticipated and suggest that influenza virus epidemic dynamics are not currently amenable to multi-season prediction.
Collapse
Affiliation(s)
- Simon P J de Jong
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Zandra C Felix Garza
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Joseph C Gibson
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Sarah van Leeuwen
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Robert P de Vries
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Geert-Jan Boons
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
- Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Marliek van Hoesel
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Karen de Haan
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Laura E van Groeningen
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Katina D Hulme
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Hugo D G van Willigen
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Elke Wynberg
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, The Netherlands
| | - Godelieve J de Bree
- Department of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Amy Matser
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, The Netherlands
| | - Margreet Bakker
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Lia van der Hoek
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Maria Prins
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, The Netherlands
- Department of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Neeltje A Kootstra
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Dirk Eggink
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Brooke E Nichols
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Global Health, School of Public Health, Boston University, Boston, MA, USA
| | - Alvin X Han
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Menno D de Jong
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Colin A Russell
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.
- Department of Global Health, School of Public Health, Boston University, Boston, MA, USA.
| |
Collapse
|
69
|
Uno N, Ross TM. Multivalent next generation influenza virus vaccines protect against seasonal and pre-pandemic viruses. Sci Rep 2024; 14:1440. [PMID: 38228649 PMCID: PMC10792005 DOI: 10.1038/s41598-023-51024-0] [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: 03/03/2023] [Accepted: 12/29/2023] [Indexed: 01/18/2024] Open
Abstract
Each year, new influenza virus vaccine formulations are generated to keep up with continuously circulating and mutating viral variants. A next-generation influenza virus vaccine would provide long-lasting, broadly-reactive immune protection against current and future influenza virus strains for both seasonal and pre-pandemic viruses. Next generation immunogens were designed using computationally optimized broadly reactive antigen (COBRA) methodology to protect against a broad range of strains over numerous seasons. Novel HA and NA amino acid sequences were derived from multilayered consensus sequence alignment for multiple subtypes of influenza. This multivalent formulation was hypothesized to elicit broadly protective immune responses against both seasonal and pre-pandemic influenza viruses. Mice were vaccinated with multivalent mixtures of HA and NA (H1, H2, H3, H5, H7, N1, N2) proteins. Multivalent COBRA vaccinations elicited antibodies that recognized a broad panel of strains and vaccinated mice were protected against viruses representing multiple subtypes. This is a promising candidate for a universal influenza vaccine that elicits protective immune responses against seasonal and pre-pandemic strains over multiple seasons.
Collapse
Affiliation(s)
- Naoko Uno
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA.
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA.
- Florida Research and Innovation Center, Cleveland Clinic Florida, 9801 SW Discovery Way, Port Saint Lucie, FL, 34986, USA.
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
70
|
Colombo É, Désilets A, Hassanzadeh M, Lemieux G, Marois I, Cliche D, Delbrouck JA, Murza A, Jean F, Marsault E, Richter MV, Leduc R, Boudreault PL. Optimization of Ketobenzothiazole-Based Type II Transmembrane Serine Protease Inhibitors to Block H1N1 Influenza Virus Replication. ChemMedChem 2024; 19:e202300458. [PMID: 37864572 DOI: 10.1002/cmdc.202300458] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023]
Abstract
Human influenza viruses cause acute respiratory symptoms that can lead to death. Due to the emergence of antiviral drug-resistant strains, there is an urgent requirement for novel antiviral agents and innovative therapeutic strategies. Using the peptidomimetic ketobenzothiazole protease inhibitor RQAR-Kbt (IN-1, aka N-0100) as a starting point, we report how substituting P2 and P4 positions with natural and unnatural amino acids can modulate the inhibition potency toward matriptase, a prototypical type II transmembrane serine protease (TTSP) that acts as a priming protease for influenza viruses. We also introduced modifications of the peptidomimetics N-terminal groups, leading to significant improvements (from μM to nM, 60 times more potent than IN-1) in their ability to inhibit the replication of influenza H1N1 virus in the Calu-3 cell line derived from human lungs. The selectivity towards other proteases has been evaluated and explained using molecular modeling with a crystal structure recently obtained by our group. By targeting host cell TTSPs as a therapeutic approach, it may be possible to overcome the high mutational rate of influenza viruses and consequently prevent potential drug resistance.
Collapse
Affiliation(s)
- Éloïc Colombo
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Antoine Désilets
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Malihe Hassanzadeh
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Gabriel Lemieux
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Isabelle Marois
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, J1H 5N4 Québec, Canada
- Current address: Department of Biology, Faculty of Sciences, Université de Sherbrooke, Sherbrooke, J1K 2R1 Québec, Canada
| | - Dominic Cliche
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, J1H 5N4 Québec, Canada
| | - Julien A Delbrouck
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
- Current address: Xenon Pharmaceuticals Inc., Burnaby, V5G 4W8, British Columbia, Canada
| | - Alexandre Murza
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - François Jean
- Department of Microbiology and Immunology, Faculty of Science, Life Sciences Institute, University of British Columbia, V6T 1Z3, British Columbia, Canada
| | - Eric Marsault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Martin V Richter
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, J1H 5N4 Québec, Canada
| | - Richard Leduc
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Pierre-Luc Boudreault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| |
Collapse
|
71
|
Zhang Y, Huang X, Zhang J, Tao Z. Risk factors for hospitalization and pneumonia development of pediatric patients with seasonal influenza during February-April 2023. Front Public Health 2024; 11:1300228. [PMID: 38249383 PMCID: PMC10797015 DOI: 10.3389/fpubh.2023.1300228] [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: 09/23/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024] Open
Abstract
Objectives In China influenza remains a low activity for continuous 3 years due to COVID-19 controls. We here sought to study the clinical characteristics and risk factors of the influenza infection among children after the mandatory COVID-19 restrictions were lifted. Methods We included 1,006 pediatric patients with influenza A virus (IAV) infection, enrolled in one tertiary hospital in Zhenjiang, Jiangsu Province, China, during February to April 2023. Patients were divided into the outpatient (n = 798) and inpatient (n = 208) groups, and their baseline characteristics were compared between two groups to conclude the risk factors for pediatric hospitalization. Separately, pediatric inpatients (n = 208) were further divided into the pneumonia and non-pneumonia groups with comparison of their clinical characteristics, including their laboratory test results and representative radiological features, to derive the key determinants for pneumonia development after hospitalization. Results Compared to outpatients, IAV-infected pediatric inpatients exhibited younger age, higher female: male ratio, more co-infection of influenza B virus (IBV) and hematological abnormality. Multivariate regression analysis determined the independent risk factors of hospitalization to be the clinical symptom of abdominal pain (OR = 2.63, [95% CI, 1.05-6.57], p = 0.039), co-infection of IBV (OR = 44.33, [95% CI, 25.10-78.30], p = 0.001), elevated levels of lymphocytes (OR = 2.24, [95% CI,1.65-3.05], p = 0.001) and c-reactive proteins (CRPs) (OR = 1.06, [95% CI, 1.03-1.08], p = 0.001) upon hospital admission. Furthermore, the cough symptom (OR = 17.39, [95% CI, 3.51-86.13], p = 0.001) and hospitalization length (OR = 1.36, [95% CI, 1.12-1.67], p = 0.002) were determined to be risk factors of pneumonia acquirement for pediatric inpatients. Conclusion While the abdominal pain, viral co-infection and some hematological abnormality mainly contribute to hospitalization of pediatric patients with IAV infection, the length of hospital stay and clinical sign of coughing upon hospital admission constitute the key determinants for nosocomial pneumonia development.
Collapse
Affiliation(s)
- Yuqian Zhang
- Department of Emergency Medicine, The Affiliated Hospital, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xing Huang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jianguo Zhang
- Department of Emergency Medicine, The Affiliated Hospital, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhimin Tao
- Department of Emergency Medicine, The Affiliated Hospital, Jiangsu University, Zhenjiang, Jiangsu, China
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
72
|
Wang X, Kim KW, Walker G, Stelzer‐Braid S, Scotch M, Rawlinson WD. Genome characterization of influenza A and B viruses in New South Wales, Australia, in 2019: A retrospective study using high-throughput whole genome sequencing. Influenza Other Respir Viruses 2024; 18:e13252. [PMID: 38288510 PMCID: PMC10824601 DOI: 10.1111/irv.13252] [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: 07/12/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND During the 2019 severe influenza season, New South Wales (NSW) experienced the highest number of cases in Australia. This study retrospectively investigated the genetic characteristics of influenza viruses circulating in NSW in 2019 and identified genetic markers related to antiviral resistance and potential virulence. METHODS The complete genomes of influenza A and B viruses were amplified using reverse transcription-polymerase chain reaction (PCR) and sequenced with an Illumina MiSeq platform. RESULTS When comparing the sequencing data with the vaccine strains and reference sequences, the phylogenetic analysis revealed that most NSW A/H3N2 viruses (n = 68; 94%) belonged to 3C.2a1b and a minority (n = 4; 6%) belonged to 3C.3a. These viruses all diverged from the vaccine strain A/Switzerland/8060/2017. All A/H1N1pdm09 viruses (n = 20) showed genetic dissimilarity from vaccine strain A/Michigan/45/2015, with subclades 6B.1A.5 and 6B.1A.2 identified. All B/Victoria-lineage viruses (n = 21) aligned with clade V1A.3, presenting triple amino acid deletions at positions 162-164 in the hemagglutinin protein, significantly diverging from the vaccine strain B/Colorado/06/2017. Multiple amino acid substitutions were also found in the internal proteins of influenza viruses, some of which have been previously reported in hospitalized influenza patients in Thailand. Notably, the oseltamivir-resistant marker H275Y was present in one immunocompromised patient infected with A/H1N1pdm09 and the resistance-related mutation I222V was detected in another A/H3N2-infected patient. CONCLUSIONS Considering antigenic drift and the constant evolution of circulating A and B strains, we believe continuous monitoring of influenza viruses in NSW via the high-throughput sequencing approach provides timely and pivotal information for both public health surveillance and clinical treatment.
Collapse
Affiliation(s)
- Xinye Wang
- School of Biomedical Sciences, Faculty of Medicine and HealthUniversity of New South WalesSydneyNew South WalesAustralia
- Virology Research Laboratory, Serology and Virology Division (SAViD), NSW Health PathologyPrince of Wales HospitalSydneyNew South WalesAustralia
| | - Ki Wook Kim
- Virology Research Laboratory, Serology and Virology Division (SAViD), NSW Health PathologyPrince of Wales HospitalSydneyNew South WalesAustralia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, Faculty of Medicine and HealthUniversity of New South WalesSydneyNew South WalesAustralia
| | - Gregory Walker
- School of Biomedical Sciences, Faculty of Medicine and HealthUniversity of New South WalesSydneyNew South WalesAustralia
- Virology Research Laboratory, Serology and Virology Division (SAViD), NSW Health PathologyPrince of Wales HospitalSydneyNew South WalesAustralia
| | - Sacha Stelzer‐Braid
- School of Biomedical Sciences, Faculty of Medicine and HealthUniversity of New South WalesSydneyNew South WalesAustralia
- Virology Research Laboratory, Serology and Virology Division (SAViD), NSW Health PathologyPrince of Wales HospitalSydneyNew South WalesAustralia
| | - Matthew Scotch
- Biodesign Center for Environmental Health Engineering, Biodesign InstituteArizona State UniversityPhoenixArizonaUSA
- College of Health SolutionsArizona State UniversityPhoenixArizonaUSA
- Kirby InstituteUniversity of New South WalesSydneyNew South WalesAustralia
| | - William D. Rawlinson
- School of Biomedical Sciences, Faculty of Medicine and HealthUniversity of New South WalesSydneyNew South WalesAustralia
- Virology Research Laboratory, Serology and Virology Division (SAViD), NSW Health PathologyPrince of Wales HospitalSydneyNew South WalesAustralia
| |
Collapse
|
73
|
Alvarez FP, Allard L, Bianic F, Bricout H, Crépey P, Gaillat J, Gavazzi G, Mosnier A, Launay O, Levant MC, Proshenska D, de Courville C. Cost-effectiveness and public health impact of using high dose quadrivalent influenza vaccine in the French older adults population. J Med Econ 2024; 27:1300-1307. [PMID: 39286871 DOI: 10.1080/13696998.2024.2404331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Seasonal influenza outbreaks in France cause a surge in patients, exacerbating the overburdened healthcare system each winter. Older adults are particularly vulnerable to serious events related to influenza. Quadrivalent influenza high dose (QIV HD) vaccines have been developed to offer better clinical protection in older adults, who often exhibit suboptimal immune response to quadrivalent influenza standard dose vaccines (QIV SD). This study aims to evaluate the public health impact and cost-effectiveness of administering HD versus SD vaccines to individuals aged 65+ in France. METHODOLOGY Using a static model and decision-tree approach, the study analyzed health outcomes such as influenza cases, GP (general practitioner) visits, hospitalizations, and mortality; relative vaccine efficacy (rVE) estimates were derived from a pivotal randomized-controlled trial and a meta-analysis comparing HD to SD vaccines. Two approaches were implemented to model hospitalizations (conditional on influenza or not), and analyses on bed occupancy were performed. RESULTS Results showed that using QIV HD instead of QIV SD during an average influenza season in France led to the prevention of 57,209 additional cases of influenza, 13,704 GP visits, and 764 influenza-related deaths. Moreover, switching to QIV HD resulted in an additional 1,728-15,970 hospitalizations avoided and 15,124-138,367 reduced days of hospitalization depending on the hospitalization approach used. The cost-utility analysis showed a cost per quality-adjusted life year (QALY) gained ranging from 24,020 €/QALY to 5,036 €/QALY. CONCLUSIONS Switching to QIV HD in older adults was shown to be cost-effective, with even greater public health benefits at a higher coverage rate, regardless of the season severity.
Collapse
Affiliation(s)
| | | | | | | | - P Crépey
- EHESP, Université de Rennes, REPERES, Rennes, France
| | - J Gaillat
- Centre Hospitalier Annecy-Genevois, Metz-Tessy, France
| | - G Gavazzi
- CHU Grenoble-Alpes, Grenoble, France
| | - A Mosnier
- Open Rome & Réseau des GROG, Paris, France
| | - O Launay
- Faculté de Médecine Paris Descartes, Université de Paris, Paris, France
| | | | | | | |
Collapse
|
74
|
Tamai M, Taba S, Mise T, Yamashita M, Ishikawa H, Shintake T. Effect of Ethanol Vapor Inhalation Treatment on Lethal Respiratory Viral Infection With Influenza A. J Infect Dis 2023; 228:1720-1729. [PMID: 37101418 PMCID: PMC10733743 DOI: 10.1093/infdis/jiad089] [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: 12/02/2022] [Revised: 03/12/2023] [Accepted: 04/03/2023] [Indexed: 04/28/2023] Open
Abstract
Ethanol (EtOH) effectively inactivates enveloped viruses in vitro, including influenza and severe acute respiratory syndrome coronavirus 2. Inhaled EtOH vapor may inhibit viral infection in mammalian respiratory tracts, but this has not yet been demonstrated. Here we report that unexpectedly low EtOH concentrations in solution, approximately 20% (vol/vol), rapidly inactivate influenza A virus (IAV) at mammalian body temperature and are not toxic to lung epithelial cells on apical exposure. Furthermore, brief exposure to 20% (vol/vol) EtOH decreases progeny virus production in IAV-infected cells. Using an EtOH vapor exposure system that is expected to expose murine respiratory tracts to 20% (vol/vol) EtOH solution by gas-liquid equilibrium, we demonstrate that brief EtOH vapor inhalation twice a day protects mice from lethal IAV respiratory infection by reducing viruses in the lungs without harmful side effects. Our data suggest that EtOH vapor inhalation may provide a versatile therapy against various respiratory viral infectious diseases.
Collapse
Affiliation(s)
- Miho Tamai
- Immune Signal Unit, Okinawa Institute of Science and Technology, Tancha 1919-1, Onna-son, Okinawa 904-0495, Japan
| | - Seita Taba
- Quantum Wave Microscopy Unit, Okinawa Institute of Science and Technology, Tancha 1919-1, Onna-son, Okinawa 904-0495, Japan
| | - Takeshi Mise
- Quantum Wave Microscopy Unit, Okinawa Institute of Science and Technology, Tancha 1919-1, Onna-son, Okinawa 904-0495, Japan
| | - Masao Yamashita
- Quantum Wave Microscopy Unit, Okinawa Institute of Science and Technology, Tancha 1919-1, Onna-son, Okinawa 904-0495, Japan
| | - Hiroki Ishikawa
- Immune Signal Unit, Okinawa Institute of Science and Technology, Tancha 1919-1, Onna-son, Okinawa 904-0495, Japan
| | - Tsumoru Shintake
- Quantum Wave Microscopy Unit, Okinawa Institute of Science and Technology, Tancha 1919-1, Onna-son, Okinawa 904-0495, Japan
| |
Collapse
|
75
|
Hickerson BT, Huang BK, Petrovskaya SN, Ilyushina NA. Genomic Analysis of Influenza A and B Viruses Carrying Baloxavir Resistance-Associated Substitutions Serially Passaged in Human Epithelial Cells. Viruses 2023; 15:2446. [PMID: 38140689 PMCID: PMC10748225 DOI: 10.3390/v15122446] [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: 11/16/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Baloxavir marboxil (baloxavir) is an FDA-approved inhibitor of the influenza virus polymerase acidic (PA) protein. Here, we used next-generation sequencing to compare the genomic mutational profiles of IAV H1N1 and H3N2, and IBV wild type (WT) and mutants (MUT) viruses carrying baloxavir resistance-associated substitutions (H1N1-PA I38L, I38T, and E199D; H3N2-PA I38T; and IBV-PA I38T) during passaging in normal human bronchial epithelial (NHBE) cells. We determined the ratio of nonsynonymous to synonymous nucleotide mutations (dN/dS) and identified the location and type of amino acid (AA) substitutions that occurred at a frequency of ≥30%. We observed that IAV H1N1 WT and MUT viruses remained relatively stable during passaging. While the mutational profiles for IAV H1N1 I38L, I38T, and E199D, and IBV I38T MUTs were relatively similar after each passage compared to the respective WTs, the mutational profile of the IAV H3N2 I38T MUT was significantly different for most genes compared to H3N2 WT. Our work provides insight into how baloxavir resistance-associated substitutions may impact influenza virus evolution in natural settings. Further characterization of the potentially adaptive mutations identified in this study is needed.
Collapse
Affiliation(s)
- Brady T. Hickerson
- Division of Biotechnology Review and Research II, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Bruce K. Huang
- Division of Biotechnology Review and Research II, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Svetlana N. Petrovskaya
- Division of Biotechnology Review and Research III, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Natalia A. Ilyushina
- Division of Biotechnology Review and Research II, Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
76
|
Boussarsar M, Ennouri E, Habbachi N, Bouguezzi N, Meddeb K, Gallas S, Hafdhi M, Zghidi M, Toumi R, Ben Saida I, Abid S, Boutiba-Ben Boubaker I, Maazaoui L, El Ghord H, Gzara A, Yazidi R, Ben Salah A. Epidemiology and burden of Severe Acute Respiratory Infections (SARI) in the aftermath of COVID-19 pandemic: A prospective sentinel surveillance study in a Tunisian Medical ICU, 2022/2023. PLoS One 2023; 18:e0294960. [PMID: 38100529 PMCID: PMC10723666 DOI: 10.1371/journal.pone.0294960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/10/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Severe Acute Respiratory Infections (SARI) caused by influenza and other respiratory viruses pose significant global health challenges, and the COVID-19 pandemic has further strained healthcare systems. As the focus shifts from the pandemic to other respiratory infections, assessing the epidemiology and burden of SARI is crucial for healthcare planning and resource allocation. Aim: to understand the impact of the post-pandemic period on the epidemiology of SARI cases, clinical outcomes, and healthcare resource utilization in Tunisia. METHODS This is a prospective study conducted in a Tunisian MICU part of a national sentinel surveillance system, focusing on enhanced SARI surveillance. SARI cases from week 39/2022, 26 September to week 19/2023, 13 May were included, according to a standardized case definition. Samples were collected for virological RT-PCR testing, and an electronic system ensured standardized and accurate data collection. Descriptive statistics were performed to assess epidemiology, trends, and outcomes of SARI cases, and univariate/multivariate analyses to assess factors associated with mortality. RESULTS Among 312 MICU patients, 164 SARI cases were identified during the study period. 64(39%) RT-PCR were returned positive for at least one pathogen, with influenza A and B strains accounting for 20.7% of cases at the early stages of the influenza season. The MICU experienced a significant peak in admissions during weeks 1-11/2023, leading to resource mobilization and the creation of a surge unit. SARI cases utilized 1664/3120 of the MICU-stay days and required 1157 mechanical ventilation days. The overall mortality rate among SARI cases was 22.6%. Age, non-COPD, and ARDS were identified as independent predictors of mortality. CONCLUSIONS The present study identified a relatively high rate of SARI cases, with 39% positivity for at least one respiratory virus, with influenza A and B strains occurring predominantly during the early stages of the influenza season. The findings shed light on the considerable resource utilization and mortality associated with these infections, underscoring the urgency for proactive management and efficient resource allocation strategies.
Collapse
Affiliation(s)
- Mohamed Boussarsar
- University of Sousse, Faculty of Medicine of Sousse, Sousse, Tunisia
- Medical Intensive Care Unit, Research Laboratory “Heart Failure”, Farhat Hached University Hospital, Sousse, Tunisia
| | - Emna Ennouri
- University of Sousse, Faculty of Medicine of Sousse, Sousse, Tunisia
- Medical Intensive Care Unit, Research Laboratory “Heart Failure”, Farhat Hached University Hospital, Sousse, Tunisia
| | - Naima Habbachi
- University of Sousse, Faculty of Medicine of Sousse, Sousse, Tunisia
| | - Nabil Bouguezzi
- University of Sousse, Faculty of Medicine of Sousse, Sousse, Tunisia
- Medical Intensive Care Unit, Research Laboratory “Heart Failure”, Farhat Hached University Hospital, Sousse, Tunisia
| | - Khaoula Meddeb
- University of Sousse, Faculty of Medicine of Sousse, Sousse, Tunisia
- Medical Intensive Care Unit, Research Laboratory “Heart Failure”, Farhat Hached University Hospital, Sousse, Tunisia
| | - Salma Gallas
- University of Sousse, Faculty of Medicine of Sousse, Sousse, Tunisia
- Medical Intensive Care Unit, Research Laboratory “Heart Failure”, Farhat Hached University Hospital, Sousse, Tunisia
| | - Malek Hafdhi
- University of Sousse, Faculty of Medicine of Sousse, Sousse, Tunisia
| | - Marwa Zghidi
- University of Sousse, Faculty of Medicine of Sousse, Sousse, Tunisia
- Medical Intensive Care Unit, Research Laboratory “Heart Failure”, Farhat Hached University Hospital, Sousse, Tunisia
| | - Radhouane Toumi
- University of Sousse, Faculty of Medicine of Sousse, Sousse, Tunisia
- Medical Intensive Care Unit, Research Laboratory “Heart Failure”, Farhat Hached University Hospital, Sousse, Tunisia
| | - Imen Ben Saida
- University of Sousse, Faculty of Medicine of Sousse, Sousse, Tunisia
- Medical Intensive Care Unit, Research Laboratory “Heart Failure”, Farhat Hached University Hospital, Sousse, Tunisia
| | - Salma Abid
- National Influenza Centre-Tunis, Unit Virology, Microbiology Laboratory, Charles Nicolle Hospital, Tunis, Tunisia
- University of Tunis El Manar, Faculty of Medicine of Tunis, Tunis, Tunisia
| | - Ilhem Boutiba-Ben Boubaker
- National Influenza Centre-Tunis, Unit Virology, Microbiology Laboratory, Charles Nicolle Hospital, Tunis, Tunisia
- University of Tunis El Manar, Faculty of Medicine of Tunis, Tunis, Tunisia
| | | | | | - Ahlem Gzara
- Primary Health Care Directorate, Tunis, Tunisia
| | - Rihab Yazidi
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Service of Medical Epidemiology, Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infections LR16IPT02, Institut Pasteur de Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Afif Ben Salah
- Service of Medical Epidemiology, Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Department of Family and Community Medicine, College of Medicine and Medical Sciences (CMMS), Arabian Gulf University (AGU), Manama, Bahrain
| |
Collapse
|
77
|
Jiang M, Fang C, Ma Y. Deciphering the rule of antigen-antibody amino acid interaction. Front Immunol 2023; 14:1269916. [PMID: 38111576 PMCID: PMC10725943 DOI: 10.3389/fimmu.2023.1269916] [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: 08/01/2023] [Accepted: 11/13/2023] [Indexed: 12/20/2023] Open
Abstract
Purpose Antigenic drift is the biggest challenge for mutagenic RNA virus vaccine development. The primary purpose is to determine the IEMM (immune escape mutation map) of 20 amino acids' replacement to reveal the rule of the viral immune escape. Methods To determine the relationship between epitope mutation and immune escape, we use universal protein tags as a linear epitope model. To describe and draw amino acid linkage diagrams, mutations of protein tags are classified into four categories: IEM (immune escape mutation), ADERM (antibody-dependent enhancement risk mutation), EQM (equivalent mutation), and IVM (invalid mutation). To overcome the data limitation, a general antigen-antibody (Ag-Ab) interaction map was constructed by analyzing the published three-dimensional (3D) Ag-Ab interaction patterns. Results (i) One residue interacts with multiple amino acids in antigen-antibody interaction. (ii) Most amino acid replacements are IVM and EQM. (iii) Once aromatic amino acids replace non-aromatic amino acids, the mutation is often IEM. (iv) Substituting residues with the same physical and chemical properties easily leads to IVM. Therefore, this study has important theoretical significance for future research on antigenic drift, antibody rescue, and vaccine renewal design. Conclusion The antigenic epitope mutations were typed into IEM, ADERM, EQM, and IVM types to describe and quantify the results of antigenic mutations. The antigen-antibody interaction rule was summarized as a one-to-many interaction rule. To sum up, the epitope mutation rules were defined as IVM and EQM predomination rules and the aryl mutation escape rule.
Collapse
Affiliation(s)
| | | | - Yongping Ma
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Basical Medical Collage, Chongqing Medical University, Chongqing, China
| |
Collapse
|
78
|
Mai G, Zhang C, Lan C, Zhang J, Wang Y, Tang K, Tang J, Zeng J, Chen Y, Cheng P, Liu S, Long H, Wen Q, Li A, Liu X, Zhang R, Xu S, Liu L, Niu Y, Yang L, Wang Y, Yin D, Sun C, Chen YQ, Shen W, Zhang Z, Du X. Characterizing the dynamics of BCR repertoire from repeated influenza vaccination. Emerg Microbes Infect 2023; 12:2245931. [PMID: 37542407 PMCID: PMC10438862 DOI: 10.1080/22221751.2023.2245931] [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: 05/21/2023] [Revised: 07/12/2023] [Accepted: 08/03/2023] [Indexed: 08/06/2023]
Abstract
Yearly epidemics of seasonal influenza cause an enormous disease burden around the globe. An understanding of the rules behind the immune response with repeated vaccination still presents a significant challenge, which would be helpful for optimizing the vaccination strategy. In this study, 34 healthy volunteers with 16 vaccinated were recruited, and the dynamics of the BCR repertoire for consecutive vaccinations in two seasons were tracked. In terms of diversity, length, network, V and J gene segments usage, somatic hypermutation (SHM) rate and isotype, it was found that the overall changes were stronger in the acute phase of the first vaccination than the second vaccination. However, the V gene segments of IGHV4-39, IGHV3-9, IGHV3-7 and IGHV1-69 were amplified in the acute phase of the first vaccination, with IGHV3-7 dominant. On the other hand, for the second vaccination, the changes were dominated by IGHV1-69, with potential for coding broad neutralizing antibody. Additional analysis indicates that the application of V gene segment for IGHV3-7 in the acute phase of the first vaccination was due to the elevated usage of isotypes IgM and IgG3. While for IGHV1-69 in the second vaccination, it was contributed by isotypes IgG1 and IgG2. Finally, 41 public BCR clusters were identified in the vaccine group, with both IGHV3-7 and IGHV1-69 were involved and representative complementarity determining region 3 (CDR3) motifs were characterized. This study provides insights into the immune response dynamics following repeated influenza vaccination in humans and can inform universal vaccine design and vaccine strategies in the future.
Collapse
Affiliation(s)
- Guoqin Mai
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Chi Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Chunhong Lan
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
- Center for Precision Medicine, Guangdong Academy of Medical Sciences, Medical Research Institute, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| | - Jie Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yuanyuan Wang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Kang Tang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Jing Tang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Jinfeng Zeng
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yilin Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Peiwen Cheng
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Shuning Liu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Haoyu Long
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Qilan Wen
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Aqin Li
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Xuan Liu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Ruitong Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Shuyang Xu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Lin Liu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yanlan Niu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Lan Yang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yihan Wang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Di Yin
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Caijun Sun
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yao-Qing Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Wei Shen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Zhenhai Zhang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
- Center for Precision Medicine, Guangdong Academy of Medical Sciences, Medical Research Institute, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, People’s Republic of China
| | - Xiangjun Du
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
79
|
Xia Y, Liu K, Wang F, Xu Z, Wang Y, Zong R, Xu Y, Li P, Deng B, Xu M, Chen G. Self-Assembled Virus-Like Particle Vaccines via Fluorophilic Interactions Enable Infection Mimicry and Immune Protection. Adv Healthc Mater 2023; 12:e2301647. [PMID: 37703498 DOI: 10.1002/adhm.202301647] [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: 05/23/2023] [Revised: 09/10/2023] [Indexed: 09/15/2023]
Abstract
Influenza epidemics persistently threaten global health. Vaccines based on virus-like particles (VLPs), which resemble the native conformation of viruses, have emerged as vaccine candidates. However, the production of VLPs via genetic engineering remains constrained by challenges such as low yields, high costs, and being time consuming. In this study, a novel VLP platform is developed that could mimic infection and confer influenza protection through fluorination-driven self-assembly. The VLPs closely mimick the key steps in viral infection including dendritic cell (DC) attachment and pH-responsive endo-lysosomal escape, which enhances DC maturation and antigen cross-presentation. It is also observed that the VLPs migrate from the injection site to the draining lymph nodes efficiently. Immunization with VLPs triggers both Th1 and Th2 cellular responses, thereby inducing an improved CD8+ T cell response along with strong antigen-specific antibody responses. In several infected mouse models, VLP vaccines ameliorate weight loss, lung virus titers, pulmonary pathologies, and confer full protection against H1N1, H6N2, H9N2, and mixed influenza viruses. Therefore, the results support the potential of VLPs as an effective influenza vaccine with improved immune potency against infection. A methodology to generate VLPs based on fluorophilic interactions, which can be a general approach for development of pathogenic VLPs, is reported.
Collapse
Affiliation(s)
- Yinhe Xia
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Kai Liu
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Fei Wang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Zhou Xu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Yuesheng Wang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Rongling Zong
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Yemin Xu
- Department of Gastroenterology, Affiliated Hospital, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Ping Li
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Bin Deng
- Department of Gastroenterology, Affiliated Hospital, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Maolei Xu
- The Key Laboratory of Traditional Chinese Medicine Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine, School of Pharmacy, Binzhou Medical University, Yantai, 264003, P. R. China
| | - Gang Chen
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| |
Collapse
|
80
|
Han AX, de Jong SPJ, Russell CA. Co-evolution of immunity and seasonal influenza viruses. Nat Rev Microbiol 2023; 21:805-817. [PMID: 37532870 DOI: 10.1038/s41579-023-00945-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2023] [Indexed: 08/04/2023]
Abstract
Seasonal influenza viruses cause recurring global epidemics by continually evolving to escape host immunity. The viral constraints and host immune responses that limit and drive the evolution of these viruses are increasingly well understood. However, it remains unclear how most of these advances improve the capacity to reduce the impact of seasonal influenza viruses on human health. In this Review, we synthesize recent progress made in understanding the interplay between the evolution of immunity induced by previous infections or vaccination and the evolution of seasonal influenza viruses driven by the heterogeneous accumulation of antibody-mediated immunity in humans. We discuss the functional constraints that limit the evolution of the viruses, the within-host evolutionary processes that drive the emergence of new virus variants, as well as current and prospective options for influenza virus control, including the viral and immunological barriers that must be overcome to improve the effectiveness of vaccines and antiviral drugs.
Collapse
Affiliation(s)
- Alvin X Han
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Simon P J de Jong
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Colin A Russell
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
- Department of Global Health, School of Public Health, Boston University, Boston, MA, USA.
| |
Collapse
|
81
|
Lin PH, Hsiao PJ, Pan CF, Liu MT, Wang JT, Ching C, Wu FY, Lin YH, Yang YC, Hsu LY, Yang HC, Wu UI. Association of vaccine-specific regulatory T cells with reduced antibody response to repeated influenza vaccination. Eur J Immunol 2023; 53:e2350525. [PMID: 37713727 DOI: 10.1002/eji.202350525] [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: 04/10/2023] [Revised: 08/04/2023] [Accepted: 09/14/2023] [Indexed: 09/17/2023]
Abstract
Repeated annual influenza vaccinations have been associated with reduced vaccine-induced antibody responses. This prospective study aimed to explore the role of vaccine antigen-specific regulatory T (Treg) cells in antibody response to repeated annual influenza vaccination. We analyzed pre- and postvaccination hemagglutination inhibition (HI) titers, seroconversion rates, seroprotection rates, vaccine antigen hemagglutinin (HA)-specific Treg cells, and conventional T (Tconv) cells. We compared these parameters between vaccinees with or without vaccine-induced seroconversion. Our multivariate logistic regression revealed that prior vaccination was significantly associated with a decreased likelihood of achieving seroconversion for both H1N1(adjusted OR, 0.03; 95% CI, 0.01-0.13) and H3N2 (adjusted OR, 0.09; 95% CI, 0.03-0.30). Furthermore, individuals who received repeated vaccinations had significantly higher levels of pre-existing HA-specific Treg cells than those who did not. We also found that vaccine-induced fold-increases in HI titers and seroconversion were negatively correlated with pre-existing HA-specific Treg cells and positively correlated with the ratio of Tconv to Treg cells. Overall, our findings suggest that repeated annual influenza vaccination is associated with a lower vaccine-induced antibody response and a higher frequency of vaccine-specific Treg cells. However, a lower frequency of pre-existing Treg cells correlates with a higher postvaccination antibody response.
Collapse
Affiliation(s)
- Pin-Hung Lin
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Po-Ju Hsiao
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Fu Pan
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Tsan Liu
- Center for Diagnostics and Vaccine Development, Centers for Disease Control, Taipei, Taiwan
| | - Jann-Tay Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi Ching
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Fang-Yi Wu
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Hsuan Lin
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chan Yang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Le-Yin Hsu
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Graduate Program of Data Science, National Taiwan University and Academia Sinica, Taipei, Taiwan
| | - Hung-Chih Yang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Un-In Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
| |
Collapse
|
82
|
Reischak D, Rivetti AV, Otaka JNP, Domingues CS, Freitas TDL, Cardoso FG, Montesino LO, da Silva ALS, Malta F, Amgarten D, Goés-Neto A, de Oliveira AF, Camargos MF. First report and genetic characterization of the highly pathogenic avian influenza A(H5N1) virus in Cabot's tern ( Thalasseus acuflavidus), Brazil. Vet Anim Sci 2023; 22:100319. [PMID: 38022721 PMCID: PMC10652201 DOI: 10.1016/j.vas.2023.100319] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023] Open
Abstract
In 2021, the H5N1 virus lineage 2.3.4.4b spread to the Americas, causing high mortality in wild and domestic avian populations. South American countries along the Pacific migratory route have reported wild bird deaths due to A/H5Nx virus since October 2022. However, limited genomic data resulted in no cases reported in Brazil until May 2023. Brazil reported its first case of highly pathogenic avian influenza virus (HPAI A/H5N1) in May 2023. The virus was detected in Cabot's tern specimen in Marataízes, Espírito Santo. Cases were also found in backyard poultry and other wild birds, but no human or commercial poultry cases occurred. HPAI poses risks to the poultry industry, food security, and public health. Researchers used next-gen sequencing and phylogenetic analysis to study the Brazilian sample. It confirmed its affiliation with the 2.3.4.4b clade and proximity to sequences from Chile and Peru. This sheds light on the spread and evolution of HPAI A/H5N1 in the Americas, emphasizing continuous monitoring to mitigate risks for both avian and human populations. Understanding the virus's genetics and transmission allows implementing effective control measures to protect public health and the poultry industry.
Collapse
Affiliation(s)
- Dilmara Reischak
- Ministério da Agricultura e Pecuária, Laboratório Federal de Defesa Agropecuária/SP, São Paulo 13100-105, Brazil
| | - Anselmo Vasconcelos Rivetti
- Ministério da Agricultura e Pecuária, Laboratório Federal de Defesa Agropecuária/MG, Minas Gerais 33250-220, Brazil
| | - Juliana Nabuco Pereira Otaka
- Ministério da Agricultura e Pecuária, Laboratório Federal de Defesa Agropecuária/SP, São Paulo 13100-105, Brazil
| | - Christian Steffe Domingues
- Ministério da Agricultura e Pecuária, Laboratório Federal de Defesa Agropecuária/SP, São Paulo 13100-105, Brazil
| | - Talita de Lima Freitas
- Ministério da Agricultura e Pecuária, Laboratório Federal de Defesa Agropecuária/SP, São Paulo 13100-105, Brazil
| | - Fernanda Gomes Cardoso
- Ministério da Agricultura e Pecuária, Laboratório Federal de Defesa Agropecuária/SP, São Paulo 13100-105, Brazil
| | - Lucas Oliveira Montesino
- Ministério da Agricultura e Pecuária, Laboratório Federal de Defesa Agropecuária/SP, São Paulo 13100-105, Brazil
| | - Ana Luiza Savioli da Silva
- Ministério da Agricultura e Pecuária, Laboratório Federal de Defesa Agropecuária/SP, São Paulo 13100-105, Brazil
| | - Fernanda Malta
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil
| | - Deyvid Amgarten
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil
| | - Aristóteles Goés-Neto
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Minas Gerais 31270-901, Brazil
| | - Antônio Fonseca de Oliveira
- Ministério da Agricultura e Pecuária, Laboratório Federal de Defesa Agropecuária/MG, Minas Gerais 33250-220, Brazil
| | - Marcelo Fernandes Camargos
- Ministério da Agricultura e Pecuária, Laboratório Federal de Defesa Agropecuária/MG, Minas Gerais 33250-220, Brazil
| |
Collapse
|
83
|
Gupta D, Mohan S. Influenza vaccine: a review on current scenario and future prospects. J Genet Eng Biotechnol 2023; 21:154. [PMID: 38030859 PMCID: PMC10686931 DOI: 10.1186/s43141-023-00581-y] [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: 06/29/2023] [Accepted: 10/28/2023] [Indexed: 12/01/2023]
Abstract
Vaccination is a crucial tool in preventing influenza, but it requires annual updates in vaccine composition due to the ever-changing nature of the flu virus. While healthcare and economic burdens have reduced, the virus remains a challenge. Research conducted over the past decade has revealed pathways for improvement through both basic and clinical studies. Viral surveillance plays a vital role in the better selection of candidate viruses for vaccines and the early detection of drug-resistant strains.This page offers a description of future vaccine developments and an overview of current vaccine options. In the coming years, we anticipate significant changes in vaccine production, moving away from traditional egg-based methods towards innovative technologies such as DNA and RNA vaccines. These newer approaches offer significant advantages over traditional egg-based and cell culture-based influenza vaccine manufacturing.
Collapse
Affiliation(s)
- Dipanshi Gupta
- Amity Institute of Biotechnology, Amity University Uttar Pradesh (AUUP), Sector-125, Noida, Uttar Pradesh, 201303, India
| | - Sumedha Mohan
- Amity Institute of Biotechnology, Amity University Uttar Pradesh (AUUP), Sector-125, Noida, Uttar Pradesh, 201303, India.
| |
Collapse
|
84
|
Teo QW, Wang Y, Lv H, Tan TJC, Lei R, Mao KJ, Wu NC. Stringent and complex sequence constraints of an IGHV1-69 broadly neutralizing antibody to influenza HA stem. Cell Rep 2023; 42:113410. [PMID: 37976161 PMCID: PMC10872586 DOI: 10.1016/j.celrep.2023.113410] [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: 07/27/2023] [Revised: 09/29/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023] Open
Abstract
IGHV1-69 is frequently utilized by broadly neutralizing influenza antibodies to the hemagglutinin (HA) stem. These IGHV1-69 HA stem antibodies have diverse complementarity-determining region (CDR) H3 sequences. Besides, their light chains have minimal to no contact with the epitope. Consequently, sequence determinants that confer IGHV1-69 antibodies with HA stem specificity remain largely elusive. Using high-throughput experiments, this study reveals the importance of light-chain sequence for the IGHV1-69 HA stem antibody CR9114, which is the broadest influenza antibody known to date. Moreover, we demonstrate that the CDR H3 sequences from many other IGHV1-69 antibodies, including those to the HA stem, are incompatible with CR9114. Along with mutagenesis and structural analysis, our results indicate that light-chain and CDR H3 sequences coordinately determine the HA stem specificity of IGHV1-69 antibodies. Overall, this work provides molecular insights into broadly neutralizing antibody responses to influenza virus, which have important implications for universal influenza vaccine development.
Collapse
Affiliation(s)
- Qi Wen Teo
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Yiquan Wang
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Huibin Lv
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Timothy J C Tan
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Ruipeng Lei
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Kevin J Mao
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Nicholas C Wu
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
85
|
Viney M, Cheynel L. Gut immune responses and evolution of the gut microbiome-a hypothesis. DISCOVERY IMMUNOLOGY 2023; 2:kyad025. [PMID: 38567055 PMCID: PMC10917216 DOI: 10.1093/discim/kyad025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/03/2023] [Accepted: 11/22/2023] [Indexed: 04/04/2024]
Abstract
The gut microbiome is an assemblage of microbes that have profound effects on their hosts. The composition of the microbiome is affected by bottom-up, among-taxa interactions and by top-down, host effects, which includes the host immune response. While the high-level composition of the microbiome is generally stable over time, component strains and genotypes will constantly be evolving, with both bottom-up and top-down effects acting as selection pressures, driving microbial evolution. Secretory IgA is a major feature of the gut's adaptive immune response, and a substantial proportion of gut bacteria are coated with IgA, though the effect of this on bacteria is unclear. Here we hypothesize that IgA binding to gut bacteria is a selection pressure that will drive the evolution of IgA-bound bacteria, so that they will have a different evolutionary trajectory than those bacteria not bound by IgA. We know very little about the microbiome of wild animals and even less about their gut immune responses, but it must be a priority to investigate this hypothesis to understand if and how host immune responses contribute to microbiome evolution.
Collapse
Affiliation(s)
- Mark Viney
- Department of Evolution, Ecology & Behaviour, University of Liverpool, Liverpool, UK
| | - Louise Cheynel
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, ENTPE, UMR 5023 LEHNA, Villeurbanne, France
| |
Collapse
|
86
|
Zafeiriadou A, Kaltsis L, Kostakis M, Kapes V, Thomaidis NS, Markou A. Wastewater surveillance of the most common circulating respiratory viruses in Athens: The impact of COVID-19 on their seasonality. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 900:166136. [PMID: 37567285 DOI: 10.1016/j.scitotenv.2023.166136] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/06/2023] [Accepted: 08/06/2023] [Indexed: 08/13/2023]
Abstract
Due to governments' actions to contain the spread of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the activity of common circulating respiratory viruses was significantly disrupted after the COVID-19 pandemic and thorough surveillance of respiratory pathogens was considered essential worldwide. Wastewater-based epidemiology has proven to be a valuable tool, that provides complementary information on disease outbreaks and is increasingly used to study the infection dynamics of other viruses, apart from SARS-CoV-2. The aims of the present study were the detection of four commonly circulating respiratory viruses: SARS-CoV-2, influenza A, B and Human Respiratory Syncytial Virus (RSV), the evaluation of the COVID-19 pandemic impact on their seasonality and the determination of the possible common trends in the viral load of these viruses in the wastewater of the Attica region. A standardized and validated concentration and extraction protocol was used, generic for all four viruses, followed by Reverse Transcription quantitative Polymerase Chain Reaction (RT-qPCR) assays. The study proved that there was a prolonged period when all four viruses circulated in the population and an early outbreak of seasonal influenza and RSV in 2022-2023, compared to data from the pre-COVID-19 period. SARS-CoV-2, influenza A and RSV concentrations showed peak levels during December, followed by a slight decline in influenza A concentrations, followed by steady increase of influenza B concentrations in January 2023. SARS-CoV-2 was the dominant virus throughout the whole study period. This is the first study in Greece that investigated the most common circulating viruses simultaneously and in one of the largest timelines, providing crucial information about their infection dynamics during a period when an outbreak of respiratory diseases was declared by the National Public Health Organization. Presented results highlight the establishment of environmental surveillance as a non-invasive and complementary virus outbreak monitoring tool and the importance of influenza A, B and RSV integration into a wastewater-based surveillance system to help in disease management.
Collapse
Affiliation(s)
- Anastasia Zafeiriadou
- Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, University Campus, Zografou, 15771, Athens, Greece
| | - Lazaros Kaltsis
- Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, University Campus, Zografou, 15771, Athens, Greece
| | - Marios Kostakis
- Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, University Campus, Zografou, 15771, Athens, Greece
| | - Vasileios Kapes
- Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, University Campus, Zografou, 15771, Athens, Greece
| | - Nikolaos S Thomaidis
- Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, University Campus, Zografou, 15771, Athens, Greece
| | - Athina Markou
- Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, University Campus, Zografou, 15771, Athens, Greece.
| |
Collapse
|
87
|
Zhao JH, Wang YW, Yang J, Tong ZJ, Wu JZ, Wang YB, Wang QX, Li QQ, Yu YC, Leng XJ, Chang L, Xue X, Sun SL, Li HM, Ding N, Duan JA, Li NG, Shi ZH. Natural products as potential lead compounds to develop new antiviral drugs over the past decade. Eur J Med Chem 2023; 260:115726. [PMID: 37597436 DOI: 10.1016/j.ejmech.2023.115726] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/22/2023] [Accepted: 08/13/2023] [Indexed: 08/21/2023]
Abstract
Virus infection has been one of the main causes of human death since the ancient times. Even though more and more antiviral drugs have been approved in clinic, long-term use can easily lead to the emergence of drug resistance and side effects. Fortunately, there are many kinds of metabolites which were produced by plants, marine organisms and microorganisms in nature with rich structural skeletons, and they are natural treasure house for people to find antiviral active substances. Aiming at many types of viruses that had caused serious harm to human health in recent years, this review summarizes the natural products with antiviral activity that had been reported for the first time in the past ten years, we also sort out the source, chemical structure and safety indicators in order to provide potential lead compounds for the research and development of new antiviral drugs.
Collapse
Affiliation(s)
- Jing-Han Zhao
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Yue-Wei Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Jin Yang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Zhen-Jiang Tong
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Jia-Zhen Wu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Yi-Bo Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Qing-Xin Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Qing-Qing Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Yan-Cheng Yu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Xue-Jiao Leng
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Liang Chang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Xin Xue
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Shan-Liang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - He-Min Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Ning Ding
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China.
| | - Jin-Ao Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China.
| | - Nian-Guang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China.
| | - Zhi-Hao Shi
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
88
|
Potdar V, Vijay N, Vipat V, Jadhav S, Gupta N, Aggarwal N. External quality assessment for laboratories in pan-India ILI/SARI surveillance for simultaneous detection of influenza virus and SARS-CoV-2. Front Public Health 2023; 11:1274508. [PMID: 38026348 PMCID: PMC10679675 DOI: 10.3389/fpubh.2023.1274508] [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: 08/08/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The Indian Council of Medical Research has set up a nationwide network of 28 laboratories for simultaneous surveillance of influenza virus and SARS-CoV-2 in ILI/SARI patients, using an in-house developed and validated multiplex real-time RTPCR assay. The aim of this study was to ensure the quality of testing by these laboratories by implementing an external quality assessment program (EQAP). Methods For this EQAP, a proficiency test (PT) panel comprising tissue-culture or egg-grown influenza virus and SARS-CoV-2 was developed. The PT panel was distributed to all the participant laboratories, which tested the panel and submitted the qualitative results online to the EQAP provider. The performance of the laboratories was evaluated on qualitative criteria but cycle threshold (Ct) values were also gathered for each sample. Results On a qualitative basis, all the laboratories achieved the criteria of 90% concordance with the results of the PT panel provider. Ct values of different samples across the laboratories were within ≤ ±3 cycles of the corresponding mean values of the respective sample. The results of this EQAP affirmed the quality and reliability of testing being done for simultaneous surveillance of influenza virus and SARS-CoV-2 in India.
Collapse
Affiliation(s)
- Varsha Potdar
- Indian Council of Medical Research–National Institute of Virology, Pune, India
| | - Neetu Vijay
- Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research, New Delhi, India
| | - Veena Vipat
- Indian Council of Medical Research–National Institute of Virology, Pune, India
| | - Sheetal Jadhav
- Indian Council of Medical Research–National Institute of Virology, Pune, India
| | - Nivedita Gupta
- Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research, New Delhi, India
| | - Neeraj Aggarwal
- Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research, New Delhi, India
| |
Collapse
|
89
|
Zhu M, Anirudhan V, Du R, Rong L, Cui Q. Influenza virus cell entry and targeted antiviral development. J Med Virol 2023; 95:e29181. [PMID: 37930075 DOI: 10.1002/jmv.29181] [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: 09/04/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
Influenza virus infection is currently one of the most prevalent and transmissible diseases in the world causing local outbreaks every year. It has the potential to cause devastating global pandemics as well. The development of anti-influenza drugs possessing novel mechanisms of action is urgently needed to control the spread of influenza infections; thus, drugs that inhibit influenza virus entry into target cells are emerging as a hot research topic. In addition to discussing the biological significance of hemagglutinin in viral replication, this article provides recent updates on the natural products, small molecules, proteins, peptides, and neutralizing antibody-like proteins that have anti-influenza potency.
Collapse
Affiliation(s)
- Murong Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Varada Anirudhan
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ruikun Du
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Qinghua Cui
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| |
Collapse
|
90
|
Huang CQ, Vishwanath S, Carnell GW, Chan ACY, Heeney JL. Immune imprinting and next-generation coronavirus vaccines. Nat Microbiol 2023; 8:1971-1985. [PMID: 37932355 DOI: 10.1038/s41564-023-01505-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 09/13/2023] [Indexed: 11/08/2023]
Abstract
Vaccines based on historical virus isolates provide limited protection from continuously evolving RNA viruses, such as influenza viruses or coronaviruses, which occasionally spill over between animals and humans. Despite repeated booster immunizations, population-wide declines in the neutralization of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have occurred. This has been compared to seasonal influenza vaccinations in humans, where the breadth of immune responses induced by repeat exposures to antigenically distinct influenza viruses is confounded by pre-existing immunity-a mechanism known as imprinting. Since its emergence, SARS-CoV-2 has evolved in a population with partial immunity, acquired by infection, vaccination or both. Here we critically examine the evidence for and against immune imprinting in host humoral responses to SARS-CoV-2 and its implications for coronavirus disease 2019 (COVID-19) booster vaccine programmes.
Collapse
Affiliation(s)
- Chloe Qingzhou Huang
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Sneha Vishwanath
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - George William Carnell
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Andrew Chun Yue Chan
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Jonathan Luke Heeney
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
91
|
Li Z, Xiong Y, Long J, Li T, Fu X, Yang S, Tian D, Zhao Y, Qi L. Resurgence of influenza during COVID-19 in Chongqing, China: A retrospective analysis. J Med Virol 2023; 95:e29249. [PMID: 38009822 DOI: 10.1002/jmv.29249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/12/2023] [Accepted: 11/11/2023] [Indexed: 11/29/2023]
Abstract
To better understand the trends of influenza and the impact of public health and social measures (PHSMs) implemented during the coronavirus disease 2019 (COVID-19) period in Chongqing, China. Data from the China Influenza Surveillance Information System from January 2017 to June 2022 were extracted. Epidemiological characteristics (influenza-like illness [ILI] and ILI%) and virological characteristics (influenza positive rate and circulating (sub)types) of influenza were described and compared between the pre-COVID-19 period and the COVID-19 period. Our survey showed that the implementation of PHSMs during the COVID-19 period had a positive impact on reducing influenza transmission. However, influenza activity resurged in 2021-2022 as the PHSMs were eased. Children under 5 years old constituted the highest proportion of ILI cases. The overall influenza positive rate was 23.70%, with a higher rate observed during the pre-COVID-19 period (31.55%) compared to the COVID-19 period (13.68%). Influenza virus subtypes co-circulated and the predominant subtype varied each year, with influenza A subtypes predominated in 2018/2019, while influenza B/Victoria lineage dominated in 2020/2021. PHSMs are effective measures to mitigate the spread of influenza. The findings underscore the need for bolstering monitoring systems, advocating influenza vaccination, and implementing practical PHSMs to strengthen prevention and control measures against influenza.
Collapse
Affiliation(s)
- Zhourong Li
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing, China
- School of Public Health, Chongqing Medical University, Chongqing, China
| | - Yu Xiong
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing, China
- Chongqing Municipal Key Laboratory for High Pathogenic Microbes, Chongqing, China
| | - Jiang Long
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing, China
- Chongqing Municipal Key Laboratory for High Pathogenic Microbes, Chongqing, China
| | - Tingting Li
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing, China
- Chongqing Municipal Key Laboratory for High Pathogenic Microbes, Chongqing, China
| | - Xiaoqing Fu
- Southwest Medical University, Sichuan, China
| | - Shuang Yang
- School of Public Health, Chongqing Medical University, Chongqing, China
| | - Dechao Tian
- Department of Biostatistics and Systems Biology, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yong Zhao
- School of Public Health, Chongqing Medical University, Chongqing, China
| | - Li Qi
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing, China
- Chongqing Municipal Key Laboratory for High Pathogenic Microbes, Chongqing, China
| |
Collapse
|
92
|
Sobhani K, Cheng S, Binder RA, Mantis NJ, Crawford JM, Okoye N, Braun JG, Joung S, Wang M, Lozanski G, King CL, Roback JD, Granger DA, Boppana SB, Karger AB. Clinical Utility of SARS-CoV-2 Serological Testing and Defining a Correlate of Protection. Vaccines (Basel) 2023; 11:1644. [PMID: 38005976 PMCID: PMC10674881 DOI: 10.3390/vaccines11111644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/12/2023] [Accepted: 10/18/2023] [Indexed: 11/26/2023] Open
Abstract
Herein, we review established clinical use cases for SARS-CoV-2 antibody measures, which include diagnosis of recent prior infection, isolating high titer convalescent plasma, diagnosing multisystem inflammatory syndrome in children (MIS-C), and booster dosing in the immunosuppressed and other populations. We then address whether an antibody correlate of protection (CoP) for SARS-CoV-2 has been successfully defined with the following considerations: Antibody responses in the immunocompetent, vaccine type, variants, use of binding antibody tests vs. neutralization tests, and endpoint measures. In the transition from the COVID-19 pandemic to endemic, there has been much interest in defining an antibody CoP. Due to the high mutability of respiratory viruses and our current knowledge of SARS-CoV-2 variants defining a CoP for prevention of infection is unrealistic. However, a CoP may be defined for prevention of severe disease requiring hospitalization and/or death. Most SARS-CoV-2 CoP research has focused on neutralization measurements. However, there can be significant differences in neutralization test methods, and disparate responses to new variants depending on format. Furthermore, neutralization assays are often impractical for high throughput applications (e.g., assessing humoral immune response in populations or large cohorts). Nevertheless, CoP studies using neutralization measures are reviewed to determine where there is consensus. Alternatively, binding antibody tests could be used to define a CoP. Binding antibody assays tend to be highly automatable, high throughput, and therefore practical for large population applications. Again, we review studies for consensus on binding antibody responses to vaccines, focusing on standardized results. Binding antibodies directed against the S1 receptor binding domain (S1-RBD) of the viral spike protein can provide a practical, indirect measure of neutralization. Initially, a response for S1-RBD antibodies may be selected that reflects the peak response in immunocompetent populations and may serve as a target for booster dosing in the immunocompromised. From existing studies reporting peak S1-RBD responses in standardized units, an approximate range of 1372-2744 BAU/mL for mRNA and recombinant protein vaccines was extracted that could serve as an initial CoP target. This target would need to be confirmed and potentially adjusted for updated vaccines, and almost certainly for other vaccine formats (i.e., viral vector). Alternatively, a threshold or response could be defined based on outcomes over time (i.e., prevention of severe disease). We also discuss the precedent for clinical measurement of antibodies for vaccine-preventable diseases (e.g., hepatitis B). Lastly, cellular immunity is briefly addressed for its importance in the nature and durability of protection.
Collapse
Affiliation(s)
- Kimia Sobhani
- Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (S.C.)
| | - Raquel A. Binder
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Nicholas J. Mantis
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY 12222, USA
| | - James M. Crawford
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Nkemakonam Okoye
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Jonathan G. Braun
- Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
- F. Widjaja Inflammatory Bowel Disease Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sandy Joung
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (S.C.)
| | - Minhao Wang
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (S.C.)
| | - Gerard Lozanski
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Christopher L. King
- Department of Pathology, Case Western Reserve University and Veterans Affairs Research Service, Cleveland, OH 44106, USA
| | - John D. Roback
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Douglas A. Granger
- Institute for Interdisciplinary Salivary Bioscience Research, University of California Irvine, Irvine, CA 92697, USA
| | - Suresh B. Boppana
- Department of Pediatrics and Department of Microbiology, Heersink School of Medicine, UAB, Birmingham, AL 35233, USA
| | - Amy B. Karger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
93
|
Li Y, Mo J, Liu J, Liang Y, Deng C, Huang Z, Jiang J, Liu M, Liu X, Shang L, Wang X, Xie X, Wang J. A micro-electroporation/electrophoresis-based vaccine screening system reveals the impact of vaccination orders on cross-protective immunity. iScience 2023; 26:108086. [PMID: 37860767 PMCID: PMC10582514 DOI: 10.1016/j.isci.2023.108086] [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: 07/13/2023] [Revised: 08/31/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023] Open
Abstract
The constant emergence of mutated pathogens poses great challenges to the existing vaccine system. A screening system is needed to screen for antigen designs and vaccination strategies capable of inducing cross-protective immunity. Herein, we report a screening system based on DNA vaccines and a micro-electroporation/electrophoresis system (MEES), which greatly improved the efficacy of DNA vaccines, elevating humoral and cellular immune responses by over 400- and 35-fold respectively. Eighteen vaccination strategies were screened simultaneously by sequential immunization with vaccines derived from wildtype (WT) SARS-CoV-2, Delta, or Omicron BA.1 variant. Sequential vaccination of BA.1-WT-Delta vaccines with MEES induced potent neutralizing antibodies against all three viral strains and BA.5 variant, demonstrating that cross-protective immunity against future mutants can be successfully induced by existing strain-derived vaccines when a proper combination and order of sequential vaccination are used. Our screening system could be used for fast-seeking vaccination strategies for emerging pathogens in the future.
Collapse
Affiliation(s)
- Yongyong Li
- Division of Pulmonary and Critical Care Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510080, Peoples Republic of China
| | - Jingshan Mo
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, People’s Republic of China
- School of Electronic and Information Engineering, Guangdong Ocean University, Zhanjiang 524088, People’s Republic of China
| | - Jing Liu
- Division of Pulmonary and Critical Care Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510080, Peoples Republic of China
| | - Ying Liang
- Department of Nephrology, GuangZhou Eighth People′s Hospital, GuangZhou Medical University, Guangzhou 510060, People’s Republic of China
| | - Caiguanxi Deng
- Division of Pulmonary and Critical Care Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510080, Peoples Republic of China
| | - Zhangping Huang
- Division of Pulmonary and Critical Care Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510080, Peoples Republic of China
| | - Juan Jiang
- Division of Pulmonary and Critical Care Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510080, Peoples Republic of China
| | - Ming Liu
- Division of Pulmonary and Critical Care Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510080, Peoples Republic of China
| | - Xinmin Liu
- Division of Pulmonary and Critical Care Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510080, Peoples Republic of China
| | - Liru Shang
- Division of Pulmonary and Critical Care Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510080, Peoples Republic of China
| | - Xiafeng Wang
- Division of Pulmonary and Critical Care Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510080, Peoples Republic of China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, People’s Republic of China
| | - Ji Wang
- Division of Pulmonary and Critical Care Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510080, Peoples Republic of China
| |
Collapse
|
94
|
Peissert F, Pedotti M, Corbellari R, Simonelli L, De Gasparo R, Tamagnini E, Plüss L, Elsayed A, Matasci M, De Luca R, Cassaniti I, Sammartino JC, Piralla A, Baldanti F, Neri D, Varani L. Adapting Neutralizing Antibodies to Viral Variants by Structure-Guided Affinity Maturation Using Phage Display Technology. GLOBAL CHALLENGES (HOBOKEN, NJ) 2023; 7:2300088. [PMID: 37829677 PMCID: PMC10566804 DOI: 10.1002/gch2.202300088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Indexed: 10/14/2023]
Abstract
Neutralizing monoclonal antibodies have achieved great efficacy and safety for the treatment of numerous infectious diseases. However, their neutralization potency is often rapidly lost when the target antigen mutates. Instead of isolating new antibodies each time a pathogen variant arises, it can be attractive to adapt existing antibodies, making them active against the new variant. Potential benefits of this approach include reduced development time, cost, and regulatory burden. Here a methodology is described to rapidly evolve neutralizing antibodies of proven activity, improving their function against new pathogen variants without losing efficacy against previous ones. The reported procedure is based on structure-guided affinity maturation using combinatorial mutagenesis and phage display technology. Its use against the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is demonstrated, but it is suitable for any other pathogen. As proof of concept, the method is applied to CoV-X2, a human bispecific antibody that binds with high affinity to the early SARS-CoV-2 variants but lost neutralization potency against Delta. Antibodies emerging from the affinity maturation selection exhibit significantly improved neutralization potency against Delta and no loss of efficacy against the other viral sequences tested. These results illustrate the potential application of structure-guided affinity maturation in facilitating the rapid adaptation of neutralizing antibodies to pathogen variants.
Collapse
Affiliation(s)
| | - Mattia Pedotti
- Institute for Research in BiomedicineUniversità della Svizzera italiana (USI)Bellinzona6500Switzerland
| | | | - Luca Simonelli
- Institute for Research in BiomedicineUniversità della Svizzera italiana (USI)Bellinzona6500Switzerland
| | - Raoul De Gasparo
- Institute for Research in BiomedicineUniversità della Svizzera italiana (USI)Bellinzona6500Switzerland
| | - Elia Tamagnini
- Institute for Research in BiomedicineUniversità della Svizzera italiana (USI)Bellinzona6500Switzerland
| | - Louis Plüss
- Philochem AGLibernstrasse 3Otelfingen8112Switzerland
| | | | | | | | - Irene Cassaniti
- Molecular Virology UnitMicrobiology and Virology DepartmentFondazione IRCCS Policlinico San MatteoPavia27100Italy
| | - Jose’ Camilla Sammartino
- Molecular Virology UnitMicrobiology and Virology DepartmentFondazione IRCCS Policlinico San MatteoPavia27100Italy
| | - Antonio Piralla
- Molecular Virology UnitMicrobiology and Virology DepartmentFondazione IRCCS Policlinico San MatteoPavia27100Italy
| | - Fausto Baldanti
- Molecular Virology UnitMicrobiology and Virology DepartmentFondazione IRCCS Policlinico San MatteoPavia27100Italy
- Department of Clinical Surgical Diagnostic and Pediatric SciencesUniversità degli Studi di PaviaPavia27100Italy
| | - Dario Neri
- Philochem AGLibernstrasse 3Otelfingen8112Switzerland
- Philogen SpALocalità Bellaria 35Sovicille (SI)53018Italy
| | - Luca Varani
- Institute for Research in BiomedicineUniversità della Svizzera italiana (USI)Bellinzona6500Switzerland
| |
Collapse
|
95
|
Xu S, Yu L, Teng Q, Li X, Jin Z, Qu Y, Li J, Zhang Q, Li Z, Zhao K. Enhance immune response to H9 AIV DNA vaccine based on polygene expression and DGL nanoparticle encapsulation. Poult Sci 2023; 102:102925. [PMID: 37542938 PMCID: PMC10428121 DOI: 10.1016/j.psj.2023.102925] [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: 04/06/2023] [Revised: 06/29/2023] [Accepted: 07/02/2023] [Indexed: 08/07/2023] Open
Abstract
DNA vaccination has great potential to treat or prevent avian influenza pandemics, but the technique may be limited by low immunogenicity and gene delivery in clinical testing. Here, to improve the immune efficacy of DNA vaccines against avian influenza, we prepared and tested the immunogenicity of 4 recombinant DNA vaccines containing 2 or 3 AIV antigens. The results revealed that chickens and mice immunized with plasmid DNA containing 3 antigens (HA gene from H9N2, and NA and HA genes from H5N1) exhibited a robust immune response than chickens and mice immunized with plasmid DNAs containing 2 antigenic genes. Subsequently, this study used pβH9N1SH5 as a model antigen to study the effect of dendritic polylysine (DGL) nanoparticles as a gene delivery system and adjuvant on antigen-specific immunity in mice models. At a ratio of 1:3 DGL/pβH9N1SH5 (w/w), the pβH9N1SH5/DGL NPs showed excellent physical and chemical properties, induced higher levels of HI antibodies, and larger CD3+/CD4+ T lymphocyte and CD3+/CD8+ T lymphocyte population, as well as the production of cytokines, namely, interferon (IFN)-γ, interleukin (IL)-2 compared with the naked pβH9N1SH5. Therefore, multiantigen gene expression methods using DGL as a delivery system may have broad application prospects in gene therapy.
Collapse
Affiliation(s)
- Shangen Xu
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou 318000, China
| | - Lu Yu
- Department of Avian Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Qiaoyang Teng
- Department of Avian Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Xuesong Li
- Department of Avian Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Zheng Jin
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou 318000, China
| | - Yang Qu
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou 318000, China
| | - Jiawei Li
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou 318000, China
| | - Qihong Zhang
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou 318000, China
| | - Zejun Li
- Department of Avian Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China.
| | - Kai Zhao
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou 318000, China.
| |
Collapse
|
96
|
Zhang G, Tang T, Chen Y, Huang X, Liang T. mRNA vaccines in disease prevention and treatment. Signal Transduct Target Ther 2023; 8:365. [PMID: 37726283 PMCID: PMC10509165 DOI: 10.1038/s41392-023-01579-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/01/2023] [Accepted: 07/30/2023] [Indexed: 09/21/2023] Open
Abstract
mRNA vaccines have emerged as highly effective strategies in the prophylaxis and treatment of diseases, thanks largely although not totally to their extraordinary performance in recent years against the worldwide plague COVID-19. The huge superiority of mRNA vaccines regarding their efficacy, safety, and large-scale manufacture encourages pharmaceutical industries and biotechnology companies to expand their application to a diverse array of diseases, despite the nonnegligible problems in design, fabrication, and mode of administration. This review delves into the technical underpinnings of mRNA vaccines, covering mRNA design, synthesis, delivery, and adjuvant technologies. Moreover, this review presents a systematic retrospective analysis in a logical and well-organized manner, shedding light on representative mRNA vaccines employed in various diseases. The scope extends across infectious diseases, cancers, immunological diseases, tissue damages, and rare diseases, showcasing the versatility and potential of mRNA vaccines in diverse therapeutic areas. Furthermore, this review engages in a prospective discussion regarding the current challenge and potential direction for the advancement and utilization of mRNA vaccines. Overall, this comprehensive review serves as a valuable resource for researchers, clinicians, and industry professionals, providing a comprehensive understanding of the technical aspects, historical context, and future prospects of mRNA vaccines in the fight against various diseases.
Collapse
Affiliation(s)
- Gang Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, 310003, Hangzhou, Zhejiang, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, 310009, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Tianyu Tang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, 310003, Hangzhou, Zhejiang, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, 310009, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Yinfeng Chen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, 310003, Hangzhou, Zhejiang, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, 310009, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China.
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, 310003, Hangzhou, Zhejiang, China.
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, 310009, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China.
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, 310003, Hangzhou, Zhejiang, China.
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, 310009, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
| |
Collapse
|
97
|
Rabalski L, Kosinski M, Cybulski P, Stadejek T, Lepek K. Genetic Diversity of Type A Influenza Viruses Found in Swine Herds in Northwestern Poland from 2017 to 2019: The One Health Perspective. Viruses 2023; 15:1893. [PMID: 37766299 PMCID: PMC10536349 DOI: 10.3390/v15091893] [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: 08/04/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Influenza A viruses (IAV) are still a cause of concern for public health and veterinary services worldwide. With (-) RNA-segmented genome architecture, influenza viruses are prone to reassortment and can generate a great variety of strains, some capable of crossing interspecies barriers. Seasonal IAV strains continuously spread from humans to pigs, leading to multiple reassortation events with strains endemic to swine. Due to its high adaptability to humans, a reassortant strain based on "human-like" genes could potentially be a carrier of avian origin segments responsible for high virulence, and hence become the next pandemic strain with unseen pathogenicity. The rapid evolution of sequencing methods has provided a fast and cost-efficient way to assess the genetic diversity of IAV. In this study, we investigated the genetic diversity of swine influenza viruses (swIAVs) collected from Polish farms. A total of 376 samples were collected from 11 farms. The infection was confirmed in 112 cases. The isolates were subjected to next-generation sequencing (NGS), resulting in 93 full genome sequences. Phylogenetic analysis classified 59 isolates as genotype T (H1avN2g) and 34 isolates as genotype P (H1pdmN1pdm), all of which had an internal gene cassette (IGC) derived from the H1N1pdm09-like strain. These data are consistent with evolutionary trends in European swIAVs. The applied methodology proved to be useful in monitoring the genetic diversity of IAV at the human-animal interface.
Collapse
Affiliation(s)
- Lukasz Rabalski
- Laboratory of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland
- Biological Threats Identification and Countermeasure Center of the General Karol Kaczkowski Military Institute of Hygiene and Epidemiology, Lubelska 4 St, 24-100 Pulawy, Poland
| | - Maciej Kosinski
- Laboratory of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland
| | - Piotr Cybulski
- Goodvalley Agro S.A., Dworcowa 25, 77-320 Przechlewo, Poland
| | - Tomasz Stadejek
- Department of Pathology and Veterinary Diagnostic, Institute of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, 02-776 Warsaw, Poland
| | - Krzysztof Lepek
- Laboratory of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland
| |
Collapse
|
98
|
Sun X, Hu X, Zhang Q, Zhao L, Sun X, Yang L, Jin M. Sodium taurocholate hydrate inhibits influenza virus replication and suppresses influenza a Virus-triggered inflammation in vitro and in vivo. Int Immunopharmacol 2023; 122:110544. [PMID: 37392567 DOI: 10.1016/j.intimp.2023.110544] [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: 04/10/2023] [Revised: 06/07/2023] [Accepted: 06/17/2023] [Indexed: 07/03/2023]
Abstract
Influenza A virus is an important respiratory pathogen that poses serious threats to human health. Owing to the high mutation rate of viral genes, weaker cross-protection of vaccines, and rapid emergence of drug resistance, there is an urgent need to develop new antiviral drugs against influenza viruses. Taurocholic acid is a primary bile acid that promotes digestion, absorption, and excretion of dietary lipids. Here, we demonstrate that sodium taurocholate hydrate (STH) exhibits broad-spectrum antiviral activity against influenza strains H5N6, H1N1, H3N2, H5N1, and H9N2 in vitro. STH significantly inhibited the early stages of influenza A virus replication. The levels of influenza virus viral RNA (vRNA), complementary RNA (cRNA), and mRNA were specifically reduced in virus-infected cells following STH treatment. In vivo, STH treatment of infected mice alleviated clinical signs and reduced weight loss and mortality. STH also reduced TNF-α, IL-1β, and IL-6 overexpression. STH significantly inhibited the upregulation of TLR4 and the NF-kB family member p65, both in vivo and in vitro. These results suggest that STH exerts a protective effect against influenza infection via suppression of the NF-kB pathway, highlighting the potential use of STH as a drug for treating influenza infection.
Collapse
Affiliation(s)
- Xiaolu Sun
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan, Hubei, China
| | - Xiaotong Hu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan, Hubei, China
| | - Qiang Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China; College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Li Zhao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, China
| | - Xiaomei Sun
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan, Hubei, China
| | - Li Yang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan, Hubei, China
| | - Meilin Jin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan, Hubei, China.
| |
Collapse
|
99
|
Einav T, Ma R. Using interpretable machine learning to extend heterogeneous antibody-virus datasets. CELL REPORTS METHODS 2023; 3:100540. [PMID: 37671020 PMCID: PMC10475791 DOI: 10.1016/j.crmeth.2023.100540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 05/14/2023] [Accepted: 06/30/2023] [Indexed: 09/07/2023]
Abstract
A central challenge in biology is to use existing measurements to predict the outcomes of future experiments. For the rapidly evolving influenza virus, variants examined in one study will often have little to no overlap with other studies, making it difficult to discern patterns or unify datasets. We develop a computational framework that predicts how an antibody or serum would inhibit any variant from any other study. We validate this method using hemagglutination inhibition data from seven studies and predict 2,000,000 new values ± uncertainties. Our analysis quantifies the transferability between vaccination and infection studies in humans and ferrets, shows that serum potency is negatively correlated with breadth, and provides a tool for pandemic preparedness. In essence, this approach enables a shift in perspective when analyzing data from "what you see is what you get" into "what anyone sees is what everyone gets."
Collapse
Affiliation(s)
- Tal Einav
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Rong Ma
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
100
|
N’Guessan A, Kailasam S, Mostefai F, Poujol R, Grenier JC, Ismailova N, Contini P, De Palma R, Haber C, Stadler V, Bourque G, Hussin JG, Shapiro BJ, Fritz JH, Piccirillo CA. Selection for immune evasion in SARS-CoV-2 revealed by high-resolution epitope mapping and sequence analysis. iScience 2023; 26:107394. [PMID: 37599818 PMCID: PMC10433132 DOI: 10.1016/j.isci.2023.107394] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 02/10/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023] Open
Abstract
Here, we exploit a deep serological profiling strategy coupled with an integrated, computational framework for the analysis of SARS-CoV-2 humoral immune responses. Applying a high-density peptide array (HDPA) spanning the entire proteomes of SARS-CoV-2 and endemic human coronaviruses allowed identification of B cell epitopes and relate them to their evolutionary and structural properties. We identify hotspots of pre-existing immunity and identify cross-reactive epitopes that contribute to increasing the overall humoral immune response to SARS-CoV-2. Using a public dataset of over 38,000 viral genomes from the early phase of the pandemic, capturing both inter- and within-host genetic viral diversity, we determined the evolutionary profile of epitopes and the differences across proteins, waves, and SARS-CoV-2 variants. Lastly, we show that mutations in spike and nucleocapsid epitopes are under stronger selection between than within patients, suggesting that most of the selective pressure for immune evasion occurs upon transmission between hosts.
Collapse
Affiliation(s)
- Arnaud N’Guessan
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill Genome Centre, McGill University, Montréal, QC, Canada
| | - Senthilkumar Kailasam
- Canadian Center for Computational Genomics, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Fatima Mostefai
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC, Canada
| | - Raphaël Poujol
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
| | | | - Nailya Ismailova
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Paola Contini
- Department of Internal Medicine, University of Genoa and IRCCS IST-Ospedale San Martino, Genoa, Italy
| | - Raffaele De Palma
- Department of Internal Medicine, University of Genoa and IRCCS IST-Ospedale San Martino, Genoa, Italy
| | | | | | - Guillaume Bourque
- Canadian Center for Computational Genomics, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Julie G. Hussin
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Département de Médecine, Université de Montréal, Montréal, QC, Canada
| | - B. Jesse Shapiro
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill Genome Centre, McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Jörg H. Fritz
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Ciriaco A. Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program of the Research Institute of McGill Health Center, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| |
Collapse
|