1
|
Nugroho CMH, Silaen OSM, Kurnia RS, Krisnamurti DGB, Putra MA, Indrawati A, Poetri ON, Wibawan IWT, Widyaningtyas ST, Soebandrio A. In vitro antiviral activity of NanB bacterial sialidase against avian influenza H9N2 virus in MDCK cells. Avian Pathol 2025; 54:96-107. [PMID: 39069790 DOI: 10.1080/03079457.2024.2386315] [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: 10/10/2023] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
The avian influenza virus is an infectious agent that may cause global health problems in poultry and is potentially zoonotic. In the recent decades, bacterial-derived sialidases have been extensively studied for their ability to inhibit avian influenza virus infections. In this study, the antiviral activity of NanB sialidase from Pasteurella multocida was investigated through in vitro analysis using Madin-Darby canine kidney (MDCK) cells. NanB sialidase was purified from P. multocida to test its toxicity and its ability to hydrolyse its sialic acid receptors on MDCK cells. The H9N2 challenge virus was propagated in MDCK cells until cytopathic effects appeared. Antiviral activity of NanB sialidase was tested using MDCK cells, and then observed based on cell morphology, viral copy number, and expression of apoptosis-mediating genes. NanB sialidase effectively hydrolysed Neu5Acα(2,6)-Gal sialic acid at a dose of 129 mU/ml, while at 258 mU/ml, it caused toxicity to MDCK cells. Antiviral activity of sialidase was evident based on the significant decrease in viral copy number at all doses administered. The increase of p53 and caspase-3 expression was observed in infected cells without sialidase. Our study demonstrates the ability of NanB sialidase to inhibit H9N2 virus replication based on observations of sialic acid hydrolysis, reduction in viral copy number, and expression of apoptosis-related genes. The future application of sialidase may be considered as an antiviral strategy against avian influenza H9N2 virus infections. RESEARCH HIGHLIGHTSNanB sialidase effectively hydrolyses Neu5Acα(2,6)-Gal at a dose of 129 mU/ml.NanB sialidase from Pasteurella multocida can inhibit the entry of H9N2 virus into cells.NanB sialidase of Pasteurella multocida prevents infection-induced cell apoptosis.NanB sialidase reduces the H9N2 viral copy number in MDCK cells.
Collapse
Affiliation(s)
- Christian Marco Hadi Nugroho
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, University of Indonesia, DKI Jakarta, Indonesia
- Animal Health Research and Diagnostic Unit, PT Medika Satwa Laboratoris, Bogor, Indonesia
| | - Otto Sahat Martua Silaen
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, University of Indonesia, DKI Jakarta, Indonesia
| | - Ryan Septa Kurnia
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, University of Indonesia, DKI Jakarta, Indonesia
- Animal Health Research and Diagnostic Unit, PT Medika Satwa Laboratoris, Bogor, Indonesia
| | | | - Muhammad Ade Putra
- Department of Animal Disease and Veterinary Health, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor, Indonesia
| | - Agustin Indrawati
- Department of Animal Disease and Veterinary Health, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor, Indonesia
| | - Okti Nadia Poetri
- Department of Animal Disease and Veterinary Health, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor, Indonesia
| | - I Wayan Teguh Wibawan
- Department of Animal Disease and Veterinary Health, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor, Indonesia
| | - Silvia Tri Widyaningtyas
- Virology and Cancer Pathobiology Research Centre, Faculty of Medicine, University of Indonesia, DKI Jakarta, Indonesia
| | - Amin Soebandrio
- Department of Microbiology, Faculty of Medicine, University of Indonesia, DKI Jakarta, Indonesia
| |
Collapse
|
2
|
Al-Shuhaib MBS, Alam S, Khan SA, Al-Shuhaib JMB, Chen YK, M Alshabrmi F. Hemagglutinin 3 and 8 can be the most efficient influenza subtypes for human host invasion; a comparative in silico approach. J Biomol Struct Dyn 2025; 43:108-126. [PMID: 37965722 DOI: 10.1080/07391102.2023.2280674] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/28/2023] [Indexed: 11/16/2023]
Abstract
The severity of the influenza virus infection is largely determined by its ability to invade the human host receptor. This critical step is conducted by utilizing hemagglutinin (HA) due to its binding with sialic acid 2,6 (SA). Though 18 subtypes (H1-H18) of HA have been identified, the most efficient one for conducting the host entry has not yet been resolved. This study aims to assess the severity of infections for HA variants by conducting a comparative docking of H1-H18 with the human SA receptor. Eighteen viral 3D structures were retrieved, minimized, and optimized for docking with human SA. In all retrieved structures, five conserved amino acid residues were selected for docking with human SA. Special protein grids were prepared by locating these five residues in the 18 selected subtypes. Results showed that H3 and H8 exerted the highest standard precision and extra precision docking scores, and the highest binding affinities with the human SA, respectively. Phylogenetic analyses confirmed the actual positioning of the selected 3D structures and showed these docked structures belonged to their usual classes due to the extremely close distances found in each docked subtype compared with its corresponding non-docked structures. H8-SA showed slightly better RMSD and SASA values than H3-SA, while H3-SIA showed more favourable radius of gyration scores than H8-SIA in the majority of the simulation period. Due to the highest affinity of binding of H3 and H8 with the human receptor, special caution should be exercised regarding any possible outbreak mediated by these subtypes in human populations. However, it is important to acknowledge a limitation inherent to the computational approach; it may hold relative rather than absolute significance. Further research is needed to deepen our understanding of the intricate interplay between HA variants and the host receptor, taking into account the broader context of viral infection dynamics.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Sarfaraz Alam
- Tunneling Group Biotechnology Centre, Gliwice, Poland
| | | | | | - Yan-Kun Chen
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Fahad M Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
3
|
Sriwilaijaroen N, Hanamatsu H, Yokota I, Nishikaze T, Ijichi T, Takahashi T, Sakoda Y, Furukawa JI, Suzuki Y. Edible bird's nest: N- and O-glycan analysis and synergistic anti-avian influenza virus activity with neuraminidase inhibitors. Antiviral Res 2024; 232:106040. [PMID: 39577572 DOI: 10.1016/j.antiviral.2024.106040] [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: 07/08/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Zoonotic avian influenza viruses have continued to infect people on occasion. During treatment, antiviral resistant viruses have occasionally emerged, highlighting the need for a novel strategy for treating human illness. After pancreatin treatment, edible bird's nest (EBN), swiftlet saliva consumed for health purposes, possesses anti-avian viral activity by inhibiting receptor-binding hemagglutinin (HA) activity. Glycan analysis revealed an abundance of α2,3Neu5Ac decoy receptors in pancreatin-treated EBN. Fucosylated tri-α2,3Neu5Ac tri-antennary N-glycans (N-35) and di-α2,3Neu5Ac core 2 O-glycans (O-15) are predominant, accounting for 53.46% and 44.66% of total N- and O-glycan amounts, respectively. Isobologram analysis revealed that the treated EBN had a strong synergistic effect with either oseltamivir carboxylate or zanamivir, a competitive inhibitor of receptor-destroying neuraminidases (NAs), against the avian H5N1 virus. Taken together, EBN has the potential to be developed as a food-derived avian viral trap to prevent and decrease avian virus infection as well as in combination with a viral releasing-NA inhibitor to increase therapeutic potency, reduce toxicity, delay resistance development, and potentially prevent pandemic onset.
Collapse
Affiliation(s)
- Nongluk Sriwilaijaroen
- Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand; Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka-shi, Shizuoka, 422-8526, Japan.
| | - Hisatoshi Hanamatsu
- Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Ikuko Yokota
- Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Takashi Nishikaze
- Global Application Development Center, Shimadzu Corporation, Kyoto, Kyoto, 604-8511, Japan
| | - Tetsuo Ijichi
- Life Science Division, Combi Corporation, Saitama-shi, Saitama, 338-0832, Japan
| | - Tadanobu Takahashi
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka-shi, Shizuoka, 422-8526, Japan
| | - Yoshihiro Sakoda
- Laboratory of Microbiology, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| | - Jun-Ichi Furukawa
- Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Aichi, 464-8601, Japan; Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, 060-8638, Japan
| | - Yasuo Suzuki
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka-shi, Shizuoka, 422-8526, Japan
| |
Collapse
|
4
|
Storms SM, Leonardi-Cattolica A, Prezioso T, Varga C, Wang L, Lowe J. Influenza A virus shedding and reinfection during the post-weaning period in swine: longitudinal study of two nurseries. Front Vet Sci 2024; 11:1482225. [PMID: 39606665 PMCID: PMC11601151 DOI: 10.3389/fvets.2024.1482225] [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: 08/17/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
Introduction Influenza A virus in swine (IAV-S) is common in the United States commercial swine population and has the potential for zoonotic transmission. Objective To elucidate influenza shedding the domestic pig population, we evaluated two commercial swine farms in Illinois, United States, for 7 weeks. Farm 1 had a recent IAV-S outbreak. Farm 2 has had IAV-S circulating for several years. Methods Forty post-weaning pigs on Farm 1 and 51 pigs from Farm 2 were individually monitored and sampled by nasal swabs for 7 weeks. Results RT-PCR results over time showed most piglets shed in the first 2 weeks post weaning, with 91.2% shedding in week one, and 36.3% in week two. No difference in the number of pigs shedding was found between the two nurseries. Reinfection events did differ between the farms, with 30% of piglets on Farm 1 becoming reinfected, compared to 7.8% on Farm 2. In addition, whole genome sequencing of nasal swab samples from each farm showed identical viruses circulating between the initial infection and the reinfection periods. Sequencing also allowed for nucleic and amino acid mutation analysis in the circulating viruses, as well the identification of a potential reverse zoonosis event. We saw antigenic site mutations arising in some pigs and MxA resistance genes in almost all samples. Conclusion This study provided information on IAV-S circulation in nurseries to aid producers and veterinarians to screen appropriately for IAV-S, determine the duration of IAV-S shedding, and predict the occurrence of reinfection in the nursery period.
Collapse
Affiliation(s)
- Suzanna M. Storms
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | | | - Tara Prezioso
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Csaba Varga
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Leyi Wang
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - James Lowe
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| |
Collapse
|
5
|
Raza MA, Ashraf MA. Drug resistance and possible therapeutic options against influenza A virus infection over past years. Arch Microbiol 2024; 206:458. [PMID: 39499323 DOI: 10.1007/s00203-024-04181-3] [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: 09/09/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/07/2024]
Abstract
Influenza A virus infection, commonly known as the flu, has persisted in the community for centuries. Although we have yearly vaccinations to prevent seasonal flu, there remains a dire need for antiviral drugs to treat active infections. The constantly evolving genome of the influenza A virus limits the number of effective antiviral therapeutic options. Over time, antiviral drugs become inefficient due to the development of resistance, as seen with adamantanes, which are now largely ineffective against most circulating strains of the virus. Neuraminidase inhibitors have long been the drug of choice, but due to selection pressure, strains are becoming resistant to this class of drugs. Baloxavir marboxil, a drug with a novel mode of action, can be used against strains resistant to other classes of drugs but is still not available in many countries. Deep research into nanoparticles has shown they are effective as antiviral drugs, opening a new avenue of research to use them as antiviral agents with novel modes of action. As this deadly virus, which has killed millions of people in the past, continues to develop resistance, there is an urgent need for new therapeutic agents with novel modes of action to halt active infections in patients. This review article covers the available therapeutic antiviral drug options with different modes of action, their effectiveness, and resistance to various strains of influenza A virus.
Collapse
Affiliation(s)
- Muhammad Asif Raza
- CAS Key Laboratory of Molecular Virology and Immunology, Institutional Center for Shared Technologies and Facilities, Pathogen Discovery and Big Data Platform, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, No. 320 Yueyang Road, Shanghai, 200031, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Muhammad Awais Ashraf
- CAS Key Laboratory of Molecular Virology and Immunology, Institutional Center for Shared Technologies and Facilities, Pathogen Discovery and Big Data Platform, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, No. 320 Yueyang Road, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
6
|
Dorji K, Klungthong C, Dorji T, Wangchuk T, Yuden P, Pelki T, Ghishing TD, Gyemiry G, Gyeltshen S, Chinnawirotpisan P, Manasatienkij W, Wangchuk S, Farmer A. Epidemiology and genetic characterization of influenza viruses circulating in Bhutan in 2022. PLoS One 2024; 19:e0304849. [PMID: 39288111 PMCID: PMC11407632 DOI: 10.1371/journal.pone.0304849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/21/2024] [Indexed: 09/19/2024] Open
Abstract
INTRODUCTION Influenza (Flu) causes considerable morbidity and mortality globally, and in Bhutan, Flu viruses are a leading cause of acute respiratory infection and cause outbreaks during Flu seasons. In this study, we aim to analyze the epidemiology and the genetic characterization of Flu viruses circulated in Bhutan in 2022. METHOD Respiratory specimens were collected from patients who meet the case definition for influenza-like illness (ILI) and severe acute respiratory infection (SARI) from sentinel sites. Specimens were tested for Flu and SARS-CoV-2 viruses by RT-PCR using the Multiplex Assay. Selected positive specimens were utilized for Flu viral genome sequencing by next-generation sequencing. Descriptive analysis was performed on patient demographics to see the proportion of Flu-associated ILI and SARI. All data were analyzed using Epi Info7 and QGIS 3.16 software. RESULT A weekly average of 16.2 ILI cases per 1000 outpatient visits and 18 SARI cases per 1000 admitted cases were reported in 2022. The median age among ILI was 12 years (IQR: 5-28) and SARI was 6.2 (IQR: 2.5-15) years. Flu A(H3N2) (70.2%) subtype was the most predominant circulating strain. Flu A(H1N1)pdm09 and Flu B viruses belonged to subclades that were mismatched to the vaccine strains recommended for the 2021-2022 season but matched the vaccine strain for the 2022-2023 season with vaccine efficacy 85.14% and 88.07% respectively. Flu A(H3N2) virus belonged to two subclades which differed from the vaccine strains recommended in both the 2021-2022 and 2022-2023 seasons with vaccine efficacy 68.28%. CONCLUSION Flu virus positivity rates were substantially elevated during the Flu season in 2022 compared to 2021. Flu A(H3N2) subtype was the most predominant circulating strain in the country and globally. Genetic characterization of the Flu viruses in Bhutan showed a close relatedness of high vaccine efficacy with the vaccine strain that WHO recommended for the 2022-23 season.
Collapse
Affiliation(s)
- Kunzang Dorji
- National Influenza Centre, Royal Centre for Disease Control, Ministry of Health, Thimphu, Bhutan
| | - Chonticha Klungthong
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Tshering Dorji
- National Influenza Centre, Royal Centre for Disease Control, Ministry of Health, Thimphu, Bhutan
| | - Tandin Wangchuk
- National Influenza Centre, Royal Centre for Disease Control, Ministry of Health, Thimphu, Bhutan
| | - Pema Yuden
- National Influenza Centre, Royal Centre for Disease Control, Ministry of Health, Thimphu, Bhutan
| | - Tshering Pelki
- National Influenza Centre, Royal Centre for Disease Control, Ministry of Health, Thimphu, Bhutan
| | - Tara Devi Ghishing
- National Influenza Centre, Royal Centre for Disease Control, Ministry of Health, Thimphu, Bhutan
| | - Govinda Gyemiry
- ICT, Royal Centre for Disease Control, Ministry of Health, Thimphu, Bhutan
| | - Sonam Gyeltshen
- National Influenza Centre, Royal Centre for Disease Control, Ministry of Health, Thimphu, Bhutan
| | | | - Wudtichai Manasatienkij
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Sonam Wangchuk
- Royal Centre for Disease Control, Ministry of Health, Thimphu, Bhutan
| | - Aaron Farmer
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| |
Collapse
|
7
|
Putra MA, Soebandrio A, Wibawan IWT, Nugroho CMHN, Kurnia RS, Silaen OSM, Rizkiantino R, Indrawati A, Poetri ON, Krisnamurti DGB. Analyzing Molecular Traits of H9N2 Avian Influenza Virus Isolated from a Same Poultry Farm in West Java Province, Indonesia, in 2017 and 2023. F1000Res 2024; 13:571. [PMID: 39610402 PMCID: PMC11602698 DOI: 10.12688/f1000research.150975.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/11/2024] [Indexed: 11/30/2024] Open
Abstract
Background Indonesia is one of the countries that is endemic to avian influenza virus subtype H9N2. This study aims to compare the molecular characteristics of avian influenza virus (AIV) subtype H9N2 from West Java. Methods Specific pathogen-free (SPF) embryonated chicken eggs were used to inoculate samples. RNA extraction and RT-qPCR confirmed the presence of H9 and N2 genes in the samples. RT-PCR was employed to amplify the H9N2-positive sample. Nucleotide sequences were obtained through Sanger sequencing and analyzed using MEGA 7. Homology comparison and phylogenetic tree analysis, utilizing the neighbor-joining tree method, assessed the recent isolate's similarity to reference isolates from GenBank. Molecular docking analysis was performed on the HA1 protein of the recent isolate and the A/Layer/Indonesia/WestJava-04/2017 isolate, comparing their interactions with the sialic acids Neu5Ac2-3Gal and Neu5Ac2-6Gal. Results RT-qPCR confirmed the isolate samples as AIV subtype H9N2. The recent virus exhibited 11 amino acid residue differences compared to the A/Layer/Indonesia/WestJava-04/2017 isolate. Phylogenetically, the recent virus remains within the h9.4.2.5 subclade. Notably, at antigenic site II, the recent isolate featured an amino acid N at position 183, unlike A/Layer/Indonesia/WestJava-04/2017. Molecular docking analysis revealed a preference of HA1 from the 2017 virus for Neu5Ac2-3Gal, while the 2023 virus displayed a tendency to predominantly bind with Neu5Ac2-6Gal. Conclusion In summary, the recent isolate displayed multiple mutations and a strong affinity for Neu5Ac2-6Gal, commonly found in mammals.
Collapse
Affiliation(s)
- Muhammad Ade Putra
- Master of Animal Biomedical Sciences, School of Veterinary and Biomedical, IPB University, Bogor, West Java, 16680, Indonesia
| | - Amin Soebandrio
- Department of Microbiology, Faculty of Medicine, University of Indonesia, Jakarta, Jakarta, 10320, Indonesia
| | - I Wayan Teguh Wibawan
- Division of Medical Microbiology, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor, West Java, 16680, Indonesia
| | | | - Ryan Septa Kurnia
- Animal Health Diagnostic Unit, PT. Medika Satwa Laboratoris, Bogor, West Java, 16166, Indonesia
| | | | - Rifky Rizkiantino
- Division of Central Laboratory and Disease Research Center, Technology and Research Development, Central Proteina Prima (CP Prima) Inc., Tangerang, Banten, 15560, Indonesia
| | - Agustin Indrawati
- Division of Medical Microbiology, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor, West Java, 16680, Indonesia
| | - Okti Nadia Poetri
- Division of Medical Microbiology, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor, West Java, 16680, Indonesia
| | - Desak Gede Budi Krisnamurti
- Department of Medical Pharmacy, Faculty of Medicine, University of Indonesia, Jakarta, Jakarta, 10430, Indonesia
| |
Collapse
|
8
|
Neog S, Vinjamuri SR, Vijayan K, Kumar S, Trivedi V. NDV targets the invasion pathway in malaria parasite through cell surface sialic acid interaction. FASEB J 2024; 38:e23856. [PMID: 39092913 DOI: 10.1096/fj.202400004rr] [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: 01/03/2024] [Revised: 07/01/2024] [Accepted: 07/21/2024] [Indexed: 08/04/2024]
Abstract
Merozoites utilize sialic acids on the red blood cell (RBC) cell surface to rapidly adhere to and invade the RBCs. Newcastle disease virus (NDV) displays a strong affinity toward membrane-bound sialic acids. Incubation of NDV with the malaria parasites dose-dependently reduces its cellular viability. The antiplasmodial activity of NDV is specific, as incubation with Japanese encephalitis virus, duck enteritis virus, infectious bronchitis virus, and influenza virus did not affect the parasite propagation. Interestingly, NDV is reducing more than 80% invasion when RBCs are pretreated with the virus. Removal of the RBC surface proteins or the NDV coat proteins results in disruption of the virus binding to RBC. It suggests the involvement of specific protein: ligand interaction in virus binding. We established that the virus engages with the parasitized RBCs (PRBCs) through its hemagglutinin neuraminidase (HN) protein by recognizing sialic acid-containing glycoproteins on the cell surface. Blocking of the HN protein with free sialic acid or anti-HN antibodies abolished the virus binding as well as its ability to reduce parasite growth. Interestingly, the purified HN from the virus alone could inhibit the parasite's growth in a dose-dependent manner. NDV binds strongly to knobless murine parasite strain Plasmodium yoelii and restricted the parasite growth in mice. Furthermore, the virus was found to preferentially target the PRBCs compared to normal erythrocytes. Immunolocalization studies reveal that NDV is localized on the plasma membrane as well as weakly inside the PRBC. NDV causes neither any infection nor aggregation of the human RBCs. Our findings suggest that NDV is a potential candidate for developing targeted drug delivery platforms for the Plasmodium-infected RBCs.
Collapse
Affiliation(s)
- Siddharth Neog
- Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati, India
| | - Sandeep Reddy Vinjamuri
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Kamalakannan Vijayan
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Sachin Kumar
- Viral Immunology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati, India
| | - Vishal Trivedi
- Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati, India
| |
Collapse
|
9
|
Korsun N, Trifonova I, Madzharova I, Christova I. Resurgence of influenza with increased genetic diversity of circulating viruses during the 2022-2023 season. J Med Microbiol 2024; 73. [PMID: 39073070 DOI: 10.1099/jmm.0.001864] [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] [Indexed: 07/30/2024] Open
Abstract
Introduction. After two seasons of absence and low circulation, influenza activity increased significantly in the winter of 2022-2023. This study aims to characterize virological and epidemiological aspects of influenza infection in Bulgaria during the 2022-2023 season and perform a phylogenetic/molecular analysis of the hemagglutinin (HA) and neuraminidase (NA) sequences of representative influenza strains.Hypothesis/Gap Statement. Influenza A and B viruses generate new genetic groups/clades each season, replacing previously circulating variants. This results in increased antigenic distances from current vaccine strains. Strengthening existing influenza surveillance is essential to meet the challenges posed by the co-circulation of influenza and SARS-CoV-2.Methodology. We tested 2713 clinical samples from patients with acute respiratory illnesses using a multiplex real-time RT-PCR kit (FluSC2) to detect influenza A/B and Severe acute respiratory syndrome coronavirus-2(SARS-CoV-2) simultaneously. Representative Bulgarian influenza strains were sequenced at the WHO Collaborating Centres in London, UK, and Atlanta, USA.Results. Influenza virus was detected in 694 (25.6 %) patients. Of these, 364 (52.4 %), 213 (30.7 %) and 117 (16.9 %) were positive for influenza A(H1N1)pdm09, A(H3N2) and B/Victoria lineage virus, respectively. HA genes of the 47 influenza A(H1N1)pdm09 viruses fell into clades 5a.2. and 5a.2a.1 within the 6B.5A.1A.5a.2 group. Twenty-seven A(H3N2) viruses belonging to subclades 2b, 2a.1, 2a.1b and 2a.3a.1 within the 3C.2a1b.2a.2 group were analysed. All 23 sequenced B/Victoria lineage viruses were classified into the V1A.3a.2 group. We identified amino acid substitutions in HA and NA compared with the vaccine strains, including several substitutions in the HA antigenic sites.Conclusion. The study's findings showed genetic diversity among the influenza A viruses and, to a lesser extent, among B viruses, circulating in the first season after the lifting of anti-COVID-19 measures.
Collapse
MESH Headings
- Humans
- Influenza, Human/virology
- Influenza, Human/epidemiology
- Genetic Variation
- Phylogeny
- Influenza B virus/genetics
- Influenza B virus/classification
- Influenza B virus/isolation & purification
- SARS-CoV-2/genetics
- SARS-CoV-2/classification
- Neuraminidase/genetics
- Adult
- Male
- Middle Aged
- Female
- Bulgaria/epidemiology
- Young Adult
- Aged
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Child, Preschool
- Child
- Adolescent
- COVID-19/epidemiology
- COVID-19/virology
- Infant
- Seasons
- Influenza A virus/genetics
- Influenza A virus/classification
- Influenza A virus/isolation & purification
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/classification
- Influenza A Virus, H1N1 Subtype/isolation & purification
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/classification
- Influenza A Virus, H3N2 Subtype/isolation & purification
Collapse
Affiliation(s)
- Neli Korsun
- National Laboratory "Influenza and ARI", Department of Virology, National Center of Infectious and Parasitic Diseases, 44A Stoletov Blvd, 1233 Sofia, Bulgaria
| | - Ivelina Trifonova
- National Laboratory "Influenza and ARI", Department of Virology, National Center of Infectious and Parasitic Diseases, 44A Stoletov Blvd, 1233 Sofia, Bulgaria
| | - Iveta Madzharova
- National Laboratory "Influenza and ARI", Department of Virology, National Center of Infectious and Parasitic Diseases, 44A Stoletov Blvd, 1233 Sofia, Bulgaria
| | - Iva Christova
- National Laboratory "Influenza and ARI", Department of Virology, National Center of Infectious and Parasitic Diseases, 44A Stoletov Blvd, 1233 Sofia, Bulgaria
| |
Collapse
|
10
|
Pérez-Isidoro R, Valdez-Lara AG, Díaz-Salazar AJ, Hoeppener S, Guerrero-Sánchez C, Quintana-Owen P, Ruiz-Suárez JC, Schubert US, Ayora-Talavera G, De Jesús-Téllez MA, Saldívar-Guerra E. Biophysical investigation of liposome systems decorated with bioconjugated copolymers in the presence of amantadine. J Mater Chem B 2024; 12:5823-5837. [PMID: 38757473 DOI: 10.1039/d4tb00171k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Liposome-based technologies derived from lipids and polymers (e.g., PEGylated liposomes) have been recognized because of their applications in nanomedicine. However, since such systems represent myriad challenges and may promote immune responses, investigation of new biomaterials is mandatory. Here, we report on a biophysical investigation of liposomes decorated with bioconjugated copolymers in the presence (or absence) of amantadine (an antiviral medication). First, copolymers of poly(N,N-dimethylacrylamide-co-fluoresceinacrylate-co-acrylic acid-N-succinimide ester)-block-poly(N-isopropylacrylamide) (PDMA-b-PNIPAM) containing a fluorescence label were biofunctionalized with short peptides that resemble the sequence of the loops 220 and 130 of the binding receptor of the hemagglutinin (HA) protein of the influenza A virus. Then, the bioconjugated copolymers were self-assembled along with liposomes composed of 1,2 dimyristoyl-sn-glycero-3-phosphocholine, sphingomyelin, and cholesterol (MSC). These biohybrid systems, with and without amantadine, were systematically characterized using differential scanning calorimetry (DSC), dynamic light scattering (DLS), and cryogenic transmission electron microscopy (cryoTEM). Finally, the systems were tested in an in vitro study to evaluate cytotoxicity and direct immunofluorescence in Madin Darbin Canine Kidney (MDCK) cells. The biohybrid systems displayed long-term stability, thermo-responsiveness, hydrophilic-hydrophobic features, and fluorescence properties and were presumable endowed with cell targeting properties intrinsically integrated into the amino acid sequences of the utilized peptides, which indeed turn them into promising nanodevices for biomedical applications.
Collapse
Affiliation(s)
- Rosendo Pérez-Isidoro
- Centro de Investigación en Química Aplicada (CIQA), Enrique Reyna, 140, 25294 Saltillo, Coahuila, Mexico.
| | | | - Alma Jessica Díaz-Salazar
- Laboratorio de Bio-fisicoquímica, Departamento de Fisicoquímica, Facultad de Química, Universidad Nacional Autónoma de México, 04510 México City, Mexico
| | - Stephanie Hoeppener
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 97743 Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Carlos Guerrero-Sánchez
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 97743 Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Patricia Quintana-Owen
- Departamento de Física Aplicada, CINVESTAV-IPN, Unidad Mérida, A.P. 73, Cordemex, 97310 Mérida, Yucatán, Mexico
| | | | - Ulrich S Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 97743 Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Guadalupe Ayora-Talavera
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 97743 Jena, Germany.
| | | | - Enrique Saldívar-Guerra
- Centro de Investigación en Química Aplicada (CIQA), Enrique Reyna, 140, 25294 Saltillo, Coahuila, Mexico.
| |
Collapse
|
11
|
Kim YH, Park C, Nguyen HD, V Ngo H, Lee BJ. Self-assembled nanonization of fatty acid-conjugated vaccine antigen for enhanced thermal stability. Int J Pharm 2024; 658:124176. [PMID: 38688427 DOI: 10.1016/j.ijpharm.2024.124176] [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: 12/20/2023] [Revised: 04/02/2024] [Accepted: 04/28/2024] [Indexed: 05/02/2024]
Abstract
The aim of this study was to evaluate the enhanced thermal stability and physicochemical properties of fattigated vaccine antigens. High molecular weight influenza hemagglutinin (Heg) was used as a model antigen because of low heat stability requiring cold chamber. Heg was conjugated with long-chain oleic acid (C18) and short-chain 3-decenoic acid (C10) to prepare fattigated Heg. Circular dichroism analysis revealed no significant changes in the three-dimensional structure post-conjugation. In the liquid state, the fattigated Heg was self-assembled into nanoparticles (NPs) due to its amphiphilic nature, with sizes of 136.27 ± 12.78 nm for oleic acid-conjugated Heg (HOC) and 88.73 ± 3.27 nm for 3-decenoic acid-conjugated Heg (HDC). Accelerated thermal stability studies at 60 °C for 7 days demonstrated that fattigated Heg exhibited higher thermal stability than Heg in various liquid or solid states. The longer-chained HOC showed better thermal stability than HDC in the liquid state, attributed to increased hydrophobic interactions during self-assembly. In bio-mimicking liquid states at 37 °C, HOC exhibited higher thermal stability than Heg. Furthermore, solid-state HOC with cryoprotectants (trehalose, mannitol, and Tween® 80) had significantly increased thermal stability due to reduced exposure of protein surface area via nanonization behavior. The current fattigation platform could be a promising strategy for developing thermostable nano vaccines of heat-labile vaccine antigens.
Collapse
Affiliation(s)
- Yeon-Ho Kim
- Department of Pharmacy, College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Chulhun Park
- College of Pharmacy, Jeju National University, Jeju 63243, Republic of Korea
| | - Hy D Nguyen
- Department of Pharmacy, College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Hai V Ngo
- Department of Pharmacy, College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Beom-Jin Lee
- Department of Pharmacy, College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea; Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
12
|
Hoang PT, Luong QXT, Ayun RQ, Lee Y, Oh KJ, Kim T, Lee TK, Lee S. A synergistic therapy against influenza virus A/H1N1/PR8 by a HA1 specific neutralizing single-domain V L and an RNA hydrolyzing scFv. Front Microbiol 2024; 15:1355599. [PMID: 38706966 PMCID: PMC11066198 DOI: 10.3389/fmicb.2024.1355599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/01/2024] [Indexed: 05/07/2024] Open
Abstract
The emergence of anti-influenza drug-resistant strains poses a challenge for influenza therapy due to mutations in the virus's surface protein. Recently, there has been increasing interest in combination therapy consisting of two or more drugs as a potential alternative approach, aiming to enhance therapeutic efficacy. In this study, we investigated a novel synergistic therapy with a vertical effect using a single-domain VL-HA1-specific antibody against H1N1/PR8 and a horizontal effect using an RNA catalytic antibody with broad-spectrum influenza antiviral drug. We isolated a single-domain VL-HA1-specific (NVLH8) antibody binding to the virus particles showing a neutralizing activity against influenza virus A, specifically H1N1/PR8, as determined by the reduction in plaque number and lower viral HA protein expression in vitro. The neutralizing antibody likely prevented the viral entry, specifically at the viral genome-releasing step. Additionally, the 3D8 scFv hydrolyzed viral RNAs in the cytoplasm, including mRNA, vRNA, and cRNA in MDCK cells. The combined treatment of neutralizing antibodies for a vertical effect and 3D8 scFv for a horizontal effect produced a synergistic effect providing a novel approach against viral diseases when compared with a single treatment. Our results indicated that combining treatment, in particular two proteins exhibiting different mechanisms of action increased the antiviral activity against the influenza virus.
Collapse
Affiliation(s)
- Phuong Thi Hoang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
- Novelgen Co., Ltd., R&D Center, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Quynh Xuan Thi Luong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ramadhani Qurrota Ayun
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yongjun Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kwang-Ji Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
- Novelgen Co., Ltd., R&D Center, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Taehyun Kim
- Novelgen Co., Ltd., R&D Center, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Taek-Kyun Lee
- Risk Assessment Research Center, Korea Institute of Ocean Science and Technology, Geoje, Republic of Korea
| | - Sukchan Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
13
|
Yu L, Liu X, Wei X, Ren J, Wang X, Wu S, Lan K. C1QTNF5 is a novel attachment factor that facilitates the entry of influenza A virus. Virol Sin 2024; 39:277-289. [PMID: 38246238 PMCID: PMC11074642 DOI: 10.1016/j.virs.2024.01.003] [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: 10/17/2023] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
Influenza A virus (IAV) binds sialic acid receptors on the cell surface to enter the host cells, which is the key step in initiating infection, transmission and pathogenesis. Understanding the factors that contribute to the highly efficient entry of IAV into human cells will help elucidate the mechanism of viral entry and pathogenicity, and provide new targets for intervention. In the present study, we reported a novel membrane protein, C1QTNF5, which binds to the hemagglutinin protein of IAV and promotes IAV infection in vitro and in vivo. We found that the HA1 region of IAV hemagglutinin is critical for the interaction with C1QTNF5 protein, and C1QTNF5 interacts with hemagglutinin mainly through its N-terminus (1-103 aa). In addition, we further demonstrated that overexpression of C1QTNF5 promotes IAV entry, while blocking the interaction between C1QTNF5 and IAV hemagglutinin greatly inhibits viral entry. However, C1QTNF5 does not function as a receptor to mediate IAV infection in sialic acid-deficient CHO-Lec2 cells, but promotes IAV to attach to these cells, suggesting that C1QTNF5 is an important attachment factor for IAV. This work reveals C1QTNF5 as a novel IAV attachment factor and provides a new perspective for antiviral strategies.
Collapse
Affiliation(s)
- Lei Yu
- State Key Laboratory of Virology, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Xinjin Liu
- State Key Laboratory of Virology, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Xiaoqin Wei
- State Key Laboratory of Virology, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Junrui Ren
- State Key Laboratory of Virology, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Xueyun Wang
- State Key Laboratory of Virology, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Shuwen Wu
- State Key Laboratory of Virology, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Ke Lan
- State Key Laboratory of Virology, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430072, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430072, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
14
|
Heider A, Wedde M, Weinheimer V, Döllinger S, Monazahian M, Dürrwald R, Wolff T, Schweiger B. Characteristics of two zoonotic swine influenza A(H1N1) viruses isolated in Germany from diseased patients. Int J Med Microbiol 2024; 314:151609. [PMID: 38286065 DOI: 10.1016/j.ijmm.2024.151609] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024] Open
Abstract
Interspecies transmission of influenza A viruses (IAV) from pigs to humans is a concerning event as porcine IAV represent a reservoir of potentially pandemic IAV. We conducted a comprehensive analysis of two porcine A(H1N1)v viruses isolated from human cases by evaluating their genetic, antigenic and virological characteristics. The HA genes of those human isolates belonged to clades 1C.2.1 and 1C.2.2, respectively, of the A(H1N1) Eurasian avian-like swine influenza lineage. Antigenic profiling revealed substantial cross-reactivity between the two zoonotic H1N1 viruses and human A(H1N1)pdm09 virus and some swine viruses, but did not reveal cross-reactivity to H1N2 and earlier human seasonal A(H1N1) viruses. The solid-phase direct receptor binding assay analysis of both A(H1N1)v showed a predominant binding to α2-6-sialylated glycans similar to human-adapted IAV. Investigation of the replicative potential revealed that both A(H1N1)v viruses grow in human bronchial epithelial cells to similar high titers as the human A(H1N1)pdm09 virus. Cytokine induction was studied in human alveolar epithelial cells A549 and showed that both swine viruses isolated from human cases induced higher amounts of type I and type III IFN, as well as IL6 compared to a seasonal A(H1N1) or a A(H1N1)pdm09 virus. In summary, we demonstrate a remarkable adaptation of both zoonotic viruses to propagate in human cells. Our data emphasize the needs for continuous monitoring of people and regions at increased risk of such trans-species transmissions, as well as systematic studies to quantify the frequency of these events and to identify viral molecular determinants enhancing the zoonotic potential of porcine IAV.
Collapse
Affiliation(s)
- Alla Heider
- Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Robert Koch-Institute, Seestrasse 10, Berlin 13353, Germany.
| | - Marianne Wedde
- Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Robert Koch-Institute, Seestrasse 10, Berlin 13353, Germany
| | - Viola Weinheimer
- Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Robert Koch-Institute, Seestrasse 10, Berlin 13353, Germany
| | - Stephanie Döllinger
- Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Robert Koch-Institute, Seestrasse 10, Berlin 13353, Germany
| | | | - Ralf Dürrwald
- Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Robert Koch-Institute, Seestrasse 10, Berlin 13353, Germany
| | - Thorsten Wolff
- Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Robert Koch-Institute, Seestrasse 10, Berlin 13353, Germany
| | - Brunhilde Schweiger
- Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Robert Koch-Institute, Seestrasse 10, Berlin 13353, Germany
| |
Collapse
|
15
|
Pushan SS, Samantaray M, Rajagopalan M, Ramaswamy A. Structural dynamics of influenza A (H1N1) hemagglutinin protein: a comparative study of Indian (2018) isolate with its evolutionary neighbor, Californian (2009) strain. J Biomol Struct Dyn 2024:1-14. [PMID: 38379377 DOI: 10.1080/07391102.2024.2317985] [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: 11/09/2023] [Accepted: 02/07/2024] [Indexed: 02/22/2024]
Abstract
This work highlights the structure and dynamics of two trimeric HA proteins of the H1N1 virus from different origins, the pandemic Californian (HACal) and its closest Indian neighbor (HAInd), reported in 2009 and 2018, respectively. Because of mutation, HAInd acquires new N-glycosylation and epitope binding sites along with mutations at RBD, which might trigger an altered viral-host interaction mechanism. Molecular dynamics simulations performed on HA trimers for a period of 250 ns reveal the highly dynamic nature of HACal trimers inherited by the flexibility of HA monomers. In the trimer, the dynamics of one monomer are more pronounced compared to others, and the enhanced dynamics of RBD especially gain attention as they plays a key role during fusion. Conversely, the mutant HAInd trimer effectively establishes more H-bond interactions, and accordingly, the trimer undergoes more stabilized dynamics with a relatively lower amplitude of RBD dynamics, as endorsed by the reduced RMSD, Rg, and SASA variations. The cooperative and anti-cooperative motions dissected for the subdomains of both strains also reveal a prominent anticorrelative motion of RBD against other subdomains. In agreement, the free energy landscape of stable HAInd is also characterized by a single lowest wide energy basin instead of the two minimum energy basins of the HACal trimer. In essence, the mutant HAInd acquires a highly stable conformation with novel functional features, which calls for (i) further investigation on the emerging mutation-mediated variation in viral-host binding mechanism and (ii) the need for further design of site-specific potential inhibitors to face future challenges.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shilpa Sri Pushan
- Department of Bioinformatics, Pondicherry University (A Central University), Kalapet, Puducherry, India
| | - Mahesh Samantaray
- Department of Bioinformatics, Pondicherry University (A Central University), Kalapet, Puducherry, India
| | - Muthukumaran Rajagopalan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur
| | - Amutha Ramaswamy
- Department of Bioinformatics, Pondicherry University (A Central University), Kalapet, Puducherry, India
| |
Collapse
|
16
|
Carter T, Iqbal M. The Influenza A Virus Replication Cycle: A Comprehensive Review. Viruses 2024; 16:316. [PMID: 38400091 PMCID: PMC10892522 DOI: 10.3390/v16020316] [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: 01/18/2024] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Influenza A virus (IAV) is the primary causative agent of influenza, colloquially called the flu. Each year, it infects up to a billion people, resulting in hundreds of thousands of human deaths, and causes devastating avian outbreaks with worldwide losses worth billions of dollars. Always present is the possibility that a highly pathogenic novel subtype capable of direct human-to-human transmission will spill over into humans, causing a pandemic as devastating if not more so than the 1918 influenza pandemic. While antiviral drugs for influenza do exist, they target very few aspects of IAV replication and risk becoming obsolete due to antiviral resistance. Antivirals targeting other areas of IAV replication are needed to overcome this resistance and combat the yearly epidemics, which exact a serious toll worldwide. This review aims to summarise the key steps in the IAV replication cycle, along with highlighting areas of research that need more focus.
Collapse
Affiliation(s)
- Toby Carter
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK;
| | | |
Collapse
|
17
|
Al Farroukh M, Kiseleva I, Stepanova E, Bazhenova E, Krutikova E, Tkachev A, Chistyakova A, Rekstin A, Puchkova L, Rudenko L. The Effect of Mice Adaptation Process on the Pathogenicity of Influenza A/South Africa/3626/2013 (H1N1)pdm09 Model Strain. Int J Mol Sci 2023; 24:17386. [PMID: 38139214 PMCID: PMC10743444 DOI: 10.3390/ijms242417386] [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: 11/02/2023] [Revised: 11/24/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
Influenza virus strain A/South Africa/3626/2013 (H1N1)pdm09 (SA-WT) is a non-mouse-adapted model strain that has naturally high pathogenic properties in mice. It has been suggested that the high pathogenicity of this strain for mice could be due to the three strain-specific substitutions in the polymerase complex (Q687R in PB1, N102T in PB2, and E358E/K heterogeneity in PB2). To evaluate the role of these replacements, SA-WT was passaged five times in mouse lungs, and the genome of the mouse-adapted version of the SA-WT strain (SA-M5) was sequenced. SA-M5 lost E358E/K heterogeneity and retained E358, which is the prevalent amino acid at this position among H1N1pdm09 strains. In addition, in the hemagglutinin of SA-M5, two heterogeneous substitutions (G155G/E and S190S/R) were identified. Both viruses, SA-M5 and SA-WT, were compared for their toxicity, ability to replicate, pathogenicity, and immunogenicity in mice. In mice infected with SA-M5 or SA-WT strains, toxicity, virus titer in pulmonary homogenates, and mouse survival did not differ significantly. In contrast, an increase in the immunogenicity of SA-M5 compared to SA-WT was observed. This increase could be due to the substitutions G155G/E and S190S/R in the HA of SA-M5. The prospects for using SA-M5 in studying the immunogenicity mechanisms were also discussed.
Collapse
Affiliation(s)
- Mohammad Al Farroukh
- Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, St. Petersburg 197022, Russia; (E.S.); (E.B.); (E.K.); (A.C.); (A.R.); (L.P.); (L.R.)
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, St. Petersburg 194021, Russia;
| | - Irina Kiseleva
- Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, St. Petersburg 197022, Russia; (E.S.); (E.B.); (E.K.); (A.C.); (A.R.); (L.P.); (L.R.)
| | - Ekaterina Stepanova
- Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, St. Petersburg 197022, Russia; (E.S.); (E.B.); (E.K.); (A.C.); (A.R.); (L.P.); (L.R.)
| | - Ekaterina Bazhenova
- Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, St. Petersburg 197022, Russia; (E.S.); (E.B.); (E.K.); (A.C.); (A.R.); (L.P.); (L.R.)
| | - Elena Krutikova
- Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, St. Petersburg 197022, Russia; (E.S.); (E.B.); (E.K.); (A.C.); (A.R.); (L.P.); (L.R.)
| | - Artem Tkachev
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, St. Petersburg 194021, Russia;
| | - Anna Chistyakova
- Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, St. Petersburg 197022, Russia; (E.S.); (E.B.); (E.K.); (A.C.); (A.R.); (L.P.); (L.R.)
| | - Andrey Rekstin
- Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, St. Petersburg 197022, Russia; (E.S.); (E.B.); (E.K.); (A.C.); (A.R.); (L.P.); (L.R.)
| | - Ludmila Puchkova
- Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, St. Petersburg 197022, Russia; (E.S.); (E.B.); (E.K.); (A.C.); (A.R.); (L.P.); (L.R.)
| | - Larisa Rudenko
- Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, St. Petersburg 197022, Russia; (E.S.); (E.B.); (E.K.); (A.C.); (A.R.); (L.P.); (L.R.)
| |
Collapse
|
18
|
López-Valiñas Á, Valle M, Pérez M, Darji A, Chiapponi C, Ganges L, Segalés J, Núñez JI. Genetic diversification patterns in swine influenza A virus (H1N2) in vaccinated and nonvaccinated animals. Front Cell Infect Microbiol 2023; 13:1258321. [PMID: 37780850 PMCID: PMC10540852 DOI: 10.3389/fcimb.2023.1258321] [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: 07/13/2023] [Accepted: 08/25/2023] [Indexed: 10/03/2023] Open
Abstract
Influenza A viruses (IAVs) are characterized by having a segmented genome, low proofreading polymerases, and a wide host range. Consequently, IAVs are constantly evolving in nature causing a threat to animal and human health. In 2009 a new human pandemic IAV strain arose in Mexico because of a reassortment between two strains previously circulating in pigs; Eurasian "avian-like" (EA) swine H1N1 and "human-like" H1N2, highlighting the importance of swine as adaptation host of avian to human IAVs. Nowadays, although of limited use, a trivalent vaccine, which include in its formulation H1N1, H3N2, and, H1N2 swine IAV (SIAV) subtypes, is one of the most applied strategies to reduce SIAV circulation in farms. Protection provided by vaccines is not complete, allowing virus circulation, potentially favoring viral evolution. The evolutionary dynamics of SIAV quasispecies were studied in samples collected at different times from 8 vaccinated and 8 nonvaccinated pigs, challenged with H1N2 SIAV. In total, 32 SIAV genomes were sequenced by next-generation sequencing, and subsequent variant-calling genomic analysis was carried out. Herein, a total of 364 de novo single nucleotide variants (SNV) were found along all genetic segments in both experimental groups. The nonsynonymous substitutions proportion found was greater in vaccinated animals suggesting that H1N2 SIAV was under positive selection in this scenario. The impact of each substitution with an allele frequency greater than 5% was hypothesized according to previous literature, particularly in the surface glycoproteins hemagglutinin and neuraminidase. The H1N2 SIAV quasispecies evolution capacity was evidenced, observing different evolutionary trends in vaccinated and nonvaccinated animals.
Collapse
Affiliation(s)
- Álvaro López-Valiñas
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, Barcelona, Spain
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
| | - Marta Valle
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, Barcelona, Spain
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
| | - Marta Pérez
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, Barcelona, Spain
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
| | - Ayub Darji
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, Barcelona, Spain
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
| | - Chiara Chiapponi
- WOAH Reference Laboratory for Swine Influenza, Istituto Zooprofilattico Sperimentale della Lombardia ed Emilia-Romagna, Brescia, Italy
| | - Llilianne Ganges
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, Barcelona, Spain
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
- WOAH Reference Laboratory for Classical Swine Fever, IRTA-CReSA, Barcelona, Spain
| | - Joaquim. Segalés
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - José I. Núñez
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, Barcelona, Spain
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
| |
Collapse
|
19
|
Abd Alhadi M, Friedman LM, Karlsson EA, Cohen-Lavi L, Burkovitz A, Schultz-Cherry S, Noah TL, Weir SS, Shulman LM, Beck MA, Hertz T. Obesity Is Associated with an Impaired Baseline Repertoire of Anti-Influenza Virus Antibodies. Microbiol Spectr 2023; 11:e0001023. [PMID: 37098954 PMCID: PMC10269616 DOI: 10.1128/spectrum.00010-23] [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: 01/02/2023] [Accepted: 03/27/2023] [Indexed: 04/27/2023] Open
Abstract
Obesity is a risk factor for severe disease and mortality for both influenza and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. While previous studies show that individuals with obesity generate antibody responses following influenza vaccination, infection rates within the obese group were twice as high as those in the healthy-weight group. The repertoire of antibodies raised against influenza viruses following previous vaccinations and/or natural exposures is referred to here as baseline immune history (BIH). To investigate the hypothesis that obesity impacts immune memory to infections and vaccines, we profiled the BIH of obese and healthy-weight adults vaccinated with the 2010-2011 seasonal influenza vaccine in response to conformational and linear antigens. Despite the extensive heterogeneity of the BIH profiles in both groups, there were striking differences between obese and healthy subjects, especially with regard to A/H1N1 strains and the 2009 pandemic virus (Cal09). Individuals with obesity had lower IgG and IgA magnitude and breadth for a panel of A/H1N1 whole viruses and hemagglutinin proteins from 1933 to 2009 but increased IgG magnitude and breadth for linear peptides from the Cal09 H1 and N1 proteins. Age was also associated with A/H1N1 BIH, with young individuals with obesity being more likely to have reduced A/H1N1 BIH. We found that individuals with low IgG BIH had significantly lower neutralizing antibody titers than individuals with high IgG BIH. Taken together, our findings suggest that increased susceptibility of obese participants to influenza infection may be mediated in part by obesity-associated differences in the memory B-cell repertoire, which cannot be ameliorated by current seasonal vaccination regimens. Overall, these data have vital implications for the next generation of influenza virus and SARS-CoV-2 vaccines. IMPORTANCE Obesity is associated with increased morbidity and mortality from influenza and SARS-CoV-2 infection. While vaccination is the most effective strategy for preventing influenza virus infection, our previous studies showed that influenza vaccines fail to provide optimal protection in obese individuals despite reaching canonical correlates of protection. Here, we show that obesity may impair immune history in humans and cannot be overcome by seasonal vaccination, especially in younger individuals with decreased lifetime exposure to infections and seasonal vaccines. Low baseline immune history is associated with decreased protective antibody responses. Obesity potentially handicaps overall responses to vaccination, biasing it toward responses to linear epitopes, which may reduce protective capacity. Taken together, our data suggest that young obese individuals are at an increased risk of reduced protection by vaccination, likely due to altered immune history biased toward nonprotective antibody responses. Given the worldwide obesity epidemic coupled with seasonal respiratory virus infections and the inevitable next pandemic, it is imperative that we understand and improve vaccine efficacy in this high-risk population. The design, development, and usage of vaccines for and in obese individuals may need critical evaluation, and immune history should be considered an alternate correlate of protection in future vaccine clinical trials.
Collapse
Affiliation(s)
- Marwa Abd Alhadi
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Lilach M. Friedman
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Erik A. Karlsson
- Virology Unit, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Liel Cohen-Lavi
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Industrial Engineering and Management, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anat Burkovitz
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Terry L. Noah
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Samuel S. Weir
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lester M. Shulman
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Melinda A. Beck
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Tomer Hertz
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
20
|
Zorić JM, Veljović L, Radosavljević V, Glišić D, Kureljušić J, Maletić J, Savić B. Protein sequence features of H1N1 swine influenza A viruses detected on commercial swine farms in Serbia. J Vet Res 2023; 67:147-154. [PMID: 38143831 PMCID: PMC10740377 DOI: 10.2478/jvetres-2023-0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 06/02/2023] [Indexed: 12/26/2023] Open
Abstract
Introduction Swine influenza A viruses (swIAVs) are characterised by high mutation rates and zoonotic and pandemic potential. In order to draw conclusions about virulence in swine and pathogenicity to humans, we examined the existence of molecular markers and accessory proteins, cross-reactivity with vaccine strains, and resistance to antiviral drugs in five strains of H1N1 swIAVs. Material and Methods Amino acid (AA) sequences of five previously genetically characterised swIAVs were analysed in MEGA 7.0 software and the Influenza Research Database. Results Amino acid analysis revealed three virus strains with 590S/591R polymorphism and T271A substitution within basic polymerase 2 (PB2) AA chains, which cause enhanced virus replication in mammalian cells. The other two strains possessed D701N and R251K substitutions within PB2 and synthesised PB1-F2 protein, which are the factors of increased polymerase activity and virulence in swine. All strains synthesised PB1-N40, PA-N155, PA-N182, and PA-X proteins responsible for enhanced replication in mammalian cells and downregulation of the immune response of the host. Mutations detected within haemagglutinin antigenic sites imply the antigenic drift of the five analysed viruses in relation to the vaccine strains. All viruses show susceptibility to neuraminidase inhibitors and baloxavir marboxil, which is important in situations of incidental human infections. Conclusion The detection of virulence markers and accessory proteins in the analysed viruses suggests their higher propensity for replication in mammalian cells, increased virulence, and potential for transmission to humans, and implies compromised efficacy of influenza vaccines.
Collapse
Affiliation(s)
| | - Ljubiša Veljović
- Department of Virology, Institute of Veterinary Medicine of Serbia, 11000Belgrade, Serbia
| | - Vladimir Radosavljević
- Department of Virology, Institute of Veterinary Medicine of Serbia, 11000Belgrade, Serbia
| | - Dimitrije Glišić
- Department of Virology, Institute of Veterinary Medicine of Serbia, 11000Belgrade, Serbia
| | - Jasna Kureljušić
- Department of Food and Feed Safety, Institute of Veterinary Medicine of Serbia, 11000Belgrade, Serbia
| | - Jelena Maletić
- Department of Poultry Diseases, Institute of Veterinary Medicine of Serbia, 11000Belgrade, Serbia
| | - Božidar Savić
- Department of Pathology, Institute of Veterinary Medicine of Serbia, 11000Belgrade, Serbia
| |
Collapse
|
21
|
Ferrando VA, Friedrich ME, Gandhi S, Mellmann A, Masemann D, Christersson A, Anhlan D, Brunotte L, Stoll M, Harder T, Beer M, Boergeling Y, Ludwig S. Cell-intrinsic genomic reassortment of pandemic H1N1 2009 and Eurasian avian-like swine influenza viruses results in potentially zoonotic variants. Emerg Microbes Infect 2023; 12:2212809. [PMID: 37191590 DOI: 10.1080/22221751.2023.2212809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Influenza A viruses (IAV) cause annual epidemics and occasional pandemics in humans. The most recent pandemic outbreak occurred in 2009 with H1N1pdm09. This virus, which most likely reassorted in swine before its transmission to humans, was reintroduced into the swine population and continues circulating ever since. In order to assess its potential to cause reassortants on a cellular level, human origin H1N1pdm09 and a recent Eurasian avian-like H1N1 swine IAV were (co-)passaged in the newly generated swine lung cell line C22. Co-infection with both viruses gave rise to numerous reassortants that additionally carry different mutations which can partially be found in nature as well. Reassortment most frequently affected the PB1, PA and NA segments with the swine IAV as recipient. These reassortants reached higher titers in swine lung cells and were able to replicate in genuine human lung tissue explants ex vivo, suggesting a possible zoonotic potential. Interestingly, reassortment and mutations in the viral ribonucleoprotein complex influence the viral polymerase activity in a cell type and species-specific manner. In summary, we demonstrate reassortment promiscuity of these viruses in a novel swine lung cell model and indicate a possible zoonotic potential of the reassortants.
Collapse
Affiliation(s)
- Verónica A Ferrando
- Institute of Virology Münster, Westfälische Wilhelms-University 48149 Münster, Germany
| | - Marcel E Friedrich
- Institute of Virology Münster, Westfälische Wilhelms-University 48149 Münster, Germany
| | - Shrey Gandhi
- Department of Genetic Epidemiology, Institute of Human Genetics, Westfälische Wilhelms-University 48149 Münster, Germany
| | - Alexander Mellmann
- Institute of Hygiene Münster, Westfälische Wilhelms-University 48149 Münster, Germany
| | - Dörthe Masemann
- Institute of Virology Münster, Westfälische Wilhelms-University 48149 Münster, Germany
| | - Anmari Christersson
- Institute of Virology Münster, Westfälische Wilhelms-University 48149 Münster, Germany
| | - Darisuren Anhlan
- Institute of Virology Münster, Westfälische Wilhelms-University 48149 Münster, Germany
| | - Linda Brunotte
- Institute of Virology Münster, Westfälische Wilhelms-University 48149 Münster, Germany
| | - Monika Stoll
- Department of Genetic Epidemiology, Institute of Human Genetics, Westfälische Wilhelms-University 48149 Münster, Germany
| | - Timm Harder
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute 17493 Greifswald, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute 17493 Greifswald, Germany
| | - Yvonne Boergeling
- Institute of Virology Münster, Westfälische Wilhelms-University 48149 Münster, Germany
| | - Stephan Ludwig
- Institute of Virology Münster, Westfälische Wilhelms-University 48149 Münster, Germany
| |
Collapse
|
22
|
Aganovic A. pH-dependent endocytosis mechanisms for influenza A and SARS-coronavirus. Front Microbiol 2023; 14:1190463. [PMID: 37234537 PMCID: PMC10206014 DOI: 10.3389/fmicb.2023.1190463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
The ongoing SARS-CoV-2 pandemic and the influenza epidemics have revived the interest in understanding how these highly contagious enveloped viruses respond to alterations in the physicochemical properties of their microenvironment. By understanding the mechanisms and conditions by which viruses exploit the pH environment of the host cell during endocytosis, we can gain a better understanding of how they respond to pH-regulated anti-viral therapies but also pH-induced changes in extracellular environments. This review provides a detailed explanation of the pH-dependent viral structural changes preceding and initiating viral disassembly during endocytosis for influenza A (IAV) and SARS coronaviruses. Drawing upon extensive literature from the last few decades and latest research, I analyze and compare the circumstances in which IAV and SARS-coronavirus can undertake endocytotic pathways that are pH-dependent. While there are similarities in the pH-regulated patterns leading to fusion, the mechanisms and pH activation differ. In terms of fusion activity, the measured activation pH values for IAV, across all subtypes and species, vary between approximately 5.0 to 6.0, while SARS-coronavirus necessitates a lower pH of 6.0 or less. The main difference between the pH-dependent endocytic pathways is that the SARS-coronavirus, unlike IAV, require the presence of specific pH-sensitive enzymes (cathepsin L) during endosomal transport. Conversely, the conformational changes in the IAV virus under acidic conditions in endosomes occur due to the specific envelope glycoprotein residues and envelope protein ion channels (viroporins) getting protonated by H+ ions. Despite extensive research over several decades, comprehending the pH-triggered conformational alterations of viruses still poses a significant challenge. The precise mechanisms of protonation mechanisms of certain during endosomal transport for both viruses remain incompletely understood. In absence of evidence, further research is needed.
Collapse
Affiliation(s)
- Amar Aganovic
- Faculty of Engineering Science and Technology, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
23
|
Ghafoori SM, Petersen GF, Conrady DG, Calhoun BM, Stigliano MZZ, Baydo RO, Grice R, Abendroth J, Lorimer DD, Edwards TE, Forwood JK. Structural characterisation of hemagglutinin from seven Influenza A H1N1 strains reveal diversity in the C05 antibody recognition site. Sci Rep 2023; 13:6940. [PMID: 37117205 PMCID: PMC10140725 DOI: 10.1038/s41598-023-33529-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/14/2023] [Indexed: 04/30/2023] Open
Abstract
Influenza virus (IV) causes several outbreaks of the flu each year resulting in an economic burden to the healthcare system in the billions of dollars. Several influenza pandemics have occurred during the last century and estimated to have caused 100 million deaths. There are four genera of IV, A (IVA), B (IVB), C (IVC), and D (IVD), with IVA being the most virulent to the human population. Hemagglutinin (HA) is an IVA surface protein that allows the virus to attach to host cell receptors and enter the cell. Here we have characterised the high-resolution structures of seven IVA HAs, with one in complex with the anti-influenza head-binding antibody C05. Our analysis revealed conserved receptor binding residues in all structures, as seen in previously characterised IV HAs. Amino acid conservation is more prevalent on the stalk than the receptor binding domain (RBD; also called the head domain), allowing the virus to escape from antibodies targeting the RBD. The equivalent site of C05 antibody binding to A/Denver/57 HA appears hypervariable in the other H1N1 IV HAs. Modifications within this region appear to disrupt binding of the C05 antibody, as these HAs no longer bind the C05 antibody by analytical SEC. Our study brings new insights into the structural and functional recognition of IV HA proteins and can contribute to further development of anti-influenza vaccines.
Collapse
Affiliation(s)
- Seyed Mohammad Ghafoori
- School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2650, Australia
| | - Gayle F Petersen
- School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2650, Australia
| | - Deborah G Conrady
- UCB BioSciences, Bainbridge Island, WA, 98110, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, 98109, USA
| | - Brandy M Calhoun
- UCB BioSciences, Bainbridge Island, WA, 98110, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, 98109, USA
| | - Matthew Z Z Stigliano
- UCB BioSciences, Bainbridge Island, WA, 98110, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, 98109, USA
| | - Ruth O Baydo
- UCB BioSciences, Bainbridge Island, WA, 98110, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, 98109, USA
| | - Rena Grice
- UCB BioSciences, Bainbridge Island, WA, 98110, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, 98109, USA
| | - Jan Abendroth
- UCB BioSciences, Bainbridge Island, WA, 98110, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, 98109, USA
| | - Donald D Lorimer
- UCB BioSciences, Bainbridge Island, WA, 98110, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, 98109, USA
| | - Thomas E Edwards
- UCB BioSciences, Bainbridge Island, WA, 98110, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, 98109, USA
| | - Jade K Forwood
- School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2650, Australia.
| |
Collapse
|
24
|
Raj S, Vishwakarma P, Saxena S, Kumar V, Khatri R, Kumar A, Singh M, Mishra S, Asthana S, Ahmed S, Samal S. Intradermal Immunization of Soluble Influenza HA Derived from a Lethal Virus Induces High Magnitude and Breadth of Antibody Responses and Provides Complete Protection In Vivo. Vaccines (Basel) 2023; 11:780. [PMID: 37112692 PMCID: PMC10141624 DOI: 10.3390/vaccines11040780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 04/05/2023] Open
Abstract
Immunogens mimicking the native-like structure of surface-exposed viral antigens are considered promising vaccine candidates. Influenza viruses are important zoonotic respiratory viruses with high pandemic potential. Recombinant soluble hemagglutinin (HA) glycoprotein-based protein subunit vaccines against Influenza have been shown to induce protective efficacy when administered intramuscularly. Here, we have expressed a recombinant soluble trimeric HA protein in Expi 293F cells and purified the protein derived from the Inf A/Guangdong-Maonan/ SWL1536/2019 virus which was found to be highly virulent in the mouse. The trimeric HA protein was found to be in the oligomeric state, highly stable, and the efficacy study in the BALB/c mouse challenge model through intradermal immunization with the prime-boost regimen conferred complete protection against a high lethal dose of homologous and mouse-adapted InfA/PR8 virus challenge. Furthermore, the immunogen induced high hemagglutinin inhibition (HI) titers and showed cross-protection against other Inf A and Inf B subtypes. The results are promising and warrant trimeric HA as a suitable vaccine candidate.
Collapse
Affiliation(s)
- Sneha Raj
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Preeti Vishwakarma
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Shikha Saxena
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Varun Kumar
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Ritika Khatri
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Amit Kumar
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Mrityunjay Singh
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Surbhi Mishra
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Shailendra Asthana
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Shubbir Ahmed
- Centralized Core Research Facility (CCRF), All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sweety Samal
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| |
Collapse
|
25
|
Pushan SS, Samantaray M, Rajagopalan M, Ramaswamy A. Evolution of Indian Influenza A (H1N1) Hemagglutinin Strains: A Comparative Analysis of the Pandemic Californian HA Strain. Front Mol Biosci 2023; 10:1111869. [PMID: 37006623 PMCID: PMC10061220 DOI: 10.3389/fmolb.2023.1111869] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/20/2023] [Indexed: 03/18/2023] Open
Abstract
The need for a vaccine/inhibitor design has become inevitable concerning the emerging epidemic and pandemic viral infections, and the recent outbreak of the influenza A (H1N1) virus is one such example. From 2009 to 2018, India faced severe fatalities due to the outbreak of the influenza A (H1N1) virus. In this study, the potential features of reported Indian H1N1 strains are analyzed in comparison with their evolutionarily closest pandemic strain, A/California/04/2009. The focus is laid on one of its surface proteins, hemagglutinin (HA), which imparts a significant role in attacking the host cell surface and its entry. The extensive analysis performed, in comparison with the A/California/04/2009 strain, revealed significant point mutations in all Indian strains reported from 2009 to 2018. Due to these mutations, all Indian strains disclosed altered features at the sequence and structural levels, which are further presumed to be associated with their functional diversity as well. The mutations observed with the 2018 HA sequence such as S91R, S181T, S200P, I312V, K319T, I419M, and E523D might improve the fitness of the virus in a new host and environment. The higher fitness and decreased sequence similarity of mutated strains may compromise therapeutic efficacy. In particular, the mutations observed commonly, such as serine-to-threonine, alanine-to-threonine, and lysine-to-glutamine at various regions, alter the physico-chemical features of receptor-binding domains, N-glycosylation, and epitope-binding sites when compared with the reference strain. Such mutations render diversity among all Indian strains, and the structural and functional characterization of these strains becomes inevitable. In this study, we observed that mutational drift results in the alteration of the receptor-binding domain, the generation of new variant N-glycosylation along with novel epitope-binding sites, and modifications at the structural level. Eventually, the pressing need to develop potentially distinct next-generation therapeutic inhibitors against the HA strains of the Indian influenza A (H1N1) virus is also highlighted here.
Collapse
Affiliation(s)
- Shilpa Sri Pushan
- Department of Bioinformatics, Pondicherry University, Puducherry, India
| | - Mahesh Samantaray
- Department of Bioinformatics, Pondicherry University, Puducherry, India
| | - Muthukumaran Rajagopalan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, India
| | - Amutha Ramaswamy
- Department of Bioinformatics, Pondicherry University, Puducherry, India
| |
Collapse
|
26
|
López-Valiñas Á, Valle M, Wang M, Darji A, Cantero G, Chiapponi C, Segalés J, Ganges L, Núñez JI. Vaccination against swine influenza in pigs causes different drift evolutionary patterns upon swine influenza virus experimental infection and reduces the likelihood of genomic reassortments. Front Cell Infect Microbiol 2023; 13:1111143. [PMID: 36992684 PMCID: PMC10040791 DOI: 10.3389/fcimb.2023.1111143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/14/2023] [Indexed: 03/16/2023] Open
Abstract
Influenza A viruses (IAVs) can infect a wide variety of bird and mammal species. Their genome is characterized by 8 RNA single stranded segments. The low proofreading activity of their polymerases and the genomic reassortment between different IAVs subtypes allow them to continuously evolve, constituting a constant threat to human and animal health. In 2009, a pandemic of an IAV highlighted the importance of the swine host in IAVs adaptation between humans and birds. The swine population and the incidence of swine IAV is constantly growing. In previous studies, despite vaccination, swine IAV growth and evolution were proven in vaccinated and challenged animals. However, how vaccination can drive the evolutionary dynamics of swine IAV after coinfection with two subtypes is poorly studied. In the present study, vaccinated and nonvaccinated pigs were challenged by direct contact with H1N1 and H3N2 independent swine IAVs seeder pigs. Nasal swab samples were daily recovered and broncho-alveolar lavage fluid (BALF) was also collected at necropsy day from each pig for swine IAV detection and whole genome sequencing. In total, 39 swine IAV whole genome sequences were obtained by next generation sequencing from samples collected from both experimental groups. Subsequently, genomic, and evolutionary analyses were carried out to detect both, genomic reassortments and single nucleotide variants (SNV). Regarding the segments found per sample, the simultaneous presence of segments from both subtypes was much lower in vaccinated animals, indicating that the vaccine reduced the likelihood of genomic reassortment events. In relation to swine IAV intra-host diversity, a total of 239 and 74 SNV were detected within H1N1 and H3N2 subtypes, respectively. Different proportions of synonymous and nonsynonymous substitutions were found, indicating that vaccine may be influencing the main mechanism that shape swine IAV evolution, detecting natural, neutral, and purifying selection in the different analyzed scenarios. SNV were detected along the whole swine IAV genome with important nonsynonymous substitutions on polymerases, surface glycoproteins and nonstructural proteins, which may have an impact on virus replication, immune system escaping and virulence of virus, respectively. The present study further emphasized the vast evolutionary capacity of swine IAV, under natural infection and vaccination pressure scenarios.
Collapse
Affiliation(s)
- Álvaro López-Valiñas
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
- *Correspondence: José I. Núñez, ; Álvaro López-Valiñas,
| | - Marta Valle
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
| | - Miaomiao Wang
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
| | - Ayub Darji
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
| | - Guillermo Cantero
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
| | - Chiara Chiapponi
- WOAH Reference Laboratory for Swine Influenza, Istituto Zooprofilattico Sperimentale della Lombardia ed Emilia-Romagna, Brescia, Italy
| | - Joaquim Segalés
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Llilianne Ganges
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
- WOAH Reference Laboratory for Classical Swine Fever, IRTA-CReSA, Barcelona, Spain
| | - José I. Núñez
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Barcelona, Spain
- *Correspondence: José I. Núñez, ; Álvaro López-Valiñas,
| |
Collapse
|
27
|
Son S, Ahn SB, Kim G, Jang Y, Ko C, Kim M, Kim SJ. Identification of broad-spectrum neutralizing antibodies against influenza A virus and evaluation of their prophylactic efficacy in mice. Antiviral Res 2023; 213:105591. [PMID: 37003306 DOI: 10.1016/j.antiviral.2023.105591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/12/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Influenza A virus continuously infects humans, and the antigenic shifts of this respiratory virus enable it to cross the species barrier, threatening public health with the risk of pandemics. Broadly neutralizing antibodies (bnAbs) that target the antigenic surface glycoprotein, hemagglutinin (HA), of influenza A virus protect against various subtypes of the virus. Here, we screened a human scFv library, through phage display and panning against recombinant HA proteins, to discover human monoclonal antibodies (mAbs) that are broadly active. Consequently, two human mAbs, named G1 and G2, were identified, which target the HA proteins of the H1N1 and H3N2 subtypes, respectively. G1, was shown to have broad binding ability to different HA subtypes of group 1. By contrast, G2 had higher binding affinity but sensed exclusively H3 subtype-derived HAs. In a cell culture-based virus-neutralizing assay, both G1 and G2 efficiently suppressed infection of the parental influenza A viruses of H1N1 and H3N2 subtypes. Mode-of-action studies showed that the G1 antibody blocked HA2-mediated membrane fusion. Meanwhile, G2 inhibited HA1-mediated viral attachment to host cells. It is noteworthy that both antibodies elicited antibody-dependent cellular cytotoxicity (ADCC) activities by recruiting FcγRIIIA-expressing effector cells. In mouse challenge models, single-shot, intraperitoneal administration of chimeric G1 and G2 antibodies with the mouse IgG constant region completely protected mice from viral infections at doses above 10 and 1 mg/kg, respectively. The newly identified bnAbs, G1 and G2, could provide insight into the development of broad-spectrum antivirals against future pandemic influenza A virus involving group 1- or H3-subtyped strains.
Collapse
Affiliation(s)
- Sumin Son
- Synthetic Biology and Bioengineering Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Soo Bin Ahn
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea; Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Geonyeong Kim
- Synthetic Biology and Bioengineering Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Yejin Jang
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Chunkyu Ko
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Meehyein Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea; Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Sang Jick Kim
- Synthetic Biology and Bioengineering Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
28
|
Fumagalli SE, Padhiar NH, Meyer D, Katneni U, Bar H, DiCuccio M, Komar AA, Kimchi-Sarfaty C. Analysis of 3.5 million SARS-CoV-2 sequences reveals unique mutational trends with consistent nucleotide and codon frequencies. Virol J 2023; 20:31. [PMID: 36812119 PMCID: PMC9936480 DOI: 10.1186/s12985-023-01982-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/02/2023] [Indexed: 02/19/2023] Open
Abstract
BACKGROUND Since the onset of the SARS-CoV-2 pandemic, bioinformatic analyses have been performed to understand the nucleotide and synonymous codon usage features and mutational patterns of the virus. However, comparatively few have attempted to perform such analyses on a considerably large cohort of viral genomes while organizing the plethora of available sequence data for a month-by-month analysis to observe changes over time. Here, we aimed to perform sequence composition and mutation analysis of SARS-CoV-2, separating sequences by gene, clade, and timepoints, and contrast the mutational profile of SARS-CoV-2 to other comparable RNA viruses. METHODS Using a cleaned, filtered, and pre-aligned dataset of over 3.5 million sequences downloaded from the GISAID database, we computed nucleotide and codon usage statistics, including calculation of relative synonymous codon usage values. We then calculated codon adaptation index (CAI) changes and a nonsynonymous/synonymous mutation ratio (dN/dS) over time for our dataset. Finally, we compiled information on the types of mutations occurring for SARS-CoV-2 and other comparable RNA viruses, and generated heatmaps showing codon and nucleotide composition at high entropy positions along the Spike sequence. RESULTS We show that nucleotide and codon usage metrics remain relatively consistent over the 32-month span, though there are significant differences between clades within each gene at various timepoints. CAI and dN/dS values vary substantially between different timepoints and different genes, with Spike gene on average showing both the highest CAI and dN/dS values. Mutational analysis showed that SARS-CoV-2 Spike has a higher proportion of nonsynonymous mutations than analogous genes in other RNA viruses, with nonsynonymous mutations outnumbering synonymous ones by up to 20:1. However, at several specific positions, synonymous mutations were overwhelmingly predominant. CONCLUSIONS Our multifaceted analysis covering both the composition and mutation signature of SARS-CoV-2 gives valuable insight into the nucleotide frequency and codon usage heterogeneity of SARS-CoV-2 over time, and its unique mutational profile compared to other RNA viruses.
Collapse
Affiliation(s)
- Sarah E Fumagalli
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Nigam H Padhiar
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Douglas Meyer
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Upendra Katneni
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Haim Bar
- Department of Statistics, University of Connecticut, Storrs, CT, USA
| | | | - Anton A Komar
- Department of Biological, Geological and Environmental Sciences, Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, USA
| | - Chava Kimchi-Sarfaty
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
29
|
Designing multi-epitope mRNA construct as a universal influenza vaccine candidate for future epidemic/pandemic preparedness. Int J Biol Macromol 2023; 226:885-899. [PMID: 36521707 DOI: 10.1016/j.ijbiomac.2022.12.066] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/25/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Despite the availability of prevention and treatment strategies and advancing immunization approaches, the influenza virus remains a global threat that continues to plague humanity with unpredictable pandemics. Due to the unusual genetic variability and segmented genome, the reassortment between different strains of influenza is facilitated and the viruses continuously evolve and adapt to the host cell's immunity. This underlies the seasonal vaccine mismatches that decrease the vaccine efficacy and increase the risk of outbreaks. Thus, the development of a universal vaccine covering all the influenza A and B strains would reduce the pervasiveness of the influenza virus. In the current study, a potentially universal influenza multi-epitope vaccine was designed based on the experimentally tested conserved T cell and B cell epitopes of hemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP), and matrix-2 proton channel (M2) of the virus. The immune simulation and molecular docking of the vaccine construct with TLR2, TLR3, and TLR4 elicited the favorable immunogenicity of the vaccine and the formation of stable complexes, respectively. Ultimately, based on the immunoinformatics analysis, the universal mRNA multi-epitope vaccine designed in this study might have a protection potential against the various subtypes of influenza A and B.
Collapse
|
30
|
Büyüksünetçi YT, Anık Ü. Electro-Nano Diagnostic Platform Based on Antibody-Antigen Interaction: An Electrochemical Immunosensor for Influenza A Virus Detection. BIOSENSORS 2023; 13:176. [PMID: 36831942 PMCID: PMC9953406 DOI: 10.3390/bios13020176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 06/18/2023]
Abstract
H1N1 is a kind of influenza A virus that causes serious health issues throughout the world. Its symptoms are more serious than seasonal flu and can sometimes be lethal. For this reason, rapid, accurate, and effective diagnostic tests are needed. In this study, an electrochemical immunosensor for the sensitive, selective, and practical detection of the H1N1 virus was developed. The sensor platform included multi-walled carbon nanotube gold-platinum (MWCNT-Au-Pt) hybrid nanomaterial and anti-hemagglutinin (anti-H1) monoclonal antibody. For the construction of this biosensor, a gold screen-printed electrode (AuSPE) was used as a transducer. Firstly, AuSPE was modified with MWCNT-Au-Pt hybrid nanomaterial via drop casting. Anti-H1 antibody was immobilized onto the electrode surface after the modification process with cysteamine was applied. Then, the effect of the interaction time with cysteamine for surface modification was investigated. Following that, the experimental parameters, such as the amount of hybrid nanomaterial and the concentration of anti-H1 were optimized. Under the optimized conditions, the analytical characteristics of the developed electrochemical immunosensor were investigated for the H1N1 virus by using electrochemical impedance spectroscopy. As a result, a linear range was obtained between 2.5-25.0 µg/mL with a limit of the detection value of 3.54 µg/mL. The relative standard deviation value for 20 µg/mL of the H1N1 virus was also calculated and found as 0.45% (n = 3). In order to determine the selectivity of the developed anti-H1-based electrochemical influenza A immunosensor, the response of this system towards the H3N2 virus was investigated. The matrix effect was also investigated by using synthetic saliva supplemented with H1N1 virus.
Collapse
Affiliation(s)
- Yudum Tepeli Büyüksünetçi
- Sensors, Biosensors and Nano-Diagnostic Laboratory, Research Laboratory Center, Mugla Sitki Kocman University, Kotekli, 48000 Mugla, Turkey
| | - Ülkü Anık
- Sensors, Biosensors and Nano-Diagnostic Laboratory, Research Laboratory Center, Mugla Sitki Kocman University, Kotekli, 48000 Mugla, Turkey
- Chemistry Department, Faculty of Science, Mugla Sitki Kocman University, Kotekli, 48000 Mugla, Turkey
| |
Collapse
|
31
|
Weidenbacher P, Musunuri S, Powell AE, Tang S, Do J, Sanyal M, Kim PS. Simplified Purification of Glycoprotein-Modified Ferritin Nanoparticles for Vaccine Development. Biochemistry 2023; 62:292-299. [PMID: 35960597 PMCID: PMC9850919 DOI: 10.1021/acs.biochem.2c00241] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/27/2022] [Indexed: 02/02/2023]
Abstract
Ferritin-based, self-assembling protein nanoparticle vaccines are being developed against a range of viral pathogens, including SARS-CoV-2, influenza, HIV-1, and Epstein-Barr virus. However, purification of these nanoparticles is often laborious and requires customization for each potential nanoparticle vaccine. We propose that the simple insertion of a polyhistidine tag into exposed flexible loops on the ferritin surface (His-Fer) can mitigate the need for complex purifications and enable facile metal-chelate-based purification, thereby allowing for optimization of early stage vaccine candidates. Using sequence homology and computational modeling, we identify four sites that can accommodate insertion of a polyhistidine tag and demonstrate purification of both hemagglutinin-modified and SARS-CoV-2 spike-modified ferritins, highlighting the generality of the approach. A site at the 4-fold axis of symmetry enables optimal purification of both protein nanoparticles. We demonstrate improved purification through modulating the polyhistidine length and optimizing both the metal cation and the resin type. Finally, we show that purified His-Fer proteins remain multimeric and elicit robust immune responses similar to those of their wild-type counterparts. Collectively, this work provides a simplified purification scheme for ferritin-based vaccines.
Collapse
Affiliation(s)
- Payton Weidenbacher
- Stanford
ChEM-H, Stanford University, Stanford, California 94305, United States
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Sriharshita Musunuri
- Stanford
ChEM-H, Stanford University, Stanford, California 94305, United States
- Department
of Biochemistry, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Abigail E. Powell
- Stanford
ChEM-H, Stanford University, Stanford, California 94305, United States
- Department
of Biochemistry, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Shaogeng Tang
- Stanford
ChEM-H, Stanford University, Stanford, California 94305, United States
- Department
of Biochemistry, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Jonathan Do
- Stanford
ChEM-H, Stanford University, Stanford, California 94305, United States
- Department
of Biochemistry, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Mrinmoy Sanyal
- Stanford
ChEM-H, Stanford University, Stanford, California 94305, United States
- Department
of Biochemistry, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Peter S. Kim
- Stanford
ChEM-H, Stanford University, Stanford, California 94305, United States
- Department
of Biochemistry, School of Medicine, Stanford
University, Stanford, California 94305, United States
- Chan
Zuckerberg Biohub, San Francisco, California 94158, United States
| |
Collapse
|
32
|
Stokes R, Kohlbrand AJ, Seo H, Sankaran B, Karges J, Cohen SM. Carboxylic Acid Isostere Derivatives of Hydroxypyridinones as Core Scaffolds for Influenza Endonuclease Inhibitors. ACS Med Chem Lett 2022; 14:75-82. [PMID: 36655124 PMCID: PMC9841593 DOI: 10.1021/acsmedchemlett.2c00434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022] Open
Abstract
Among the most important influenza virus targets is the RNA-dependent RNA polymerase acidic N-terminal (PAN) endonuclease, which is a critical component of the viral replication machinery. To inhibit the activity of this metalloenzyme, small-molecule inhibitors employ metal-binding pharmacophores (MBPs) that coordinate to the dinuclear Mn2+ active site. In this study, several metal-binding isosteres (MBIs) were examined where the carboxylic acid moiety of a hydroxypyridinone MBP is replaced with other groups to modulate the physicochemical properties of the compound. MBIs were evaluated for their ability to inhibit PAN using a FRET-based enzymatic assay, and their mode of binding in PAN was determined using X-ray crystallography.
Collapse
Affiliation(s)
- Ryjul
W. Stokes
- Department
of Chemistry and Biochemistry, University
of California, La Jolla, California 92093, United States
| | - Alysia J. Kohlbrand
- Department
of Chemistry and Biochemistry, University
of California, La Jolla, California 92093, United States
| | - Hyeonglim Seo
- Department
of Chemistry and Biochemistry, University
of California, La Jolla, California 92093, United States
| | - Banumathi Sankaran
- The
Berkeley Center for Structural Biology, Advanced Light Source, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Johannes Karges
- Department
of Chemistry and Biochemistry, University
of California, La Jolla, California 92093, United States
| | - Seth M. Cohen
- Department
of Chemistry and Biochemistry, University
of California, La Jolla, California 92093, United States,
| |
Collapse
|
33
|
Elkhatib WF, Abdelkareem SS, Khalaf WS, Shahin MI, Elfadil D, Alhazmi A, El-Batal AI, El-Sayyad GS. Narrative review on century of respiratory pandemics from Spanish flu to COVID-19 and impact of nanotechnology on COVID-19 diagnosis and immune system boosting. Virol J 2022; 19:167. [PMID: 36280866 PMCID: PMC9589879 DOI: 10.1186/s12985-022-01902-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 09/26/2022] [Indexed: 12/15/2022] Open
Abstract
The rise of the highly lethal severe acute respiratory syndrome-2 (SARS-2) as corona virus 2019 (COVID-19) reminded us of the history of other pandemics that happened in the last century (Spanish flu) and stayed in the current century, which include Severe-Acute-Respiratory-Syndrome (SARS), Middle-East-Respiratory-Syndrome (MERS), Corona Virus 2019 (COVID-19). We review in this report the newest findings and data on the origin of pandemic respiratory viral diseases, reservoirs, and transmission modes. We analyzed viral adaption needed for host switch and determinants of pathogenicity, causative factors of pandemic viruses, and symptoms and clinical manifestations. After that, we concluded the host factors associated with pandemics morbidity and mortality (immune responses and immunopathology, ages, and effect of pandemics on pregnancy). Additionally, we focused on the burdens of COVID-19, non-pharmaceutical interventions (quarantine, mass gatherings, facemasks, and hygiene), and medical interventions (antiviral therapies and vaccines). Finally, we investigated the nanotechnology between COVID-19 analysis and immune system boosting (Nanoparticles (NPs), antimicrobial NPs as antivirals and immune cytokines). This review presents insights about using nanomaterials to treat COVID-19, improve the bioavailability of the abused drugs, diminish their toxicity, and improve their performance.
Collapse
Affiliation(s)
- Walid F Elkhatib
- Microbiology and Immunology Department, Faculty of Pharmacy, Ain Shams University, African Union Organization St., Abbassia, Cairo, 11566, Egypt.
- Department of Microbiology and Immunology, Faculty of Pharmacy, Galala University, New Galala City, Suez, Egypt.
| | - Shereen S Abdelkareem
- Department of Alumni, School of Pharmacy and Pharmaceutical Industries, Badr University in Cairo (BUC), Entertainment Area, Badr City, Cairo, Egypt
| | - Wafaa S Khalaf
- Department of Microbiology and Immunology, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo, 11751, Egypt
| | - Mona I Shahin
- Zoology Department, Faculty of Tymaa, Tabuk University, Tymaa, 71491, Kingdom of Saudi Arabia
| | - Dounia Elfadil
- Biology and Chemistry Department, Hassan II University of Casablanca, Casablanca, Morocco
| | - Alaa Alhazmi
- Medical Laboratory Technology Department, Jazan University, Jazan, Saudi Arabia
- SMIRES for Consultation in Specialized Medical Laboratories, Jazan University, Jazan, Saudi Arabia
| | - Ahmed I El-Batal
- Drug Microbiology Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Gharieb S El-Sayyad
- Department of Microbiology and Immunology, Faculty of Pharmacy, Galala University, New Galala City, Suez, Egypt.
- Drug Microbiology Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt.
| |
Collapse
|
34
|
Rcheulishvili N, Papukashvili D, Liu C, Ji Y, He Y, Wang PG. Promising strategy for developing mRNA-based universal influenza virus vaccine for human population, poultry, and pigs- focus on the bigger picture. Front Immunol 2022; 13:1025884. [PMID: 36325349 PMCID: PMC9618703 DOI: 10.3389/fimmu.2022.1025884] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/03/2022] [Indexed: 08/08/2023] Open
Abstract
Since the first outbreak in the 19th century influenza virus has remained emergent owing to the huge pandemic potential. Only the pandemic of 1918 caused more deaths than any war in world history. Although two types of influenza- A (IAV) and B (IBV) cause epidemics annually, influenza A deserves more attention as its nature is much wilier. IAVs have a large animal reservoir and cause the infection manifestation not only in the human population but in poultry and domestic pigs as well. This many-sided characteristic of IAV along with the segmented genome gives rise to the antigenic drift and shift that allows evolving the new strains and new subtypes, respectively. As a result, the immune system of the body is unable to recognize them. Importantly, several highly pathogenic avian IAVs have already caused sporadic human infections with a high fatality rate (~60%). The current review discusses the promising strategy of using a potentially universal IAV mRNA vaccine based on conserved elements for humans, poultry, and pigs. This will better aid in averting the outbreaks in different susceptible species, thus, reduce the adverse impact on agriculture, and economics, and ultimately, prevent deadly pandemics in the human population.
Collapse
Affiliation(s)
| | | | | | | | - Yunjiao He
- *Correspondence: Yunjiao He, ; Peng George Wang,
| | | |
Collapse
|
35
|
Prada LSD, Sanz-Muñoz I, de Lejarazu RO, Eiros JM, García-Sastre A, Aydillo T. Immunodominance hierarchy after seasonal influenza vaccination. Emerg Microbes Infect 2022; 11:2670-2679. [PMID: 36219456 DOI: 10.1080/22221751.2022.2135460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Current influenza vaccines elicit humoral immune responses against the hemagglutinin (HA) protein of influenza viruses. Different antigenic sites have been identified in the HA head as the main target of hemagglutination inhibition (HAI) antibodies (Sb, Sa, Cb, Ca1 and Ca2). To determine immunodominance (ID) of each site, we performed HAI assays against a panel of mutant viruses, each one lacking one of the classically defined antigenic sites and compared it to wild type (Wt). Agglutinating antibodies were measured before and after vaccination in two different regimens: Quadrivalent Influenza Vaccine (QIV) in young adults; or Adjuvanted Trivalent influenza Vaccine (ATIV) in elderly. Our results showed abs before vaccination were significantly reduced against all antigenic sites in the elderly and only against Sb and Ca2 in young adults compared to the Wt. Humoral response to vaccination was significantly reduced against all viruses compared to the Wt for the ATIV and only against Sb and Ca2 for the QIV. The strongest reduction was observed in all cases against Sb followed by Ca2. We concluded that ID profile was clearly dominated by Sb followed by Ca2. Additionally, the antibody response evolved with age, increasing the response towards less immunodominant epitopes of HA head. Adjuvants can positively influence ID hierarchy broadening responses towards multiple antigenic sites of HA head.
Collapse
Affiliation(s)
- Laura Sánchez-de Prada
- National Influenza Centre of Valladolid, 47010, Spain.,Hospital Clínico Universitario de Valladolid, 47003, Spain
| | | | | | | | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Teresa Aydillo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
36
|
Zhao C, Pu J. Influence of Host Sialic Acid Receptors Structure on the Host Specificity of Influenza Viruses. Viruses 2022; 14:v14102141. [PMID: 36298694 PMCID: PMC9608321 DOI: 10.3390/v14102141] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 11/23/2022] Open
Abstract
Influenza viruses need to use sialic acid receptors to invade host cells, and the α-2,3 and α-2,6 sialic acids glycosidic bonds linking the terminal sialic acids are generally considered to be the most important factors influencing the cross-species transmission of the influenza viruses. The development of methods to detect the binding of influenza virus HA proteins to sialic acid receptors, as well as the development of glycobiological techniques, has led to a richer understanding of the structure of the sialylated glycan in influenza virus hosts. It was found that, in addition to the sialic acid glycosidic bond, sialic acid variants, length of the sialylated glycan, Gal-GlcNAc-linked glycosidic bond within the sialylated glycan, and sulfation/fucosylation of the GlcNAc within the sialylated glycan all affect the binding properties of influenza viruses to the sialic acid receptors, thus indirectly affecting the host specificity of influenza viruses. This paper will review the sialic acid variants, internal structural differences of sialylated glycan molecules that affect the host specificity of influenza viruses, and distribution characteristics of sialic acid receptors in influenza virus hosts, in order to provide a more reliable theoretical basis for the in-depth investigation of cross-species transmission of influenza viruses and the development of new antiviral drugs.
Collapse
|
37
|
An C, Wu Y, Wu J, Liu H, Zhou S, Ge D, Dong R, You L, Hao Y. Berberine ameliorates pulmonary inflammation in mice with influenza viral pneumonia by inhibiting NLRP3 inflammasome activation and gasdermin D‐mediated pyroptosis. Drug Dev Res 2022; 83:1707-1721. [DOI: 10.1002/ddr.21995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/10/2022] [Accepted: 08/23/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Chen An
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Yanmin Wu
- Department of Immunology, School of Medical Technology Qiqihar Medical University Qiqihar China
| | - Jun Wu
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Huanwei Liu
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Siyao Zhou
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Dongyu Ge
- Research and Test Center, School of Traditional Chinese Medicine Beijing University of Chinese Medicine Beijing China
| | - Ruijuan Dong
- Research and Test Center, School of Traditional Chinese Medicine Beijing University of Chinese Medicine Beijing China
| | - Leiming You
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Yu Hao
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| |
Collapse
|
38
|
Maddocks GM, Peterson KL, Downey ML, Lee BH, Lavoie JH, Menegatti S, Daniele M. Aptasensor for Detection of Influenza-A in Human Saliva. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:1262-1265. [PMID: 36086000 DOI: 10.1109/embc48229.2022.9871837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Access to low-cost, rapid, individualized diagnostics at point-of-care and point-of-need is vital to minimize the impact of highly infectious viruses, such as influenza. Herein, a biosensor for detecting hemagglutinin (HA), an abundant capsid protein in H1N1 viruses, is demonstrated. A gold working electrode was functionalized with a thiol-modified, HA-binding aptamer derivatized with a methylene blue modification for redox reporting. The aptamer was characterized by surface plasmon resonance to confirm its biorecognition activity for HA. The aptasensor was characterized by square wave voltammetry to quantify the sensor's response to varying concentrations of HA. The sensor exhibited a lower limit of detection of 1.5 pM with linear detection of up to 1.2 nM in both Tris buffer and simulated human saliva, thus encompassing the clinically relevant HA range in saliva. Average sensitivity was measured at 21.083 nA·nM-1in Tris and 14.5 nA·nM-1in artificial saliva across clinically relevant HA titers. Sensor stability across time was also investigated, providing a preliminary understanding of the translational viability of the aptasensors for mobile and remote diagnostic applications.
Collapse
|
39
|
The Contribution of Viral Proteins to the Synergy of Influenza and Bacterial Co-Infection. Viruses 2022; 14:v14051064. [PMID: 35632805 PMCID: PMC9143653 DOI: 10.3390/v14051064] [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: 04/06/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
A severe course of acute respiratory disease caused by influenza A virus (IAV) infection is often linked with subsequent bacterial superinfection, which is difficult to cure. Thus, synergistic influenza-bacterial co-infection represents a serious medical problem. The pathogenic changes in the infected host are accelerated as a consequence of IAV infection, reflecting its impact on the host immune response. IAV infection triggers a complex process linked with the blocking of innate and adaptive immune mechanisms required for effective antiviral defense. Such disbalance of the immune system allows for easier initiation of bacterial superinfection. Therefore, many new studies have emerged that aim to explain why viral-bacterial co-infection can lead to severe respiratory disease with possible fatal outcomes. In this review, we discuss the key role of several IAV proteins-namely, PB1-F2, hemagglutinin (HA), neuraminidase (NA), and NS1-known to play a role in modulating the immune defense of the host, which consequently escalates the development of secondary bacterial infection, most often caused by Streptococcus pneumoniae. Understanding the mechanisms leading to pathological disorders caused by bacterial superinfection after the previous viral infection is important for the development of more effective means of prevention; for example, by vaccination or through therapy using antiviral drugs targeted at critical viral proteins.
Collapse
|
40
|
Ryt-Hansen P, Nielsen HG, Sørensen SS, Larsen I, Kristensen CS, Larsen LE. The role of gilts in transmission dynamics of swine influenza virus and impacts of vaccination strategies and quarantine management. Porcine Health Manag 2022; 8:19. [PMID: 35513878 PMCID: PMC9069814 DOI: 10.1186/s40813-022-00261-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Along with an expanding global swine production, the commercial housing and management of swine herds, provide an optimal environment for constant circulation of swine influenza virus (swIAV), thereby challenging farmers and veterinarian in determining optimal control measures. The aim of this study was to investigate the role of gilts in the swIAV transmission dynamics, and to evaluate the impact of different control measures such as quarantine and gilt vaccination. METHODS The study was conducted as a cross-sectional study in ten Danish sow herds, including five swIAV vaccinated and five unvaccinated herds. Blood- and nasal swab samples of gilts, first parity sows and their piglets were collected at different stages in the production system (quarantine in/out, mating, gestation and farrowing) and analyzed for the presence of swIAV and swIAV antibodies. Associations between the detection of swIAV, seroprevalence, antibody levels, sow and gilt vaccination strategy and quarantine biosecurity were thereafter investigated to identify possible risk factors for swIAV introductions and persistence within the herds. RESULTS Nine of the ten herds of the study had swIAV circulation and swIAV was detected in the quarantine, mating- and farrowing unit. The prevalence of seropositive gilts and first parity sows was significantly higher in the vaccinated herds, but swIAV was still present in nasal swabs from both gilts, first parity sows and piglets in these herds. Quarantine gilt vaccination and all-in/all-out management resulted in a significant reduction of swIAV positive gilts at the end of the quarantine period. CONCLUSION The results underline that herd vaccination and/or quarantine facilities are crucial to avoid swIAV introductions into sow herds.
Collapse
Affiliation(s)
- Pia Ryt-Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 2, 1870 Frederiksberg C, Denmark
| | - Henriette Guldberg Nielsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 2, 1870 Frederiksberg C, Denmark
| | - Simon Smed Sørensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 2, 1870 Frederiksberg C, Denmark
| | - Inge Larsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 2, 1870 Frederiksberg C, Denmark
| | | | - Lars Erik Larsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 2, 1870 Frederiksberg C, Denmark
| |
Collapse
|
41
|
Bull MB, Gu H, Ma FNL, Perera LP, Poon LLM, Valkenburg SA. Next-generation T cell-activating vaccination increases influenza virus mutation prevalence. SCIENCE ADVANCES 2022; 8:eabl5209. [PMID: 35385318 PMCID: PMC8986104 DOI: 10.1126/sciadv.abl5209] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 02/11/2022] [Indexed: 06/14/2023]
Abstract
To determine the potential for viral adaptation to T cell responses, we probed the full influenza virus genome by next-generation sequencing directly ex vivo from infected mice, in the context of an experimental T cell-based vaccine, an H5N1-based viral vectored vaccinia vaccine Wyeth/IL-15/5Flu, versus the current standard-of-care, seasonal inactivated influenza vaccine (IIV) and unvaccinated conditions. Wyeth/IL-15/5Flu vaccination was coincident with increased mutation incidence and frequency across the influenza genome; however, mutations were not enriched within T cell epitope regions, but high allele frequency mutations within conserved hemagglutinin stem regions and PB2 mammalian adaptive mutations arose. Depletion of CD4+ and CD8+ T cell subsets led to reduced frequency of mutants in vaccinated mice; therefore, vaccine-mediated T cell responses were important drivers of virus diversification. Our findings suggest that Wyeth/IL-15/5Flu does not generate T cell escape mutants but increases stochastic events for virus adaptation by stringent bottlenecks.
Collapse
Affiliation(s)
- Maireid B. Bull
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Haogao Gu
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Fionn N. L. Ma
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Liyanage P. Perera
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1374, USA
| | - Leo L. M. Poon
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sophie A. Valkenburg
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Microbiology and Immunology, at The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
42
|
SARS-CoV-2 Spike Protein Binding of Glycated Serum Albumin-Its Potential Role in the Pathogenesis of the COVID-19 Clinical Syndromes and Bias towards Individuals with Pre-Diabetes/Type 2 Diabetes and Metabolic Diseases. Int J Mol Sci 2022; 23:ijms23084126. [PMID: 35456942 PMCID: PMC9030890 DOI: 10.3390/ijms23084126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 01/08/2023] Open
Abstract
The immune response to SARS-CoV-2 infection requires antibody recognition of the spike protein. In a study designed to examine the molecular features of anti-spike and anti-nucleocapsid antibodies, patient plasma proteins binding to pre-fusion stabilised complete spike and nucleocapsid proteins were isolated and analysed by matrix-assisted laser desorption ionisation–time of flight (MALDI-ToF) mass spectrometry. Amongst the immunoglobulins, a high affinity for human serum albumin was evident in the anti-spike preparations. Careful mass comparison revealed the preferential capture of advanced glycation end product (AGE) forms of glycated human serum albumin by the pre-fusion spike protein. The ability of bacteria and viruses to surround themselves with serum proteins is a recognised immune evasion and pathogenic process. The preference of SARS-CoV-2 for AGE forms of glycated serum albumin may in part explain the severity and pathology of acute respiratory distress and the bias towards the elderly and those with (pre)diabetic and atherosclerotic/metabolic disease.
Collapse
|
43
|
Martins de Camargo M, Caetano AR, Ferreira de Miranda Santos IK. Evolutionary pressures rendered by animal husbandry practices for avian influenza viruses to adapt to humans. iScience 2022; 25:104005. [PMID: 35313691 PMCID: PMC8933668 DOI: 10.1016/j.isci.2022.104005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Commercial poultry operations produce and crowd billions of birds every year, which is a source of inexpensive animal protein. Commercial poultry is intensely bred for desirable production traits, and currently presents very low variability at the major histocompatibility complex. This situation dampens the advantages conferred by the MHC’s high genetic variability, and crowding generates immunosuppressive stress. We address the proteins of influenza A viruses directly and indirectly involved in host specificities. We discuss how mutants with increased virulence and/or altered host specificity may arise if few class I alleles are the sole selective pressure on avian viruses circulating in immunocompromised poultry. This hypothesis is testable with peptidomics of MHC ligands. Breeding strategies for commercial poultry can easily and inexpensively include high variability of MHC as a trait of interest, to help save billions of dollars as a disease burden caused by influenza and decrease the risk of selecting highly virulent strains.
Collapse
|
44
|
Pandey VK, Sharma R, Prajapati GK, Mohanta TK, Mishra AK. N-glycosylation, a leading role in viral infection and immunity development. Mol Biol Rep 2022; 49:8109-8120. [PMID: 35364718 PMCID: PMC8974804 DOI: 10.1007/s11033-022-07359-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 03/10/2022] [Indexed: 12/26/2022]
Abstract
N-linked protein glycosylation is an essential co-and posttranslational protein modification that occurs in all three domains of life; the assembly of N-glycans follows a complex sequence of events spanning the (Endoplasmic Reticulum) ER and the Golgi apparatus. It has a significant impact on both physicochemical properties and biological functions. It plays a significant role in protein folding and quality control, glycoprotein interaction, signal transduction, viral attachment, and immune response to infection. Glycoengineering of protein employed for improving protein properties and plays a vital role in the production of recombinant glycoproteins and struggles to humanize recombinant therapeutic proteins. It considers an alternative platform for biopharmaceuticals production. Many immune proteins and antibodies are glycosylated. Pathogen’s glycoproteins play vital roles during the infection cycle and their expression of specific oligosaccharides via the N-glycosylation pathway to evade detection by the host immune system. This review focuses on the aspects of N-glycosylation processing, glycoengineering approaches, their role in viral attachment, and immune responses to infection.
Collapse
Affiliation(s)
- Vijay Kant Pandey
- Department of Agriculture, Netaji Subhas University, Jamshedpur, Jharkhand, India
| | - Rajani Sharma
- Department of Biotechnology, Amity University Jharkhand, Niwaranpur, Ranchi, 834002, India.
| | | | | | - Awdhesh Kumar Mishra
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea.
| |
Collapse
|
45
|
Di Gennaro F, Petrosillo N. New endemic and pandemic pathologies with interhuman airborne transmission through ear, nose and throat anatomical sites. ACTA OTORHINOLARYNGOLOGICA ITALICA : ORGANO UFFICIALE DELLA SOCIETA ITALIANA DI OTORINOLARINGOLOGIA E CHIRURGIA CERVICO-FACCIALE 2022; 42:S5-S13. [PMID: 35763270 PMCID: PMC9137375 DOI: 10.14639/0392-100x-suppl.1-42-2022-01] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/10/2022] [Indexed: 12/23/2022]
Abstract
The current severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has once again stigmatised the importance of airborne pathogens and their clinical, social and public health impact. Respiratory viruses are transmitted between individuals when the pathogen is released from the upper airways or from the lower respiratory tract of an infected individual. Airborne transmission is defined as the inhalation of the infectious aerosol, named droplet nuclei which size is smaller than 5 mm and that can be inhaled at a distance up to 2 metres. This route of transmission is relevant for viral respiratory pathogens, including severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome (MERS)-CoV, influenza virus, human rhinovirus, respiratory syncytial virus (RSV) and other respiratory virus families that differ in viral and genomic structures, susceptibility of a population to the infection, severity, transmissibility, ways of transmission and seasonal recurrence. Human respiratory viruses generally infect cells of the upper respiratory tract, eliciting respiratory signs and symptoms, sometimes without the possibility to differentiate them clinically. As seen by the current Coronavirus Disease 2019 (COVID-19) pandemic, human respiratory viruses can substantially contribute to increased morbidity and mortality, economic losses and, eventually, social disruption. In this article, we describe the structural, clinical and transmission aspects of the main respiratory viruses responsible for endemic, epidemic and pandemic infections.
Collapse
Affiliation(s)
| | - Nicola Petrosillo
- Infection Control & Infectious Disease Service, University Hospital Campus Bio-Medico, Rome, Italy
| |
Collapse
|
46
|
Characterization of Immune Response towards Generation of Universal Anti-HA-Stalk Antibodies after Immunization of Broiler Hens with Triple H5N1/NA-HA-M1 VLPs. Viruses 2022; 14:v14040730. [PMID: 35458460 PMCID: PMC9029564 DOI: 10.3390/v14040730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
(1) Background: Avian influenza viruses (AIVs) promptly evade preexisting immunity by constantly altering the immunodominant neutralizing antibody epitopes (antigenic drift) or by procuring new envelope serotypes (antigenic shift). As a consequence, the majority of antibodies elicited by infection or vaccination protect only against closely related strains. The immunodominance of the globular head of the main glycoprotein has been shown to mask the immunogenicity of the conserved regions located within the hemagglutinin (HA) protein. It has been shown that the broadly neutralizing universal antibodies recognize the HA2 domain in headless hemagglutinin (HA-stalk). Therefore, the HA-stalk is a highly conserved antigen, which makes it a good candidate to be used in universal vaccine development against AIVs. (2) Methods: Sf9 insect cells were used to produce triple H5N1/NA-HA-M1 influenza virus-like particles (VLPs) via co-expression of neuraminidase, hemagglutinin and matrix proteins from a tricistronic expression cassette. Purified influenza VLPs were used to immunize broiler hens. An in-depth characterization of the immune response was performed with an emphasis on the pool of elicited universal antibodies. (3) Results: Our findings suggest, that after vaccination with triple H5N1/NA-HA-M1 VLPs, hens generate a pool of broad-spectrum universal anti-HA-stalk antibodies. Furthermore, these universal antibodies are able to recognize the mammalian-derived HA-stalk recombinant proteins from homologous H5N1 and heterologous H7N9 AIVs as well as from the heterosubtypic human H1N1 influenza strain. (4) Conclusions: Our findings may suggest that highly pathogenic avian influenza H5 HA protein contain functional epitopes that are attractive targets for the generation of broad-spectrum antibodies against AIVs in their native hosts.
Collapse
|
47
|
MCMV-based vaccine vectors expressing full-length viral proteins provide long-term humoral immune protection upon a single-shot vaccination. Cell Mol Immunol 2022; 19:234-244. [PMID: 34992275 PMCID: PMC8739032 DOI: 10.1038/s41423-021-00814-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/25/2021] [Indexed: 11/15/2022] Open
Abstract
Global pandemics caused by influenza or coronaviruses cause severe disruptions to public health and lead to high morbidity and mortality. There remains a medical need for vaccines against these pathogens. CMV (cytomegalovirus) is a β-herpesvirus that induces uniquely robust immune responses in which remarkably large populations of antigen-specific CD8+ T cells are maintained for a lifetime. Hence, CMV has been proposed and investigated as a novel vaccine vector for expressing antigenic peptides or proteins to elicit protective cellular immune responses against numerous pathogens. We generated two recombinant murine CMV (MCMV) vaccine vectors expressing hemagglutinin (HA) of influenza A virus (MCMVHA) or the spike protein of severe acute respiratory syndrome coronavirus 2 (MCMVS). A single injection of MCMVs expressing either viral protein induced potent neutralizing antibody responses, which strengthened over time. Importantly, MCMVHA-vaccinated mice were protected from illness following challenge with the influenza virus, and we excluded that this protection was due to the effects of memory T cells. Conclusively, we show here that MCMV vectors induce not only long-term cellular immunity but also humoral responses that provide long-term immune protection against clinically relevant respiratory pathogens.
Collapse
|
48
|
Sriwilaijaroen N, Suzuki Y. Roles of Glycans and Non-glycans on the Epithelium and in the Immune System in H1-H18 Influenza A Virus Infections. Methods Mol Biol 2022; 2556:205-242. [PMID: 36175637 DOI: 10.1007/978-1-0716-2635-1_16] [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] [Indexed: 06/16/2023]
Abstract
The large variation of influenza A viruses (IAVs) in various susceptible hosts and their rapid evolution, which allows host/tissue switching, host immune escape, vaccine escape, and drug resistance, are difficult challenges for influenza control in all countries worldwide. Access and binding of the IAV to actual receptors at endocytic sites is critical for the establishment of influenza infection. In this chapter, the progress in identification of and roles of glycans and non-glycans on the epithelium and in the immune system in H1-H18 IAV infections are reviewed. The first part of the review is on current knowledge of H1-H16 IAV receptors on the epithelium including sialyl glycans, other negatively charged glycans, and annexins. The second part of the review focuses on H1-H16 IAV receptors in the immune system including acidic surfactant phospholipids, Sia on surfactant proteins, the carbohydrate recognition domain (CRD) of surfactant proteins, Sia on mucins, Sia and C-type lectins on macrophages and dendritic cells, and Sia on NK cells. The third part of the review is about a possible H17-H18 IAV receptor. Binding of these receptors to IAVs may result in inhibition or enhancement of IAV infection depending on their location, host cell type, and IAV strain. Among these receptors, host sialyl glycans are key determinants of viral hemagglutinin (HA) lectins for H1-H16 infections. HA must acquire mutations to bind to sialyl glycans that are dominant on a new target tissue when switching to a new host for efficient transmission and to bind to long sialyl glycans found in the case of seasonal HAs with multiple glycosylation sites as a consequence of immune evasion. Although sialyl receptors/C-type lectins on immune cells are decoy receptors/pathogen recognition receptors for capturing viral HA lectin/glycans protecting HA antigenic sites, some IAV strains do not escape, such as by release with neuraminidase, but hijack these molecules to gain entry and replication in immune cells. An understanding of the virus-host battle tactics at the receptor level might lead to the establishment of novel strategies for effective control of influenza.
Collapse
Affiliation(s)
- Nongluk Sriwilaijaroen
- Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, Thailand.
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan.
| | - Yasuo Suzuki
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
49
|
Suda Y, Tajima Y, Nishi JI, Kajiya T. Diagnostic Method for COVID-19 Using Sugar Chain-Immobilized Nanoparticles and Saliva Specimens. Methods Mol Biol 2022; 2452:63-74. [PMID: 35554901 DOI: 10.1007/978-1-0716-2111-0_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Identification of viruses that infects animals or plants, and determination of their quantity are essential for the diagnosis of infectious disease and for the determination of a strategy in the treatment of virus-derived diseases. However, the concentration of viruses existing in a living body (in bodily fluid), food, drinking water, river water, and so on. is not high enough to be detected using conventional diagnostic methods. For example, since the concentration of influenza virus released from an infected person is less than the detection limit of conventional simple examination kits (rapid kit) or even a PCR process at the initial stage of infection, it is difficult to detect the presence of influenza virus which will lead to influenza disease. Our technology allows for safe, efficient, and selective concentration of viruses without troublesome ultracentrifugation, using sugar chain-immobilized metal nanoparticles based on the binding interaction between viruses and sugar chains. For COVID-19, we have developed and commercialized two molecular diagnosis kits: SUDx SARS-CoV-2 detection kit, and SGNP nCoV/Flu PCR detection kit, for the Japanese market in 2020.
Collapse
Affiliation(s)
- Yasuo Suda
- Department of Chemistry and Biotechnology, Graduate School of Science and Engineering, Kagoshima University, Kagoshima, Japan.
| | - Yasuhisa Tajima
- Division of Infectious Disease, Hamamatsu Medical Center, Shizuoka, Japan
| | - Jun-Ichiro Nishi
- Department of Microbiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Takashi Kajiya
- Clinical Research Center, Tenyoukai Central Hospital, Kagoshima, Japan
| |
Collapse
|
50
|
Jung K, Corrigan N, Wong EHH, Boyer C. Bioactive Synthetic Polymers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105063. [PMID: 34611948 DOI: 10.1002/adma.202105063] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/13/2021] [Indexed: 05/21/2023]
Abstract
Synthetic polymers are omnipresent in society as textiles and packaging materials, in construction and medicine, among many other important applications. Alternatively, natural polymers play a crucial role in sustaining life and allowing organisms to adapt to their environments by performing key biological functions such as molecular recognition and transmission of genetic information. In general, the synthetic and natural polymer worlds are completely separated due to the inability for synthetic polymers to perform specific biological functions; in some cases, synthetic polymers cause uncontrolled and unwanted biological responses. However, owing to the advancement of synthetic polymerization techniques in recent years, new synthetic polymers have emerged that provide specific biological functions such as targeted molecular recognition of peptides, or present antiviral, anticancer, and antimicrobial activities. In this review, the emergence of this generation of bioactive synthetic polymers and their bioapplications are summarized. Finally, the future opportunities in this area are discussed.
Collapse
Affiliation(s)
- Kenward Jung
- Cluster for Advanced Macromolecular Design (CAMD), Australian Centre for Nanomedicine (ACN), and School of Chemical Engineering, University of New South Wales (UNSW) Sydney, Sydney, NSW, 2052, Australia
| | - Nathaniel Corrigan
- Cluster for Advanced Macromolecular Design (CAMD), Australian Centre for Nanomedicine (ACN), and School of Chemical Engineering, University of New South Wales (UNSW) Sydney, Sydney, NSW, 2052, Australia
| | - Edgar H H Wong
- Cluster for Advanced Macromolecular Design (CAMD), Australian Centre for Nanomedicine (ACN), and School of Chemical Engineering, University of New South Wales (UNSW) Sydney, Sydney, NSW, 2052, Australia
| | - Cyrille Boyer
- Cluster for Advanced Macromolecular Design (CAMD), Australian Centre for Nanomedicine (ACN), and School of Chemical Engineering, University of New South Wales (UNSW) Sydney, Sydney, NSW, 2052, Australia
| |
Collapse
|