1
|
Huang X, Cheng Z, Lv Y, Li W, Liu X, Huang W, Zhao C. Neutralization potency of the 2023-24 seasonal influenza vaccine against circulating influenza H3N2 strains. Hum Vaccin Immunother 2024; 20:2380111. [PMID: 39205645 PMCID: PMC11364067 DOI: 10.1080/21645515.2024.2380111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/27/2024] [Accepted: 07/11/2024] [Indexed: 09/04/2024] Open
Abstract
Seasonal influenza is a severe disease that significantly impacts public health, causing millions of infections and hundreds of thousands of deaths each year. Seasonal influenza viruses, particularly the H3N2 subtype, exhibit high antigenic variability, often leading to mismatch between vaccine strains and circulating strains. Therefore, rapidly assessing the alignment between existing seasonal influenza vaccine and circulating strains is crucial for enhancing vaccine efficacy. This study, based on a pseudovirus platform, evaluated the match between current influenza H3N2 vaccine strains and circulating strains through cross-neutralization assays using clinical human immune sera against globally circulating influenza virus strains. The research results show that although mutations are present in the circulating strains, the current H3N2 vaccine strain still imparting effective protection, providing a scientific basis for encouraging influenza vaccination. This research methodology can be sustainably applied for the neutralization potency assessment of subsequent circulating strains, establishing a persistent methodological framework.
Collapse
Affiliation(s)
- Xiande Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Ziqi Cheng
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Yake Lv
- Center of Vaccine Clinical Evaluation, Institute for Immunization Program, Shaanxi Provincial Centre for Disease Control and Prevention, Xi’an, Shaanxi Province, China
| | - Weixuan Li
- Center of Vaccine Clinical Evaluation, Institute for Immunization Program, Shaanxi Provincial Centre for Disease Control and Prevention, Xi’an, Shaanxi Province, China
| | - Xiaoyu Liu
- Center of Vaccine Clinical Evaluation, Institute for Immunization Program, Shaanxi Provincial Centre for Disease Control and Prevention, Xi’an, Shaanxi Province, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Chenyan Zhao
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| |
Collapse
|
2
|
Delgadillo-Gutiérrez K, Castelán-Vega JA, Jiménez-Alberto A, Fernández-Lizárraga MDC, Aparicio-Ozores G, Monterrubio-López GP, Ribas-Aparicio RM. Characterization and use in neutralization assays of avian influenza codon-optimized H5 and H7 retroviral pseudotypes. J Virol Methods 2021; 300:114391. [PMID: 34890710 DOI: 10.1016/j.jviromet.2021.114391] [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: 09/22/2021] [Revised: 11/12/2021] [Accepted: 11/27/2021] [Indexed: 10/19/2022]
Abstract
Influenza is a relevant problem for public and animal health, with a significant economic impact. In recent years, outbreaks of avian influenza virus have resulted in devastating losses in the poultry industry worldwide, and although its transmission to humans is very rare, there is always a potential risk for an even more severe outbreak. Currently, vaccination is considered the most effective tool for the control and prevention of influenza infections in both humans and animals. The maintenance of animal welfare and the successful implementation of animal health programs depend on the timely administration of vaccines, which must comply with quality specifications indicated by health authorities; for example, the capability to ensure a minimum antibody titer. The production of viral antigens used in these tests can pose a biosafety risk, and some viral strains can be difficult to grow. Therefore, new biotechnological alternatives are required to overcome these disadvantages. In this study, we produced pseudotypes carrying H5 and H7 hemagglutinins from lowly and highly pathogenic avian influenza viruses. These pseudotypes were used in neutralization assays to detect neutralizing antibodies in avian sera, which were confirmed positive by inhibition of the hemagglutination test. Our results showed that the pseudotype neutralization assay is a viable alternative for the detection of neutralizing antibodies, by demonstrating subtype specificity and requiring reduced biosafety requirements. Therefore, it represents a versatile platform that can facilitate technology transfer protocols between laboratories, and an immediate application in serological tools for quality control of veterinary vaccines against avian influenza.
Collapse
Affiliation(s)
- Karen Delgadillo-Gutiérrez
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Juan Arturo Castelán-Vega
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Alicia Jiménez-Alberto
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | | | - Gerardo Aparicio-Ozores
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Gloria Paulina Monterrubio-López
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Rosa María Ribas-Aparicio
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Mexico City, Mexico.
| |
Collapse
|
3
|
Chen TH, Yang YL, Jan JT, Chen CC, Wu SC. Site-Specific Glycan-Masking/Unmasking Hemagglutinin Antigen Design to Elicit Broadly Neutralizing and Stem-Binding Antibodies Against Highly Pathogenic Avian Influenza H5N1 Virus Infections. Front Immunol 2021; 12:692700. [PMID: 34335603 PMCID: PMC8317614 DOI: 10.3389/fimmu.2021.692700] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/29/2021] [Indexed: 11/17/2022] Open
Abstract
The highly pathogenic avian influenza (HPAI) H5N1 viruses with the capability of transmission from birds to humans have a serious impact on public health. To date, HPAI H5N1 viruses have evolved into ten antigenically distinct clades that could cause a mismatch of vaccine strains and reduce vaccine efficacy. In this study, the glycan masking and unmasking strategies on hemagglutinin antigen were used for designing two antigens: H5-dm/st2 and H5-tm/st2, and investigated for their elicited immunity using two-dose recombinant H5 (rH5) immunization and a first-dose adenovirus vector prime, followed by a second-dose rH5 protein booster immunization. The H5-dm/st2 antigen was found to elicit broadly neutralizing antibodies against different H5N1 clade/subclade viruses, as well as more stem-binding antibodies to inhibit HA-facilitated membrane fusion activity. Mice immunized with the H5-dm/st2 antigen had a higher survival rate when challenged with homologous and heterologous clades of H5N1 viruses. Mutant influenza virus replaced with the H5-dm/st2 gene generated by reverse genetics (RG) technology amplified well in MDCK cells and embryonated chicken eggs. Again, the inactivated H5N1-dm/st2 RG virus elicited more potent cross-clade neutralizing and anti-fusion antibodies in sera. Therefore, the H5N1-dm/st2 RG virus with the site-specific glycan-masking on the globular head and the glycan-unmasking on the stem region of H5 antigen can be used for further development of cross-protective H5N1 vaccines.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- Antigens, Viral/administration & dosage
- Antigens, Viral/immunology
- Broadly Neutralizing Antibodies/blood
- Chick Embryo
- Disease Models, Animal
- Dogs
- Female
- Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Immunization
- Immunodominant Epitopes
- Immunogenicity, Vaccine
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Madin Darby Canine Kidney Cells
- Mice, Inbred BALB C
- Orthomyxoviridae Infections/blood
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Polysaccharides/administration & dosage
- Polysaccharides/immunology
- Mice
Collapse
Affiliation(s)
- Ting-Hsuan Chen
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ya-Lin Yang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Jia-Tsrong Jan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Chung-Chu Chen
- Department of Internal Medicine, MacKay Memorial Hospital, Hsinchu, Taiwan
- Teaching Center of Natural Science, Minghsin University of Science and Technology, Hsinchu, Taiwan
| | - Suh-Chin Wu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
4
|
Toon K, Bentley EM, Mattiuzzo G. More Than Just Gene Therapy Vectors: Lentiviral Vector Pseudotypes for Serological Investigation. Viruses 2021; 13:217. [PMID: 33572589 PMCID: PMC7911487 DOI: 10.3390/v13020217] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
Serological assays detecting neutralising antibodies are important for determining the immune responses following infection or vaccination and are also often considered a correlate of protection. The target of neutralising antibodies is usually located in the Envelope protein on the viral surface, which mediates cell entry. As such, presentation of the Envelope protein on a lentiviral particle represents a convenient alternative to handling of a potentially high containment virus or for those viruses with no established cell culture system. The flexibility, relative safety and, in most cases, ease of production of lentiviral pseudotypes, have led to their use in serological assays for many applications such as the evaluation of candidate vaccines, screening and characterization of anti-viral therapeutics, and sero-surveillance. Above all, the speed of production of the lentiviral pseudotypes, once the envelope sequence is published, makes them important tools in the response to viral outbreaks, as shown during the COVID-19 pandemic in 2020. In this review, we provide an overview of the landscape of the serological applications of pseudotyped lentiviral vectors, with a brief discussion on their production and batch quality analysis. Finally, we evaluate their role as surrogates for the real virus and possible alternatives.
Collapse
Affiliation(s)
- Kamilla Toon
- Division of Virology, National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms EN6 3QG, UK;
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Emma M. Bentley
- Division of Virology, National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms EN6 3QG, UK;
| | - Giada Mattiuzzo
- Division of Virology, National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms EN6 3QG, UK;
| |
Collapse
|
5
|
Tang N, Lu CY, Sue SC, Chen TH, Jan JT, Huang MH, Huang CH, Chen CC, Chiang BL, Huang LM, Wu SC. Type IIb Heat Labile Enterotoxin B Subunit as a Mucosal Adjuvant to Enhance Protective Immunity against H5N1 Avian Influenza Viruses. Vaccines (Basel) 2020; 8:vaccines8040710. [PMID: 33266210 PMCID: PMC7768441 DOI: 10.3390/vaccines8040710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/18/2020] [Accepted: 11/29/2020] [Indexed: 11/16/2022] Open
Abstract
Human infections with highly pathogenic avian influenza H5N1 viruses persist as a major global health concern. Vaccination remains the primary protective strategy against H5N1 and other novel avian influenza virus infections. We investigated the use of E. coli type IIb heat labile enterotoxin B subunit (LTIIb-B5) as a mucosal adjuvant for intranasal immunizations with recombinant HA proteins against H5N1 avian influenza viruses. Use of LTIIb-B5 adjuvant elicited more potent IgG, IgA, and neutralizing antibody titers in both sera and bronchoalveolar lavage fluids, thus increasing protection against lethal virus challenges. LTIIb-B5 mucosal adjuvanticity was found to trigger stronger Th17 cellular response in spleen lymphocytes and cervical lymph nodes. Studies of anti-IL-17A monoclonal antibody depletion and IL-17A knockout mice also suggest the contribution from Th17 cellular response to anti-H5N1 protective immunity. Our results indicate a link between improved protection against H5N1 live virus challenges and increased Th17 response due to the use of LTIIb-B5 mucosal adjuvant with HA subunit proteins.
Collapse
Affiliation(s)
- Neos Tang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan; (N.T.); (T.-H.C.)
| | - Chun-Yi Lu
- Department of Pediatrics, National Taiwan University Children Hospital, Taipei 100226, Taiwan; (C.-Y.L.); (B.-L.C.); (L.-M.H.)
| | - Shih-Che Sue
- Department of Life Science, Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Ting-Hsuan Chen
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan; (N.T.); (T.-H.C.)
| | - Jia-Tsrong Jan
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan;
| | - Ming-Hsi Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan 35053, Taiwan;
| | - Chung-Hsiung Huang
- Department of Food Science, National Taiwan Ocean University, Keelung 202301, Taiwan;
| | - Chung-Chu Chen
- Department of Internal Medicine, MacKay Memorial Hospital, Hsinchu 30013, Taiwan;
- Teaching Center of Natural Science, Minghsin University of Science and Technology, Hsinchu 202301, Taiwan
| | - Bor-Luen Chiang
- Department of Pediatrics, National Taiwan University Children Hospital, Taipei 100226, Taiwan; (C.-Y.L.); (B.-L.C.); (L.-M.H.)
| | - Li-Min Huang
- Department of Pediatrics, National Taiwan University Children Hospital, Taipei 100226, Taiwan; (C.-Y.L.); (B.-L.C.); (L.-M.H.)
| | - Suh-Chin Wu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan; (N.T.); (T.-H.C.)
- Department of Medical Science, National Tsing Hua University, Hsinchu 30013, Taiwan
- Correspondence:
| |
Collapse
|
6
|
Highly Pathogenic Avian Influenza H5 Hemagglutinin Fused with the A Subunit of Type IIb Escherichia coli Heat Labile Enterotoxin Elicited Protective Immunity and Neutralization by Intranasal Immunization in Mouse and Chicken Models. Vaccines (Basel) 2019; 7:vaccines7040193. [PMID: 31766677 PMCID: PMC6963717 DOI: 10.3390/vaccines7040193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 02/03/2023] Open
Abstract
Highly pathogenic avian influenza viruses are classified by the World Organization for Animal Health (OIE) as causes of devastating avian diseases. This study aimed to develop type IIb Escherichiacoli heat-labile enterotoxin (LTIIb) as novel mucosal adjuvants for mucosal vaccine development. The fusion protein of H5 and LTIIb-A subunit was expressed and purified for mouse and chicken intranasal immunizations. Intranasal immunization with the H5-LTIIb-A fusion protein in mice elicited potent neutralizing antibodies in sera and bronchoalveolar lavage fluids, induced stronger Th1 and Th17 cellular responses in spleen and cervical lymph nodes, and improved protection against H5N1 influenza virus challenge. More interestingly, intranasal immunization with the H5-LTIIb-A fusion protein in chickens elicited high titers of IgY, IgA, hemagglutinin inhibition (HAI), and neutralizing antibodies in their antisera. This study employed the novel adjuvants of LTIIb for the development of a new generation of mucosal vaccines against highly pathogenic avian influenza viruses.
Collapse
|
7
|
Biuso F, Palladino L, Manenti A, Stanzani V, Lapini G, Gao J, Couzens L, Eichelberger MC, Montomoli E. Use of lentiviral pseudotypes as an alternative to reassortant or Triton X-100-treated wild-type Influenza viruses in the neuraminidase inhibition enzyme-linked lectin assay. Influenza Other Respir Viruses 2019; 13:504-516. [PMID: 31411006 PMCID: PMC6692537 DOI: 10.1111/irv.12669] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Formulation of neuraminidase (NA) within influenza vaccines is gaining importance in light of recent human studies. The enzyme-linked lectin assay (ELLA) is considered a reliable assay to evaluate human anti-NA antibodies. OBJECTIVES To overcome interference by hemagglutinin (HA)-specific antibodies and detect neuraminidase inhibitory (NI) antibodies only, two different sources of antigen have been studied in ELLA: reassortant viruses with a mismatched avian origin-HA or Triton X-100 (Tx)-treated wild-type viruses. Pseudotypes or pseudovirus (PV), characterized by a lentivirus core bearing human influenza NA and avian influenza HA, were investigated as an alternative source of antigen and compared to HA-mismatched and Tx-treated viruses, since represent a safer product to be handled. METHODS Two independent panels of sera were analyzed by ELLA to evaluate the anti-NA response against N1 (A/California/07/2009 (H1N1pdm)) and N2 (A/Hong Kong/4801/2014 (H3N2)). The NA inhibition (NI) antibody titers measured as either the 50% end point or 50% inhibitory concentration (IC50 ) were compared for every source of antigen. RESULTS The ELLA assay performed well with all three sources of antigen. NI titers measured using each antigen type correlated well when reported either as end point titers or as the IC50 . CONCLUSIONS This study suggests that HA-mismatched whole virus, Triton-treated wild-type virus or PV can be used to measure NI antibody titers of human sera, but further comparability/validation assays should be performed to assess statistical differences. The data support the use of PV as an attractive alternative source of antigen and justify further investigation to improve stability of this antigen source.
Collapse
Affiliation(s)
- Fabrizio Biuso
- VisMederi s.r.l., Strada del Petriccio e BelriguardoSienaItaly
- VisMederi Research s.r.l., Strada del Petriccio e BelriguardoSienaItaly
| | - Laura Palladino
- VisMederi s.r.l., Strada del Petriccio e BelriguardoSienaItaly
| | - Alessandro Manenti
- VisMederi s.r.l., Strada del Petriccio e BelriguardoSienaItaly
- VisMederi Research s.r.l., Strada del Petriccio e BelriguardoSienaItaly
| | | | - Giulia Lapini
- VisMederi s.r.l., Strada del Petriccio e BelriguardoSienaItaly
| | - Jin Gao
- Center for Biologics Evaluation and Research, Food and Drug AdministrationSilver SpringMDUSA
| | - Laura Couzens
- Center for Biologics Evaluation and Research, Food and Drug AdministrationSilver SpringMDUSA
| | - Maryna C. Eichelberger
- Center for Biologics Evaluation and Research, Food and Drug AdministrationSilver SpringMDUSA
| | - Emanuele Montomoli
- VisMederi s.r.l., Strada del Petriccio e BelriguardoSienaItaly
- VisMederi Research s.r.l., Strada del Petriccio e BelriguardoSienaItaly
- Department of Molecular and Developmental MedicineUniversity of SienaSienaItaly
| |
Collapse
|
8
|
Chen TH, Liu WC, Lin CY, Liu CC, Jan JT, Spearman M, Butler M, Wu SC. Glycan-masking hemagglutinin antigens from stable CHO cell clones for H5N1 avian influenza vaccine development. Biotechnol Bioeng 2018; 116:598-609. [PMID: 30080931 DOI: 10.1002/bit.26810] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/06/2018] [Accepted: 08/02/2018] [Indexed: 12/29/2022]
Abstract
Refocusing of B-cell responses can be achieved by preserving the overall fold of the antigen structure but selectively mutating the undesired antigenic sites with additional N-linked glycosylation motifs for glycan masking the vaccine antigen. We previously reported that glycan-masking recombinant H5 hemagglutinin (rH5HA) antigens on residues 83, 127, and 138 (g127 + g138 or g83 + g127 + 138 rH5HA) elicited broader neutralizing antibodies and protection against heterologous clades/subclades of high pathogenic avian influenza H5N1 viruses. In this study, we engineered the stably expressing Chinese hamster ovary (CHO) cell clones for producing the glycan-masking g127 + g138 and g83 + g127 + g138 rH5HA antigens. All of these glycan-masking rH5HA antigens produced in stable CHO cell clones were found to be mostly oligomeric structures. Only the immunization with the glycan-masking g127 + g138 but not g83 + g127 + g138 rH5HA antigens elicited more potent neutralizing antibody titers against four out of five heterologous clades/subclades of H5N1 viral strains. The increased neutralizing antibody titers against these heterologous viral strains were correlated with the increased amounts of stem-binding antibodies, only the glycan-masking g127 + g138 rH5HA antigens can translate into more protection against live viral challenges. The stable CHO cell line-produced glycan-masking g127 + g138 rH5HA can be used for H5N1 subunit vaccine development.
Collapse
Affiliation(s)
- Ting-Hsuan Chen
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Chun Liu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Ying Lin
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Chyi Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Jia-Tsrong Jan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Maureen Spearman
- Department of Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Michael Butler
- Department of Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Suh-Chin Wu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan.,Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
9
|
Delgadillo-Gutiérrez K, Ribas-Aparicio RM, Jiménez-Alberto A, Aparicio-Ozores G, Castelán-Vega JA. Stability of retroviral pseudotypes carrying the hemagglutinin of avian influenza viruses under various storage conditions. J Virol Methods 2018; 263:44-49. [PMID: 30347199 DOI: 10.1016/j.jviromet.2018.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 09/09/2018] [Accepted: 10/16/2018] [Indexed: 11/25/2022]
Abstract
Retroviral pseudotypes are broadly used as safe instruments to mimic the structure and surface of highly pathogenic viruses. They have been employed for the discovery of new drugs, as diagnostic tools in vaccine studies, and part of serological assays. Because of their widespread use in research and their potential as tools for quality control, it is important to know their shelf life, stability, and best storage conditions. In this study, we produced pseudotypes carrying the lacZ reporter gene and the hemagglutinin (HA) of avian influenza virus subtypes H5 and H7 to investigate their stability under various storage conditions. We produced pseudotypes with titers of approximately 106 RLU/mL, which decreased to 105-106 RLU/mL after short-term storage at 4 °C (up to 4 weeks). Stability was maintained after long-term storage at -20 °C (up to 12 months), even under storage variations such as freeze-thaw cycles. We conclude that, although the titers decreased by 1 log10 under the different storage conditions, the remaining titers can be readily applicable in many techniques, such as neutralization assays. These findings show that large quantities of retroviral pseudotypes can be safely stored for short- or long-term use, allowing standardization and reduced variation in assays involving retroviral pseudotypes.
Collapse
Affiliation(s)
- Karen Delgadillo-Gutiérrez
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Rosa María Ribas-Aparicio
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Alicia Jiménez-Alberto
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Gerardo Aparicio-Ozores
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Juan A Castelán-Vega
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Mexico City, Mexico.
| |
Collapse
|
10
|
Tian Y, Zhao H, Liu Q, Zhang C, Nie J, Huang W, Li C, Li X, Wang Y. Development of in vitro and in vivo neutralization assays based on the pseudotyped H7N9 virus. Sci Rep 2018; 8:8484. [PMID: 29855533 PMCID: PMC5981435 DOI: 10.1038/s41598-018-26822-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/15/2018] [Indexed: 11/08/2022] Open
Abstract
H7N9 viral infections pose a great threat to both animal and human health. This avian virus cannot be handled in level 2 biocontainment laboratories, substantially hindering evaluation of prophylactic vaccines and therapeutic agents. Here, we report a high-titer pseudoviral system with a bioluminescent reporter gene, enabling us to visually and quantitatively conduct analyses of virus replications in both tissue cultures and animals. For evaluation of immunogenicity of H7N9 vaccines, we developed an in vitro assay for neutralizing antibody measurement based on the pseudoviral system; results generated by the in vitro assay were found to be strongly correlated with those by either hemagglutination inhibition (HI) or micro-neutralization (MN) assay. Furthermore, we injected the viruses into Balb/c mice and observed dynamic distributions of the viruses in the animals, which provides an ideal imaging model for quantitative analyses of prophylactic and therapeutic monoclonal antibodies. Taken together, the pseudoviral systems reported here could be of great value for both in vitro and in vivo evaluations of vaccines and antiviral agents without the need of wild type H7N9 virus.
Collapse
Affiliation(s)
- Yabin Tian
- Division II of In Vitro Diagnostics for Infectious Diseases, National Institutes for Food and Drug Control, No. 2 Tiantanxili, Beijing, 100050, China
| | - Hui Zhao
- Division of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, No. 2 Tiantanxili, Beijing, 100050, China
| | - Qiang Liu
- Division of HIV/AIDS and Sexually-transmitted Virus Vaccines, National Institutes for Food and Drug Control, No. 2 Tiantanxili, Beijing, 100050, China
| | - Chuntao Zhang
- Division II of In Vitro Diagnostics for Infectious Diseases, National Institutes for Food and Drug Control, No. 2 Tiantanxili, Beijing, 100050, China
| | - Jianhui Nie
- Division of HIV/AIDS and Sexually-transmitted Virus Vaccines, National Institutes for Food and Drug Control, No. 2 Tiantanxili, Beijing, 100050, China
| | - Weijing Huang
- Division of HIV/AIDS and Sexually-transmitted Virus Vaccines, National Institutes for Food and Drug Control, No. 2 Tiantanxili, Beijing, 100050, China
| | - Changgui Li
- Division of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, No. 2 Tiantanxili, Beijing, 100050, China
| | - Xuguang Li
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Canada, Ottawa, On, K1A 0K9, Canada
| | - Youchun Wang
- Division of HIV/AIDS and Sexually-transmitted Virus Vaccines, National Institutes for Food and Drug Control, No. 2 Tiantanxili, Beijing, 100050, China.
| |
Collapse
|
11
|
Biuso F, Carnell G, Montomoli E, Temperton N. A Lentiviral Pseudotype ELLA for the Measurement of Antibodies Against Influenza Neuraminidase. Bio Protoc 2018. [DOI: 10.21769/bioprotoc.2936] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
12
|
CLEC5A-Mediated Enhancement of the Inflammatory Response in Myeloid Cells Contributes to Influenza Virus Pathogenicity In Vivo. J Virol 2016; 91:JVI.01813-16. [PMID: 27795434 PMCID: PMC5165214 DOI: 10.1128/jvi.01813-16] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 10/14/2016] [Indexed: 12/27/2022] Open
Abstract
Human infections with influenza viruses exhibit mild to severe clinical outcomes as a result of complex virus-host interactions. Induction of inflammatory mediators via pattern recognition receptors may dictate subsequent host responses for pathogen clearance and tissue damage. We identified that human C-type lectin domain family 5 member A (CLEC5A) interacts with the hemagglutinin protein of influenza viruses expressed on lentiviral pseudoparticles through lectin screening. Silencing CLEC5A gene expression, blocking influenza-CLEC5A interactions with anti-CLEC5A antibodies, or dampening CLEC5A-mediated signaling using a spleen tyrosine kinase inhibitor consistently reduced the levels of proinflammatory cytokines produced by human macrophages without affecting the replication of influenza A viruses of different subtypes. Infection of bone marrow-derived macrophages from CLEC5A-deficient mice showed reduced levels of tumor necrosis factor alpha (TNF-α) and IP-10 but elevated alpha interferon (IFN-α) compared to those of wild-type mice. The heightened type I IFN response in the macrophages of CLEC5A-deficient mice was associated with upregulated TLR3 mRNA after treatment with double-stranded RNA. Upon lethal challenges with a recombinant H5N1 virus, CLEC5A-deficient mice showed reduced levels of proinflammatory cytokines, decreased immune cell infiltration in the lungs, and improved survival compared to the wild-type mice, despite comparable viral loads noted throughout the course of infection. The survival difference was more prominent at a lower dose of inoculum. Our results suggest that CLEC5A-mediated enhancement of the inflammatory response in myeloid cells contributes to influenza pathogenicity in vivo and may be considered a therapeutic target in combination with effective antivirals. Well-orchestrated host responses together with effective viral clearance are critical for optimal clinical outcome after influenza infections.
IMPORTANCE Multiple pattern recognition receptors work in synergy to sense viral RNA or proteins synthesized during influenza replication and mediate host responses for viral control. Well-orchestrated host responses may help to maintain the inflammatory response to minimize tissue damage while inducing an effective adaptive immune response for viral clearance. We identified that CLEC5A, a C-type lectin receptor which has previously been reported to mediate flavivirus-induced inflammatory responses, enhanced induction of proinflammatory cytokines and chemokines in myeloid cells after influenza infections. CLEC5A-deficient mice infected with influenza virus showed reduced inflammation in the lungs and improved survival compared to that of the wild-type mice despite comparable viral loads. The survival difference was more prominent at a lower dose of inoculum. Collectively, our results suggest that dampening CLEC5A-mediated inflammatory responses in myeloid cells reduces immunopathogenesis after influenza infections.
Collapse
|
13
|
Wen L, Zheng Y, Jiang K, Zhang M, Kondengaden SM, Li S, Huang K, Li J, Song J, Wang PG. Two-Step Chemoenzymatic Detection of N-Acetylneuraminic Acid-α(2-3)-Galactose Glycans. J Am Chem Soc 2016; 138:11473-6. [PMID: 27554522 DOI: 10.1021/jacs.6b07132] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sialic acids are typically linked α(2-3) or α(2-6) to the galactose that located at the non-reducing terminal end of glycans, playing important but distinct roles in a variety of biological and pathological processes. However, details about their respective roles are still largely unknown due to the lack of an effective analytical technique. Herein, a two-step chemoenzymatic approach for the rapid and sensitive detection of N-acetylneuraminic acid-α(2-3)-galactose glycans is described.
Collapse
Affiliation(s)
| | | | - Kuan Jiang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University , Tianjin 300071, China
| | | | | | | | | | - Jing Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University , Tianjin 300071, China
| | | | - Peng George Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University , Tianjin 300071, China
| |
Collapse
|
14
|
Nogales A, Baker SF, Domm W, Martínez-Sobrido L. Development and applications of single-cycle infectious influenza A virus (sciIAV). Virus Res 2016; 216:26-40. [PMID: 26220478 PMCID: PMC4728073 DOI: 10.1016/j.virusres.2015.07.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 07/05/2015] [Accepted: 07/13/2015] [Indexed: 02/06/2023]
Abstract
The diverse host range, high transmissibility, and rapid evolution of influenza A viruses justify the importance of containing pathogenic viruses studied in the laboratory. Other than physically or mechanically changing influenza A virus containment procedures, modifying the virus to only replicate for a single round of infection similarly ensures safety and consequently decreases the level of biosafety containment required to study highly pathogenic members in the virus family. This biological containment is more ideal because it is less apt to computer, machine, or human error. With many necessary proteins that can be deleted, generation of single-cycle infectious influenza A viruses (sciIAV) can be achieved using a variety of approaches. Here, we review the recent burst in sciIAV generation and summarize the applications and findings on this important human pathogen using biocontained viral mimics.
Collapse
Affiliation(s)
- Aitor Nogales
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Steven F Baker
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - William Domm
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States.
| |
Collapse
|
15
|
Identification of Novel Fusion Inhibitors of Influenza A Virus by Chemical Genetics. J Virol 2015; 90:2690-701. [PMID: 26676787 DOI: 10.1128/jvi.02326-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/10/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED A previous screening of more than 50,000 compounds led to the identification of a pool of bioactive small molecules with inhibitory effect on the influenza A virus. One of these compounds, now widely known as nucleozin, is a small molecule that targets the influenza A virus nucleoprotein. Here we identify and characterize two structurally different novel fusion inhibitors of the influenza A virus group 1 hemagglutinin (HA), FA-583 and FA-617, with low nanomolar activities. Escape mutants that are highly resistant to each of these compounds were generated, and both were found to carry mutations localized in close proximity to the B-loop of the hemagglutinin 2 protein, which plays a crucial role in the virion-host cell fusion process. Recombinant virus, generated through reverse genetics, confirmed the resistance phenotype. In addition, the proposed binding pockets predicted by molecular docking studies are in accordance with the resistance-bearing mutation sites. We show through mechanistic studies that FA-583 and FA-617 act as fusion inhibitors by prohibiting the low-pH-induced conformational change of hemagglutinin. Our study has offered concrete biological and mechanistic explorations for the strategic development of novel fusion inhibitors of influenza A viruses. IMPORTANCE Here we report two structurally distinctive novel fusion inhibitors of influenza A virus that act by interfering with the structural change of HA at acidic pH, a process necessary for successful entry of the virus. Mutational and molecular docking studies have identified their binding pockets situated in close proximity to the B-loop region of hemagglutinin 2. The reduced sensitivity of FA-583- or FA-617-associated mutants to another compound suggests a close proximity and even partial overlap of their binding sites on hemagglutinin. Amino acid sequence alignments and crystal structure analyses of group 1 and group 2 hemagglutinins have shed light on the possible binding mode of these two compounds. This report offers new lead compounds for the design of fusion inhibitors for influenza A viruses and further shows that analysis by forward chemical genetics is a highly effective approach for the identification of novel compounds that can perturb the infectivity of viruses and to probe new druggable targets or druggable domains in various viruses.
Collapse
|
16
|
Li TW, Cheng SF, Tseng YT, Yang YC, Liu WC, Wang SC, Chou MJ, Lin YJ, Wang Y, Hsiao PW, Wu SC, Chang DK. Development of single-chain variable fragments (scFv) against influenza virus targeting hemagglutinin subunit 2 (HA2). Arch Virol 2015; 161:19-31. [PMID: 26446888 DOI: 10.1007/s00705-015-2625-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/23/2015] [Indexed: 11/26/2022]
Abstract
Influenza A viruses (IAV) are widespread in birds and domestic poultry, occasionally causing severe epidemics in humans and posing health threats. Hence, the need to develop a strategy for prophylaxis or therapy, such as a broadly neutralizing antibody against IAV, is urgent. In this study, single-chain variable fragment (scFv) phage display technology was used to select scFv fragments recognizing influenza envelope proteins. The Tomlinson I and J scFv phage display libraries were screened against the recombinant HA2 protein (rHA2) for three rounds. Only the third-round elution sample of the Tomlinson J library showed high binding affinity to rHA2, from which three clones (3JA18, 3JA62, and 3JA78) were chosen for preparative-scale production as soluble antibody by E. coli. The clone 3JA18 was selected for further tests due to its broad affinity for influenza H1N1, H3N2 and H5N1. Simulations of the scFv 3JA18-HA trimer complex revealed that the complementarity-determining region of the variable heavy chain (VH-CDR2) bound the stem region of HA. Neutralization assays using a peptide derived from VH-CDR2 also supported the simulation model. Both the selected antibody and its derived peptide were shown to suppress infection with H5N1 and H1N1 viruses, but not H3N2 viruses. The results also suggested that the scFvs selected from rHA2 could have neutralizing activity by interfering with the function of the HA stem region during virus entry into target cells.
Collapse
MESH Headings
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/immunology
- Antibody Specificity
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Immunoglobulin Fc Fragments/genetics
- Immunoglobulin Fc Fragments/immunology
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza, Human/immunology
- Influenza, Human/virology
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/immunology
Collapse
Affiliation(s)
- Tai-Wei Li
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | | | - Yen-Tzu Tseng
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Yu-Chih Yang
- Agricultural Biotech Research Center, Academia Sinica, Taipei, Taiwan
| | - Wen-Chun Liu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | | | - Mei-Ju Chou
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Yu-Jen Lin
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Yueh Wang
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Wen Hsiao
- Agricultural Biotech Research Center, Academia Sinica, Taipei, Taiwan
| | - Suh-Chin Wu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ding-Kwo Chang
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
17
|
Abstract
This article compares the treatment of living beings (unvaccinated chickens and infected cells) considered as sentinel devices in a farm and in a lab in Hong Kong. Sentinel devices are defined as living beings posted on a boundary from which they send signals of invisible threats. The ethnography looks at how they transform differences between ordinary lives and lives exposed, between good death and bad death, through the practices of those who feed them. In farms and labs exposed to Avian Influenza viruses, the logic of biosecurity intersects with a logic of care, blurring the distinction between self and other, friend and enemy through aesthetic judgments concerning what is a ‘good death’. Metabolism and immunity are redefined when sentinels are fed to produce clear signals of the mutations of viruses.
Collapse
|
18
|
Ferrara F, Molesti E, Temperton N. The application of pseudotypes to influenza pandemic preparedness. Future Virol 2015. [DOI: 10.2217/fvl.15.36] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
ABSTRACT Human and animal populations are constantly exposed to multiple influenza strains due to zoonotic spillover and rapid viral evolution driven by intrinsic error-prone replication and immunological pressure. In this context, antibody responses directed against the hemagglutinin protein on the surface of the virus are of importance since they have been shown to correlate with protective immunity. Serological techniques, detecting these responses, play a critical role in influenza pandemic preparedness in particular with regard to the measurement of vaccine immunogenicity. As the recent human pandemics (H1N1) and avian influenza outbreaks (H5 and H7) have demonstrated, there is an urgent need to be better prepared to assess the contribution of the antibody response to protection against newly emerged viruses and to evaluate the extent of pre-existing heterosubtypic immunity in populations. This review compares pseudotype-based assays with wild-type and virus-like particle virus assays and discusses their place in the pandemic preparedness against the influenza virus. It additionally addresses the state-of-the-art developments of pseudotype-based assays (chimeric hemagglutinins, multiplex and post-attachment) including the development and future deployment of assay kits and approaches toward standardization to both preclinical and clinical endpoints. Progress toward the development of an influenza pseudotype library for the purposes of pandemic preparedness is also outlined and discussed.
Collapse
Affiliation(s)
- Francesca Ferrara
- Viral Pseudotype Unit, School of Pharmacy, University of Kent, Chatham Maritime, Kent, ME4 4TB, UK
| | - Eleonora Molesti
- Viral Pseudotype Unit, School of Pharmacy, University of Kent, Chatham Maritime, Kent, ME4 4TB, UK
| | - Nigel Temperton
- Viral Pseudotype Unit, School of Pharmacy, University of Kent, Chatham Maritime, Kent, ME4 4TB, UK
| |
Collapse
|
19
|
Carnell GW, Ferrara F, Grehan K, Thompson CP, Temperton NJ. Pseudotype-based neutralization assays for influenza: a systematic analysis. Front Immunol 2015; 6:161. [PMID: 25972865 PMCID: PMC4413832 DOI: 10.3389/fimmu.2015.00161] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 03/25/2015] [Indexed: 12/02/2022] Open
Abstract
The use of vaccination against the influenza virus remains the most effective method of mitigating the significant morbidity and mortality caused by this virus. Antibodies elicited by currently licensed influenza vaccines are predominantly hemagglutination-inhibition (HI)-competent antibodies that target the globular head of hemagglutinin (HA) thus inhibiting influenza virus entry into target cells. These antibodies predominantly confer homosubtypic/strain specific protection and only rarely confer heterosubtypic protection. However, recent academia or pharma-led R&D toward the production of a “universal vaccine” has centered on the elicitation of antibodies directed against the stalk of the influenza HA that has been shown to confer broad protection across a range of different subtypes (H1–H16). The accurate and sensitive measurement of antibody responses elicited by these “next-generation” influenza vaccines is, however, hampered by the lack of sensitivity of the traditional influenza serological assays HI, single radial hemolysis, and microneutralization. Assays utilizing pseudotypes, chimeric viruses bearing influenza glycoproteins, have been shown to be highly efficient for the measurement of homosubtypic and heterosubtypic broadly neutralizing antibodies, making them ideal serological tools for the study of cross-protective responses against multiple influenza subtypes with pandemic potential. In this review, we will analyze and compare literature involving the production of influenza pseudotypes with particular emphasis on their use in serum antibody neutralization assays. This will enable us to establish the parameters required for optimization and propose a consensus protocol to be employed for the further deployment of these assays in influenza vaccine immunogenicity studies.
Collapse
Affiliation(s)
- George William Carnell
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK
| | - Francesca Ferrara
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK
| | - Keith Grehan
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK
| | - Craig Peter Thompson
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK ; Department of Zoology, University of Oxford , Oxford , UK ; The Jenner Institute Laboratories, University of Oxford , Oxford , UK
| | - Nigel James Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK
| |
Collapse
|
20
|
A viable recombinant rhabdovirus lacking its glycoprotein gene and expressing influenza virus hemagglutinin and neuraminidase is a potent influenza vaccine. J Virol 2014; 89:2820-30. [PMID: 25540378 DOI: 10.1128/jvi.03246-14] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED The emergence of novel influenza viruses that cause devastating human disease is an ongoing threat and serves as an impetus for the continued development of novel approaches to influenza vaccines. Influenza vaccine development has traditionally focused on producing humoral and/or cell-mediated immunity, often against the viral surface glycoproteins hemagglutinin (HA) and neuraminidase (NA). Here, we describe a new vaccine candidate that utilizes a replication-defective vesicular stomatitis virus (VSV) vector backbone that lacks the native G surface glycoprotein gene (VSVΔG). The expression of the H5 HA of an H5N1 highly pathogenic avian influenza virus (HPAIV), A/Vietnam/1203/04 (VN1203), and the NA of the mouse-adapted H1N1 influenza virus A/Puerto Rico/8/34 (PR8) in the VSVΔG vector restored the ability of the recombinant virus to replicate in cell culture, without the requirement for the addition of trypsin. We show here that this recombinant virus vaccine candidate was nonpathogenic in mice when given by either the intramuscular or intranasal route of immunization and that the in vivo replication of VSVΔG-H5N1 is profoundly attenuated. This recombinant virus also provided protection against lethal H5N1 infection after a single dose. This novel approach to vaccination against HPAIVs may be widely applicable to other emerging strains of influenza virus. IMPORTANCE Preparation for a potentially catastrophic influenza pandemic requires novel influenza vaccines that are safe, can be produced and administered quickly, and are effective, both soon after administration and for a long duration. We have created a new influenza vaccine that utilizes an attenuated vesicular stomatitis virus (VSV) vector, to deliver and express influenza virus proteins against which vaccinated animals develop potent antibody responses. The influenza virus hemagglutinin and neuraminidase proteins, expressed on the surface of VSV particles, allowed this vaccine to grow in cell culture and induced a potent antibody response in mice that was effective against infection with a lethal influenza virus. The mice showed no adverse reactions to the vaccine, and they were protected against an otherwise lethal influenza infection after only 14 days postvaccination and after as many as 140 days postvaccination. The ability to rapidly produce this safe and effective vaccine in cell culture is additionally advantageous.
Collapse
|
21
|
Sawoo O, Dublineau A, Batéjat C, Zhou P, Manuguerra JC, Leclercq I. Cleavage of hemagglutinin-bearing lentiviral pseudotypes and their use in the study of influenza virus persistence. PLoS One 2014; 9:e106192. [PMID: 25166303 PMCID: PMC4148439 DOI: 10.1371/journal.pone.0106192] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 07/28/2014] [Indexed: 12/30/2022] Open
Abstract
Influenza A viruses (IAVs) are a major cause of infectious respiratory human diseases and their transmission is dependent upon the environment. However, the role of environmental factors on IAV survival outside the host still raises many questions. In this study, we used lentiviral pseudotypes to study the influence of the hemagglutinin protein in IAV survival. High-titered and cleaved influenza-based lentiviral pseudoparticles, through the use of a combination of two proteases (HAT and TMPRSS2) were produced. Pseudoparticles bearing hemagglutinin proteins derived from different H1N1, H3N2 and H5N1 IAV strains were subjected to various environmental parameters over time and tested for viability through single-cycle infectivity assays. We showed that pseudotypes with different HAs have different persistence profiles in water as previously shown with IAVs. Our results also showed that pseudotypes derived from H1N1 pandemic virus survived longer than those derived from seasonal H1N1 virus from 1999, at high temperature and salinity, as previously shown with their viral counterparts. Similarly, increasing temperature and salinity had a negative effect on the survival of the H3N2 and H5N1 pseudotypes. These results showed that pseudotypes with the same lentiviral core, but which differ in their surface glycoproteins, survived differently outside the host, suggesting a role for the HA in virus stability.
Collapse
Affiliation(s)
- Olivier Sawoo
- Institut Pasteur, Environment and Infectious Risks Research and Expertise Unit, Laboratory for Urgent Response to Biological Threats, Paris, France
- University of Paris Diderot, Sorbonne Paris Cité (Cellule Pasteur), Paris, France
| | - Amélie Dublineau
- Institut Pasteur, Environment and Infectious Risks Research and Expertise Unit, Laboratory for Urgent Response to Biological Threats, Paris, France
| | - Christophe Batéjat
- Institut Pasteur, Environment and Infectious Risks Research and Expertise Unit, Laboratory for Urgent Response to Biological Threats, Paris, France
| | - Paul Zhou
- Institut Pasteur of Shanghai, Unit of Antiviral Immunity and Genetic Therapy, Chinese Academy of Sciences, Shanghai, China
| | - Jean-Claude Manuguerra
- Institut Pasteur, Environment and Infectious Risks Research and Expertise Unit, Laboratory for Urgent Response to Biological Threats, Paris, France
| | - India Leclercq
- Institut Pasteur, Environment and Infectious Risks Research and Expertise Unit, Laboratory for Urgent Response to Biological Threats, Paris, France
- University of Paris Diderot, Sorbonne Paris Cité (Cellule Pasteur), Paris, France
- * E-mail:
| |
Collapse
|
22
|
Multiplex evaluation of influenza neutralizing antibodies with potential applicability to in-field serological studies. J Immunol Res 2014; 2014:457932. [PMID: 25101305 PMCID: PMC4101955 DOI: 10.1155/2014/457932] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 05/22/2014] [Indexed: 12/26/2022] Open
Abstract
The increased number of outbreaks of H5 and H7 LPAI and HPAI viruses in poultry has major public and animal health implications. The continuous rapid evolution of these subtypes and the emergence of new variants influence the ability to undertake effective surveillance. Retroviral pseudotypes bearing influenza haemagglutinin (HA) and neuraminidase (NA) envelope glycoproteins represent a flexible platform for sensitive, readily standardized influenza serological assays. We describe a multiplex assay for the study of neutralizing antibodies that are directed against both influenza H5 and H7 HA. This assay permits the measurement of neutralizing antibody responses against two antigenically distinct HAs in the same serum/plasma sample thus increasing the amount and quality of serological data that can be acquired from valuable sera. Sera obtained from chickens vaccinated with a monovalent H5N2 vaccine, chickens vaccinated with a bivalent H7N1/H5N9 vaccine, or turkeys naturally infected with an H7N3 virus were evaluated in this assay and the results correlated strongly with data obtained by HI assay. We show that pseudotypes are highly stable under basic cold-chain storage conditions and following multiple rounds of freeze-thaw. We propose that this robust assay may have practical utility for in-field serosurveillance and vaccine studies in resource-limited regions worldwide.
Collapse
|
23
|
Wang B, Wang B, Liu P, Li T, Si W, Xiu J, Liu H. Package of NDV-pseudotyped HIV-Luc virus and its application in the neutralization assay for NDV infection. PLoS One 2014; 9:e99905. [PMID: 24937158 PMCID: PMC4061091 DOI: 10.1371/journal.pone.0099905] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 05/19/2014] [Indexed: 01/01/2023] Open
Abstract
Newcastle disease virus (NDV) is a member of the Paramyxovirinae subfamily and can infect most species of birds. It has been a great threat for the poultry industry all around the world. In this report, we successfully produced infectious pseudotyped pNL4-3-Luc-R−E− (HIV-Luc) viruses with the HN and F envelope proteins of NDV. Further investigation revealed the cytoplasmic domains of HN and F, especially HN, plays a significant role in the infection efficiency of these pseudotyped HIV-Luc viruses. Replacement of, or direct fusion to the cytoplasmic domain of the HN protein by that of vesicular stomatitis virus G (VSV-G) could greatly enhance or destroy the infective potential of HN and F-pseudotyped (NDV-pseudotyped) HIV-Luc virus. We further established a novel neutralization assay to evaluate neutralizing antibodies against NDV with the NDV-pseudotyped HIV-Luc viruses. Comparative neutralization data indicate that the results determined by using the NDV-pseudotyped HIV-Luc viruses are as reliable as those by the conventional virus-neutralization assay (VN test) with native NDV. Moreover, the results show that the novel neutralization assay is more sensitive than the VN test.
Collapse
Affiliation(s)
- Bin Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Bin Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Peixin Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Tao Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Wei Si
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jinsheng Xiu
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Henggui Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- * E-mail:
| |
Collapse
|
24
|
Cheresiz SV, Kononova AA, Razumova YV, Dubich TS, Chepurnov AA, Kushch AA, Davey R, Pokrovsky AG. A vesicular stomatitis pseudovirus expressing the surface glycoproteins of influenza A virus. Arch Virol 2014; 159:2651-8. [PMID: 24888312 DOI: 10.1007/s00705-014-2127-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 05/21/2014] [Indexed: 12/11/2022]
Abstract
Pseudotyped viruses bearing the glycoprotein(s) of a donor virus over the nucleocapsid core of a surrogate virus are widely used as safe substitutes for infectious virus in virology studies. Retroviral particles pseudotyped with influenza A virus glycoproteins have been used recently for the study of influenza hemagglutinin and neuraminidase-dependent processes. Here, we report the development of vesicular-stomatitis-virus-based pseudotypes bearing the glycoproteins of influenza A virus. We show that pseudotypes bearing the hemagglutinin and neuraminidase of H5N1 influenza A virus mimic the wild-type virus in neutralization assays and sensitivity to entry inhibitors. We demonstrate the requirement of NA for the infectivity of pseudotypes and show that viruses obtained with different NA proteins are significantly different in their transduction activities. Inhibition studies with oseltamivir carboxylate show that neuraminidase activity is required for pseudovirus production, but not for the infection of target cells with H5N1-VSV pseudovirus. The HA-NA-VSV pseudoviruses have high transduction titers and better stability than the previously reported retroviral pseudotypes and can replace live influenza virus in the development of neutralization assays, screening of potential antivirals, and the study of different HA/NA reassortants.
Collapse
Affiliation(s)
- S V Cheresiz
- Department of Medicine, Novosibirsk State University, Novosibirsk, Russia,
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Qiu C, Huang Y, Zhang A, Tian D, Wan Y, Zhang X, Zhang W, Zhang Z, Yuan Z, Hu Y, Zhang X, Xu J. Safe pseudovirus-based assay for neutralization antibodies against influenza A(H7N9) virus. Emerg Infect Dis 2014; 19:1685-7. [PMID: 24047684 PMCID: PMC3810762 DOI: 10.3201/eid1910.130728] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Serologic studies are urgently needed to assist in understanding an outbreak of influenza A(H7N9) virus. However, a biosafety level 3 laboratory is required for conventional serologic assays with live lethal virus. We describe a safe pseudovirus–based neutralization assay with preliminary assessment using subtype H7N9–infected samples and controls.
Collapse
|
26
|
Wallerström S, Lagerqvist N, Temperton NJ, Cassmer M, Moreno A, Karlsson M, Leijon M, Lundkvist A, Falk KI. Detection of antibodies against H5 and H7 strains in birds: evaluation of influenza pseudovirus particle neutralization tests. Infect Ecol Epidemiol 2014; 4:23011. [PMID: 24455106 PMCID: PMC3895261 DOI: 10.3402/iee.v4.23011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/02/2013] [Accepted: 12/13/2013] [Indexed: 01/16/2023] Open
Abstract
Introduction Avian influenza viruses circulate in bird populations, and it is important to maintain and uphold our knowledge of the viral strains that are currently of interest in this context. Here, we describe the use of hemagglutinin-pseudotype retroviruses based on highly pathogenic influenza viruses for the screening of avian sera for influenza A antibodies. Our aim was also to determine whether the pseudovirus neutralization tests that we assessed were sensitive and simple to use compared to the traditional methods, including hemagglutination inhibition assays and microneutralization tests. Material and methods H5 and H7 pseudovirus neutralization tests were evaluated by using serum from infected rabbits. Subsequently, the assays were further investigated using a panel of serum samples from avian species. The panel contained samples that were seropositive for five different hemagglutinin subtypes as well as influenza A seronegative samples. Results and discussion The results suggest that the pseudovirus neutralization test is an alternative to hemagglutination inhibition assays, as we observed comparable titers to those of both standard microneutralizations assays as well as hemagglutinin inhibition assays. When evaluated by a panel of avian sera, the method also showed its capability to recognize antibodies directed toward low-pathogenic H5 and H7. Hence, we conclude that it is possible to use pseudoviruses based on highly pathogenic avian influenza viruses to screen avian sera for antibodies directed against influenza A subtypes H5 and H7.
Collapse
Affiliation(s)
- Sofie Wallerström
- Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, Stockholm, Sweden ; Swedish Institute for Communicable Disease Control, Department of Diagnostics and Vaccinology, Solna, Sweden
| | - Nina Lagerqvist
- Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, Stockholm, Sweden ; Swedish Institute for Communicable Disease Control, Department of Diagnostics and Vaccinology, Solna, Sweden
| | | | - Michaela Cassmer
- Swedish Institute for Communicable Disease Control, Department of Diagnostics and Vaccinology, Solna, Sweden
| | - Ana Moreno
- Reparto di Virologia, Istituto Zooprofilattico Sperimentale della Lombardia ed Emilia Romagna, Brescia, Italy
| | - Malin Karlsson
- Swedish Institute for Communicable Disease Control, Department of Diagnostics and Vaccinology, Solna, Sweden
| | - Mikael Leijon
- National Veterinary Institute, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Ake Lundkvist
- Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, Stockholm, Sweden ; Swedish Institute for Communicable Disease Control, Department of Diagnostics and Vaccinology, Solna, Sweden ; Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Kerstin I Falk
- Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, Stockholm, Sweden ; Swedish Institute for Communicable Disease Control, Department of Diagnostics and Vaccinology, Solna, Sweden
| |
Collapse
|
27
|
Mather S, Scott S, Temperton N, Wright E, King B, Daly J. Current progress with serological assays for exotic emerging/re-emerging viruses. Future Virol 2013. [DOI: 10.2217/fvl.13.60] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Recent decades have witnessed an unprecedented rise in the outbreak occurrence of infectious and primarily zoonotic viruses. Contributing factors to this phenomenon include heightened global connectivity via air travel and international trade links, as well as man-made environmental alterations, such as deforestation and climate change, which all serve to bring humans into closer contact with animal reservoirs and alter the habitat of vectors, thus facilitating the transmission of viruses between species. Serological assays are integral to tracking the epidemiological spread of a virus and evaluating mass vaccination programs by quantifying neutralizing antibody responses raised against antigenic epitopes on the viral surface. However, conventional serological tests are somewhat marred by equipment and reagent costs, the necessity for high-containment laboratories for studying many emerging viruses, and interlaboratory variability, among other issues. This review details ‘next-generation’ assays aimed at addressing some of the persistent problems with viral serology, focusing on how manipulating the genomes of RNA viruses can produce attenuated or chimeric viruses that can be exploited as surrogate viruses in neutralization assays. Despite the undoubted promise of such novel serological platforms, it must be remembered that these assays have to withstand rigorous validation and standardization measures before they can play an integral role in curtailing the severity of future emerging virus outbreaks.
Collapse
Affiliation(s)
- Stuart Mather
- Viral Pseudotype Unit (Medway), School of Pharmacy, University of Kent, Chatham, Kent, ME4 4TB, UK
| | - Simon Scott
- Viral Pseudotype Unit (Medway), School of Pharmacy, University of Kent, Chatham, Kent, ME4 4TB, UK
| | - Nigel Temperton
- Viral Pseudotype Unit (Medway), School of Pharmacy, University of Kent, Chatham, Kent, ME4 4TB, UK
| | - Edward Wright
- Viral Pseudotype Unit (Fitzrovia), School of Life Sciences, University of Westminster, London, W1W 6UW, UK
| | - Barnabas King
- School of Veterinary Medicine & Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
| | - Janet Daly
- School of Veterinary Medicine & Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
| |
Collapse
|
28
|
Hidari KI, Yamaguchi M, Ueno F, Abe T, Yoshida K, Suzuki T. Influenza virus utilizes N-linked sialoglycans as receptors in A549 cells. Biochem Biophys Res Commun 2013; 436:394-9. [DOI: 10.1016/j.bbrc.2013.05.112] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 05/27/2013] [Indexed: 10/26/2022]
|
29
|
Rimmelzwaan GF, Katz JM. Immune responses to infection with H5N1 influenza virus. Virus Res 2013; 178:44-52. [PMID: 23735534 DOI: 10.1016/j.virusres.2013.05.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 04/03/2013] [Accepted: 05/20/2013] [Indexed: 01/22/2023]
Abstract
Influenza A H5N1 viruses remain a substantial threat to global public health. In particular, the expanding genetic diversity of H5N1 viruses and the associated risk for human adaptation underscore the importance of better understanding host immune responses that may protect against disease or infection. Although much emphasis has been placed on investigating early virus-host interactions and the induction of innate immune responses, little is known of the consequent adaptive immune response to H5N1 virus infection. In this review, we describe the H5N1 virus-specific and cross-reactive antibody and T cell responses in humans and animal models. Data from limited studies suggest that although initially robust, there is substantial waning of the serum antibody responses in survivors of H5N1 virus infection. Characterization of monoclonal antibodies generated from memory B cells of survivors of H5N1 virus infection has provided an understanding of the fine specificity of the human antibody response to H5N1 virus infection and identified strategies for immunotherapy. Human T cell responses induced by infection with seasonal influenza viruses are directed to relatively conserved internal proteins and cross-react with the H5N1 subtype. A role for T cell-based heterosubtypic immunity against H5N1 viruses is suggested in animal studies. Further studies on adaptive immune responses to H5N1 virus infection in both humans and animals are needed to inform the design of optimal immunological treatment and prevention modalities.
Collapse
Affiliation(s)
- Guus F Rimmelzwaan
- Viroscience Laboratory, Erasmus Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
30
|
Tao L, Chen J, Meng J, Chen Y, Li H, Liu Y, Zheng Z, Wang H. Enhanced protective efficacy of H5 subtype influenza vaccine with modification of the multibasic cleavage site of hemagglutinin in retroviral pseudotypes. Virol Sin 2013; 28:136-45. [PMID: 23728771 PMCID: PMC7091038 DOI: 10.1007/s12250-013-3326-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 04/19/2013] [Indexed: 11/02/2022] Open
Abstract
Traditionally, the multibasic cleavage site (MBCS) of surface protein H5-hemagglutinin (HA) is converted to a monobasic one so as to weaken the virulence of recombinant H5N1 influenza viruses and to produce inactivated and live attenuated vaccines. Whether such modification benefits new candidate vaccines has not been adequately investigated. We previously used retroviral vectors to generate wtH5N1 pseudotypes containing the wild-type HA (wtH5) from A/swine/Anhui/ca/2004 (H5N1) virus. Here, we generated mtH5N1 pseudotypes, which contained a mutant-type HA (mtH5) with a modified monobasic cleavage site. Groups of mice were subcutaneously injected with the two types of influenza pseudotypes. Compared to the group immunized with wtH5N1 pseudotypes, the inoculation of mtH5N1 pseudotypes induced significantly higher levels of HA specific IgG and IFN-γ in immunized mice, and enhanced protection against the challenge of mouse-adapted avian influenza virus A/Chicken/Henan/12/2004 (H5N1). This study suggests modification of the H5-hemagglutinin MBCS in retroviral pseudotypes enhances protection efficacy in mice and this information may be helpful for development of vaccines from mammalian cells to fight against H5N1 influenza viruses.
Collapse
Affiliation(s)
- Ling Tao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Development of a neutralization assay for influenza virus using an endpoint assessment based on quantitative reverse-transcription PCR. PLoS One 2013; 8:e56023. [PMID: 23437084 PMCID: PMC3577804 DOI: 10.1371/journal.pone.0056023] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Accepted: 01/09/2013] [Indexed: 12/15/2022] Open
Abstract
A microneutralization assay using an ELISA-based endpoint assessment (ELISA-MN) is widely used to measure the serological response to influenza virus infection and vaccination. We have developed an alternative microneutralization assay for influenza virus using a quantitative reverse transcription PCR-based endpoint assessment (qPCR-MN) in order to improve upon technical limitations associated with ELISA-MN. For qPCR-MN, infected MDCK-London cells in 96-well cell-culture plates are processed with minimal steps such that resulting samples are amenable to high-throughput analysis by downstream one-step quantitative reverse transcription PCR (qRT-PCR; SYBR Green chemistry with primers targeting a conserved region of the M1 gene of influenza A viruses). The growth curves of three recent vaccine strains demonstrated that the qRT-PCR signal detected at 6 hours post-infection reflected an amplification of at least 100-fold over input. Using ferret antisera, we have established the feasibility of measuring virus neutralization at 6 hours post-infection, a duration likely confined to a single virus-replication cycle. The neutralization titer for qPCR-MN was defined as the highest reciprocal serum dilution necessary to achieve a 90% inhibition of the qRT-PCR signal; this endpoint was found to be in agreement with ELISA-MN using the same critical reagents in each assay. qPCR-MN was robust with respect to assay duration (6 hours vs. 12 hours). In addition, qPCR-MN appeared to be compliant with the Percentage Law (i.e., virus neutralization results appear to be consistent over an input virus dose ranging from 500 to 12,000 TCID50). Compared with ELISA-MN, qPCR-MN might have inherent properties conducive to reducing intra- and inter-laboratory variability while affording suitability for automation and high-throughput uses. Finally, our qRT-PCR-based approach may be broadly applicable to the development of neutralization assays for a wide variety of viruses.
Collapse
|
32
|
Noisumdaeng P, Pooruk P, Kongchanagul A, Assanasen S, Kitphati R, Auewarakul P, Puthavathana P. Biological properties of H5 hemagglutinin expressed by vaccinia virus vector and its immunological reactivity with human sera. Viral Immunol 2013; 26:49-59. [PMID: 23374152 DOI: 10.1089/vim.2012.0055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
A recombinant vaccinia virus harboring the full length hemagglutinin (HA) gene derived from a highly pathogenic avian influenza A/Thailand/1(KAN-1)/2004 (H5N1) virus (rVac-H5 HA virus) was constructed. The immunogenicity of the expressed HA protein was characterized using goat antiserum, mouse monoclonal antibody, and human sera. The expressed HA protein localized both in the cytoplasm and on the cytoplasmic membrane of the thymidine kinase negative cells infected with the rVac-H5 HA virus, as determined by immunofluorescence assay. Western blot analysis demonstrated that the rVac-H5 HA protein was post-translationally processed by proteolytic cleavage of the HA0 precursor into HA1 and HA2 domains; and all of these HA forms were immunogenic in BALB/c mice. The molecular weight (MW) of each HA domain was the same as the wild-type H5 HA produced in Madin-Darby canine kidney cells infected with the H5N1 virus, but was higher than that expressed by a baculovirus-insect cell system. Sera from all H5N1 survivors reacted to HA0, HA1, and HA2 domains; whereas sera from H5N1-uninfected subjects reacted to the HA2 domain only, but not to HA0 or HA1, indicating that some cross-subtypic immunity exists in the general population. There was a lot-to-lot variation of the recombinant HA produced in the baculovirus-insect cell system that might affect the detection rate of antibody directed against certain HA domains.
Collapse
Affiliation(s)
- Pirom Noisumdaeng
- Department of Microbiology, Mahidol University, Bangkok-noi, Bangkok, Thailand
| | | | | | | | | | | | | |
Collapse
|
33
|
Millet JK, Kien F, Cheung CY, Siu YL, Chan WL, Li H, Leung HL, Jaume M, Bruzzone R, Malik Peiris JS, Altmeyer RM, Nal B. Ezrin interacts with the SARS coronavirus Spike protein and restrains infection at the entry stage. PLoS One 2012; 7:e49566. [PMID: 23185364 PMCID: PMC3504146 DOI: 10.1371/journal.pone.0049566] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 10/15/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Entry of Severe Acute Respiratory Syndrome coronavirus (SARS-CoV) and its envelope fusion with host cell membrane are controlled by a series of complex molecular mechanisms, largely dependent on the viral envelope glycoprotein Spike (S). There are still many unknowns on the implication of cellular factors that regulate the entry process. METHODOLOGY/PRINCIPAL FINDINGS We performed a yeast two-hybrid screen using as bait the carboxy-terminal endodomain of S, which faces the cytosol during and after opening of the fusion pore at early stages of the virus life cycle. Here we show that the ezrin membrane-actin linker interacts with S endodomain through the F1 lobe of its FERM domain and that both the eight carboxy-terminal amino-acids and a membrane-proximal cysteine cluster of S endodomain are important for this interaction in vitro. Interestingly, we found that ezrin is present at the site of entry of S-pseudotyped lentiviral particles in Vero E6 cells. Targeting ezrin function by small interfering RNA increased S-mediated entry of pseudotyped particles in epithelial cells. Furthermore, deletion of the eight carboxy-terminal amino acids of S enhanced S-pseudotyped particles infection. Expression of the ezrin dominant negative FERM domain enhanced cell susceptibility to infection by SARS-CoV and S-pseudotyped particles and potentiated S-dependent membrane fusion. CONCLUSIONS/SIGNIFICANCE Ezrin interacts with SARS-CoV S endodomain and limits virus entry and fusion. Our data present a novel mechanism involving a cellular factor in the regulation of S-dependent early events of infection.
Collapse
Affiliation(s)
- Jean Kaoru Millet
- HKU-Pasteur Research Centre, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
- Department of Anatomy, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - François Kien
- HKU-Pasteur Research Centre, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Chung-Yan Cheung
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Yu-Lam Siu
- HKU-Pasteur Research Centre, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Wing-Lim Chan
- HKU-Pasteur Research Centre, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
- Department of Pathology, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Huiying Li
- HKU-Pasteur Research Centre, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Hiu-Lan Leung
- HKU-Pasteur Research Centre, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Martial Jaume
- HKU-Pasteur Research Centre, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Roberto Bruzzone
- HKU-Pasteur Research Centre, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | - Joseph S. Malik Peiris
- HKU-Pasteur Research Centre, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | | | - Béatrice Nal
- HKU-Pasteur Research Centre, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
- Department of Anatomy, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
34
|
A single residue substitution in the receptor-binding domain of H5N1 hemagglutinin is critical for packaging into pseudotyped lentiviral particles. PLoS One 2012; 7:e43596. [PMID: 23133587 PMCID: PMC3487904 DOI: 10.1371/journal.pone.0043596] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 07/23/2012] [Indexed: 11/19/2022] Open
Abstract
Background Serological studies for influenza infection and vaccine response often involve microneutralization and hemagglutination inhibition assays to evaluate neutralizing antibodies against human and avian influenza viruses, including H5N1. We have previously characterized lentiviral particles pseudotyped with H5-HA (H5pp) and validated an H5pp-based assay as a safe alternative for high-throughput serological studies in BSL-2 facilities. Here we show that H5-HAs from different clades do not always give rise to efficient production of H5pp and the underlying mechanisms are addressed. Methodology/Findings We have carried out mutational analysis to delineate the molecular determinants responsible for efficient packaging of HA from A/Cambodia/40808/2005 (H5Cam) and A/Anhui/1/2005 (H5Anh) into H5pp. Our results demonstrate that a single A134V mutation in the 130-loop of the receptor binding domain is sufficient to render H5Anh the ability to generate H5Anh-pp efficiently, whereas the reverse V134A mutation greatly hampers production of H5Cam-pp. Although protein expression in total cell lysates is similar for H5Anh and H5Cam, cell surface expression of H5Cam is detected at a significantly higher level than that of H5Anh. We further demonstrate by several independent lines of evidence that the behaviour of H5Anh can be explained by a stronger binding to sialic acid receptors implicating residue 134. Conclusions We have identified a single A134V mutation as the molecular determinant in H5-HA for efficient incorporation into H5pp envelope and delineated the underlying mechanism. The reduced binding to sialic acid receptors as a result of the A134V mutation not only exerts a critical influence in pseudotyping efficiency of H5-HA, but has also an impact at the whole virus level. Because A134V substitution has been reported as a naturally occurring mutation in human host, our results may have implications for the understanding of human host adaptation of avian influenza H5N1 viruses.
Collapse
|
35
|
Geisler C, Jarvis DL. Effective glycoanalysis with Maackia amurensis lectins requires a clear understanding of their binding specificities. Glycobiology 2012; 21:988-93. [PMID: 21863598 DOI: 10.1093/glycob/cwr080] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
36
|
Desvaux S, Garcia J, Nguyen T, Reid S, Bui N, Roger F, Fenwick S, Peiris J, Ellis T. Evaluation of serological tests for H5N1 avian influenza on field samples from domestic poultry populations in Vietnam: Consequences for surveillance. Vet Microbiol 2012; 156:277-84. [DOI: 10.1016/j.vetmic.2011.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 11/09/2011] [Accepted: 11/15/2011] [Indexed: 11/25/2022]
|
37
|
Escors D, Kochan G, Stephenson H, Breckpot K. Cell and Tissue Gene Targeting with Lentiviral Vectors. SPRINGERBRIEFS IN BIOCHEMISTRY AND MOLECULAR BIOLOGY 2012. [PMCID: PMC7122860 DOI: 10.1007/978-3-0348-0402-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
One of the main advantages of using lentivectors is their capacity to transduce a wide range of cell types, independently from the cell cycle stage. However, transgene expression in certain cell types is sometimes not desirable, either because of toxicity, cell transformation, or induction of transgene-specific immune responses. In other cases, specific targeting of only cancerous cells within a tumor is sought after for the delivery of suicide genes. Consequently, great effort has been invested in developing strategies to control transgene delivery/expression in a cell/tissue-specific manner. These strategies can broadly be divided in three; particle pseudotyping (surface targeting), which entails modification of the envelope glycoprotein (ENV); transcriptional targeting, which utilizes cell-specific promoters and/or inducible promoters; and posttranscriptional targeting, recently applied in lentivectors by introducing sequence targets for cell-specific microRNAs. In this chapter we describe each of these strategies providing some illustrative examples.
Collapse
Affiliation(s)
- David Escors
- University College London, Rayne Building, 5 University Street, London, WC1E 6JF UK
| | - Grazyna Kochan
- Oxford Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building. Roosevelt Drive, Headington, Oxford, OX3 7DQ UK
| | - Holly Stephenson
- Institute of Child Health, University College London, Great Ormond Street, London, WC1N 3JH UK
| | | |
Collapse
|
38
|
Katz JM, Hancock K, Xu X. Serologic assays for influenza surveillance, diagnosis and vaccine evaluation. Expert Rev Anti Infect Ther 2011; 9:669-83. [PMID: 21692672 DOI: 10.1586/eri.11.51] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Serological techniques play a critical role in various aspects of influenza surveillance, vaccine development and evaluation, and sometimes in diagnosis, particularly for novel influenza virus infections of humans. Because individuals are repeatedly exposed to antigenically and genetically diverse influenza viruses over a lifetime, the gold standard for detection of a recent influenza virus infection or response to current vaccination is the demonstration of a seroconversion, a fourfold or greater rise in antibody titer relative to a baseline sample, to a circulating influenza strain or vaccine component. The hemagglutination-inhibition assay remains the most widely used assay to detect strain-specific serum antibodies to influenza. The hemagglutination-inhibition assay is also used to monitor antigenic changes among influenza viruses which are constantly evolving; such antigenic data is essential for consideration of changes in influenza vaccine composition. The use of the hemagglutinin-specific microneutralization assay has increased, in part, owing to its sensitivity for detection of human antibodies to novel influenza viruses of animal origin. Neutralization assays using replication-incompetent pseudotyped particles may be advantageous in some laboratory settings for detection of antibodies to influenza viruses with heightened biocontainment requirements. The use of standardized protocols and antibody standards are important steps to improve reproducibility and interlaboratory comparability of results of serologic assays for influenza viruses.
Collapse
Affiliation(s)
- Jacqueline M Katz
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA.
| | | | | |
Collapse
|
39
|
Elicitation of anti-1918 influenza virus immunity early in life prevents morbidity and lower levels of lung infection by 2009 pandemic H1N1 influenza virus in aged mice. J Virol 2011; 86:1500-13. [PMID: 22130546 DOI: 10.1128/jvi.06034-11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The Spanish influenza virus pandemic of 1918 was responsible for 40 million to 50 million deaths and is antigenically similar to the swine lineage 2009 pandemic influenza virus. Emergence of the 2009 pandemic from swine into humans has raised the possibility that low levels of cross-protective immunity to past shared epitopes could confer protection. In this study, influenza viruslike particles (VLPs) were engineered to express the hemagglutinin (HA) and genes from the 1918 influenza virus to evaluate the duration of cross-protection to the H1N1 pandemic strain by vaccinating young mice (8 to 12 weeks) and then allowing the animals to age to 20 months. This immunity was long lasting, with homologous receptor-blocking antibodies detected throughout the lifespan of vaccinated mice. Furthermore, the 1918 VLPs fully protected aged mice from 2009 pandemic H1N1 virus challenge 16 months after vaccination. Histopathological assessment showed that aged vaccinated mice had significant protection from alveolar infection but less protection of the bronchial tissue than adult vaccinated mice. Additionally, passive transfer of immune serum from aged vaccinated mice resulted in protection from death but not morbidity. This is the first report describing the lifelong duration of cross-reactive immune responses elicited by a 1918 VLP vaccine in a murine model. Importantly, these lifelong immune responses did not result in decreased total viral replication but did prevent infection of the lower respiratory tract. These findings show that immunity acquired early in life can restrict the anatomical location of influenza viral replication, rather than preventing infection, in the aged.
Collapse
|
40
|
Human annexin A6 interacts with influenza a virus protein M2 and negatively modulates infection. J Virol 2011; 86:1789-801. [PMID: 22114333 DOI: 10.1128/jvi.06003-11] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The influenza A virus M2 ion channel protein has the longest cytoplasmic tail (CT) among the three viral envelope proteins and is well conserved between different viral strains. It is accessible to the host cellular machinery after fusion with the endosomal membrane and during the trafficking, assembly, and budding processes. We hypothesized that identification of host cellular interactants of M2 CT could help us to better understand the molecular mechanisms regulating the M2-dependent stages of the virus life cycle. Using yeast two-hybrid screening with M2 CT as bait, a novel interaction with the human annexin A6 (AnxA6) protein was identified, and their physical interaction was confirmed by coimmunoprecipitation assay and a colocalization study of virus-infected human cells. We found that small interfering RNA (siRNA)-mediated knockdown of AnxA6 expression significantly increased virus production, while its overexpression could reduce the titer of virus progeny, suggesting a negative regulatory role for AnxA6 during influenza A virus infection. Further characterization revealed that AnxA6 depletion or overexpression had no effect on the early stages of the virus life cycle or on viral RNA replication but impaired the release of progeny virus, as suggested by delayed or defective budding events observed at the plasma membrane of virus-infected cells by transmission electron microscopy. Collectively, this work identifies AnxA6 as a novel cellular regulator that targets and impairs the virus budding and release stages of the influenza A virus life cycle.
Collapse
|
41
|
Jaume M, Yip MS, Cheung CY, Leung HL, Li PH, Kien F, Dutry I, Callendret B, Escriou N, Altmeyer R, Nal B, Daëron M, Bruzzone R, Peiris JSM. Anti-severe acute respiratory syndrome coronavirus spike antibodies trigger infection of human immune cells via a pH- and cysteine protease-independent FcγR pathway. J Virol 2011; 85:10582-97. [PMID: 21775467 PMCID: PMC3187504 DOI: 10.1128/jvi.00671-11] [Citation(s) in RCA: 248] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 07/05/2011] [Indexed: 11/20/2022] Open
Abstract
Public health measures successfully contained outbreaks of the severe acute respiratory syndrome coronavirus (SARS-CoV) infection. However, the precursor of the SARS-CoV remains in its natural bat reservoir, and reemergence of a human-adapted SARS-like coronavirus remains a plausible public health concern. Vaccination is a major strategy for containing resurgence of SARS in humans, and a number of vaccine candidates have been tested in experimental animal models. We previously reported that antibody elicited by a SARS-CoV vaccine candidate based on recombinant full-length Spike-protein trimers potentiated infection of human B cell lines despite eliciting in vivo a neutralizing and protective immune response in rodents. These observations prompted us to investigate the mechanisms underlying antibody-dependent enhancement (ADE) of SARS-CoV infection in vitro. We demonstrate here that anti-Spike immune serum, while inhibiting viral entry in a permissive cell line, potentiated infection of immune cells by SARS-CoV Spike-pseudotyped lentiviral particles, as well as replication-competent SARS coronavirus. Antibody-mediated infection was dependent on Fcγ receptor II but did not use the endosomal/lysosomal pathway utilized by angiotensin I converting enzyme 2 (ACE2), the accepted receptor for SARS-CoV. This suggests that ADE of SARS-CoV utilizes a novel cell entry mechanism into immune cells. Different SARS vaccine candidates elicit sera that differ in their capacity to induce ADE in immune cells despite their comparable potency to neutralize infection in ACE2-bearing cells. Our results suggest a novel mechanism by which SARS-CoV can enter target cells and illustrate the potential pitfalls associated with immunization against it. These findings should prompt further investigations into SARS pathogenesis.
Collapse
Affiliation(s)
- Martial Jaume
- HKU-Pasteur Research Centre, Dexter H. C. Man Building, 8 Sassoon Road, Pokfulam, Hong Kong SAR, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Recombinant trimeric HA protein immunogenicity of H5N1 avian influenza viruses and their combined use with inactivated or adenovirus vaccines. PLoS One 2011; 6:e20052. [PMID: 21655326 PMCID: PMC3104987 DOI: 10.1371/journal.pone.0020052] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 04/11/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The highly pathogenic avian influenza (HPAI) H5N1 virus continues to cause disease in poultry and humans. The hemagglutinin (HA) envelope protein is the primary target for subunit vaccine development. METHODOLOGY/PRINCIPAL FINDINGS We used baculovirus-insect cell expression to obtain trimeric recombinant HA (rHA) proteins from two HPAI H5N1 viruses. We investigated trimeric rHA protein immunogenicity in mice via immunizations, and found that the highest levels of neutralizing antibodies resulted from coupling with a PELC/CpG adjuvant. We also found that the combined use of trimeric rHA proteins with (a) an inactivated H5N1 vaccine virus, or (b) a recombinant adenovirus encoding full-length HA sequences for prime-boost immunization, further improved antibody responses against homologous and heterologous H5N1 virus strains. Data from cross-clade prime-boost immunization regimens indicate that sequential immunization with different clade HA antigens increased antibody responses in terms of total IgG level and neutralizing antibody titers. CONCLUSION/SIGNIFICANCE Our findings suggest that the use of trimeric rHA in prime-boost vaccine regimens represents an alternative strategy for recombinant H5N1 vaccine development.
Collapse
|
43
|
Xu K, Ling ZY, Sun L, Xu Y, Bian C, He Y, Lu W, Chen Z, Sun B. Broad humoral and cellular immunity elicited by a bivalent DNA vaccine encoding HA and NP genes from an H5N1 virus. Viral Immunol 2011; 24:45-56. [PMID: 21319978 DOI: 10.1089/vim.2010.0056] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Influenza A virus is highly variable and a major viral respiratory pathogen that can cause severe illness in humans. Therefore it is important to induce a sufficient immune response specific to current strains and to heterosubtypic viruses with vaccines. In this study, we developed a dual-promoter-based bivalent DNA vaccine that encodes both hemagglutinin (HA) and nucleoprotein (NP) proteins from a highly pathogenic A/Chicken/Henan/12/2004 (H5N1) virus. Our results show that the expression levels of HA and NP genes from the dual-promoter plasmid are similar to those seen when they are expressed individually in independent plasmids. When the bivalent DNA vaccine was inoculated via intramuscular injection and in vivo electroporation, high levels of both humoral and cellular immune responses were elicited against homologous H5N1 virus and heterosubtypic H9N2 virus. Furthermore, no obvious antigenic competition was observed between HA and NP proteins in the dual-promoter-based bivalent vaccine compared to monovalent vaccines. Our data suggest that a combination of influenza surface and internal viral genes in a dual-promoter-expressing plasmid may provide a new approach for developing a DNA vaccine that may protect not only specifically against a currently circulating strain, but also may cross-protect broadly against new heterosubtypic viruses.
Collapse
Affiliation(s)
- Ke Xu
- Molecular Virus Unit, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Giles BM, Ross TM. A computationally optimized broadly reactive antigen (COBRA) based H5N1 VLP vaccine elicits broadly reactive antibodies in mice and ferrets. Vaccine 2011; 29:3043-54. [PMID: 21320540 PMCID: PMC3090662 DOI: 10.1016/j.vaccine.2011.01.100] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 01/25/2011] [Accepted: 01/29/2011] [Indexed: 12/11/2022]
Abstract
Pandemic outbreaks of influenza are caused by the emergence of a pathogenic and transmissible virus to which the human population is immunologically naïve. Recent outbreaks of highly pathogenic avian influenza (HPAI) of the H5N1 subtype are of particular concern because of the high mortality rate (60% case fatality rate) and novel subtype. In order to develop a vaccine that elicits broadly reactive antibody responses against emerging H5N1 isolates, we utilized a novel antigen design technique termed computationally optimized broadly reactive antigen (COBRA). The COBRA HA sequence was based upon HA amino acid sequences from clade 2 H5N1 human infections and the expressed protein retained the ability to bind the receptor, as well as mediate particle fusion. Non-infectious recombinant VLP vaccines using the COBRA HA were purified from a mammalian expression system. Mice and ferrets vaccinated with COBRA HA H5N1 VLPs had protective levels of HAI antibodies to a representative isolates from each subclade of clade 2. Furthermore, VLP vaccinated animals were completely protected from a lethal challenge of the clade 2.2 H5N1 virus A/Whooper Swan/Mongolia/244/2005. This is the first report describing the use of COBRA-based antigen design. The COBRA HA H5N1 VLP vaccine elicited broadly reactive antibodies and is an effective influenza vaccine against HPAI virus.
Collapse
Affiliation(s)
- Brendan M. Giles
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ted M. Ross
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
45
|
Rimmelzwaan GF, Joyce Verburgh R, Nieuwkoop NJ, Bestebroer TM, Fouchier RA, Osterhaus AD. Use of GFP-expressing influenza viruses for the detection of influenza virus A/H5N1 neutralizing antibodies. Vaccine 2011; 29:3424-30. [DOI: 10.1016/j.vaccine.2011.02.082] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 02/16/2011] [Accepted: 02/24/2011] [Indexed: 11/29/2022]
|
46
|
Garcia JM, Lai JCC. Production of influenza pseudotyped lentiviral particles and their use in influenza research and diagnosis: an update. Expert Rev Anti Infect Ther 2011; 9:443-55. [PMID: 21504401 DOI: 10.1586/eri.11.25] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pseudotyped viral particles are being used as safe surrogates to mimic the structure and surface of many viruses, including highly pathogenic viruses such as avian influenza H5N1, to investigate biological functions mediated by the envelope proteins derived from these viruses. The first part of this article evaluates and discusses the differences in the production and characterization of influenza pseudoparticles. The second part focuses on the applications that such a flexible tool can provide in modern influenza research, in particular in the fields of drug discovery, molecular biology and diagnosis.
Collapse
Affiliation(s)
- Jean-Michel Garcia
- HKU-Pasteur Research Centre, Dexter HC Man Building, 8 Sassoon Road, Pokfulam, Hong Kong.
| | | |
Collapse
|
47
|
Wang W, Castelán-Vega JA, Jiménez-Alberto A, Vassell R, Ye Z, Weiss CD. A mutation in the receptor binding site enhances infectivity of 2009 H1N1 influenza hemagglutinin pseudotypes without changing antigenicity. Virology 2010; 407:374-80. [DOI: 10.1016/j.virol.2010.08.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 07/20/2010] [Accepted: 08/23/2010] [Indexed: 10/19/2022]
|
48
|
Generation of replication-competent recombinant influenza A viruses carrying a reporter gene harbored in the neuraminidase segment. J Virol 2010; 84:12075-81. [PMID: 20826692 DOI: 10.1128/jvi.00046-10] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Replication-competent influenza viruses carrying reporter genes are of great use for basic research, screening of antiviral drugs, and neutralizing of antibodies. In this study, two recombinant influenza A viruses with a neuraminidase (NA) segment harboring enhanced green fluorescent protein (EGFP) in the background of A/PR/8/34 (PR8) were generated. The viral RNA (vRNA)-specific packaging signals for NA were largely retained for efficient packaging. An "autocleave" 2A peptide sequence, which was inserted at the N terminus or the COOH terminus of NA to link with EGFP, enabled NA and EGFP to be expressed monocistronically. Further analysis demonstrated that both viruses, named rPR8-EGFP+NA and rPR8-NA+EGPF, although with some characteristic differences in growth and EGFP expression, could replicate in noncomplementary cells and propagate to large quantities while maintaining genome stability after multiple passages in embryonated eggs. These replication-competent influenza viruses carrying reporter genes are a great addition to the tool set for developing antiviral therapeutics and vaccines and for in vivo studies of viral dissemination and pathogenicity.
Collapse
|
49
|
Abstract
Although influenza A viruses of avian origin have long been responsible for influenza pandemics, including the "Spanish flu" pandemic of 1918, human infections caused by avian subtypes of influenza A virus, most notably H5N1, have emerged since the 1990s (H5N1 in 1997; H9N2 in 1999; and H7N7 in 2003). The wide geographic distribution of influenza A H5N1 in avian species, and the number and severity of human infections are unprecedented. Together with the ongoing genetic evolution of this virus, these features make influenza A H5N1 a likely candidate for a future influenza pandemic. This article discusses the epidemiology, pathogenesis, and diagnosis of human infections caused by influenza A H5N1 virus.
Collapse
|
50
|
Cavailler P, Chu S, Ly S, Garcia JM, Ha DQ, Bergeri I, Som L, Ly S, Sok T, Vong S, Buchy P. Seroprevalence of anti-H5 antibody in rural Cambodia, 2007. J Clin Virol 2010; 48:123-6. [DOI: 10.1016/j.jcv.2010.02.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 02/21/2010] [Accepted: 02/26/2010] [Indexed: 10/19/2022]
|