1
|
Sun JM, Zhang WQ, Li YJ, Guo TK, Zhang RR, Yang YL, Zhao Y, Yu LJ, Shi CW, Yang GL, Huang HB, Jiang YL, Wang JZ, Cao X, Wang N, Zeng Y, Yang WT, Wang CF. Recombinant probiotic Escherichia coli delivers the polymeric protein of swine influenza virus for protection. Vet Microbiol 2025; 302:110401. [PMID: 39862797 DOI: 10.1016/j.vetmic.2025.110401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/12/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
Swine influenza virus invades the host through the respiratory mucosa, which severely restricts the development of the pig breeding industry. To construct monomeric and trimeric vaccines, we developed recombinant Escherichia coli Nissle 1917 (EcN) strains that express the receptor binding site (RBS) of the hemagglutinin (HA) antigen from H1N1 swine influenza virus. After the mucosal immunization of mice, we found that probiotics activated CD40 and CD86 in DCs and increased the levels of IL-4 and IFN-γ secretion by T cells. Furthermore, the probiotics improved the function of the mucosal immune system, increased the level of SIgA, level of IgG and number of B220+IgA+, and activated germinal center B cells. The challenge experiment revealed that the probiotics alleviated weight loss, reduced pathological injury to the lungs, and protected the mice from virus infection. We also observed that the serum neutralizing antibodies of immunized piglets significantly increased, which reduced the shedding frequency of swine influenza virus in the nose of the piglets and reduced the pathological damage by activating the T cell immune response in infected piglets. Thus, the constructed probiotics are promising candidates for effective non-traditional swine influenza vaccines.
Collapse
Affiliation(s)
- Jin-Mei Sun
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Wen-Qiang Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Yan-Jin Li
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Tian-Kui Guo
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Rong-Rong Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Yong-Lei Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Ying Zhao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Ling-Jiao Yu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Chun-Wei Shi
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Gui-Lian Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Hai-Bin Huang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Yan-Long Jiang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Jian-Zhong Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Wen-Tao Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China.
| | - Chun-Feng Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
2
|
He Y, Wang J, Chi L, Dong Y, Chen H, Meng X, Liao M, Luo Y, Fan H. Combination Adjuvants Enhance Recombinant H5 Hemagglutinin Vaccine Protection Against High-Dose Viral Challenge in Chickens. Vaccines (Basel) 2024; 12:1448. [PMID: 39772109 PMCID: PMC11680309 DOI: 10.3390/vaccines12121448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Recombinant avian influenza subunit vaccines often require adjuvants to enhance immune responses. This study aims to evaluate the immune-enhancing potential of seven combination adjuvants in specific pathogen-free (SPF) chickens. METHODS SPF chickens were vaccinated with combinations of ISA78VG and adjuvants, including Quil-A, CpG, and monophosphoryl lipid A (MPLA). Their immune responses were assessed using a vaccination and viral challenge protection model. RESULTS The combinations of ISA78VG with Quil-A, CpG&MPLA or CpG&Quil-A significantly enhanced antibody responses and provided cross-protection against the H5N8-20135 strain. The ISA78VG&MPLA and ISA78VG&CpG&MPLA combinations induced the stronger IFN-γ production, with CpG further amplifying the immune response. The ISA78VG&Quil-A formulation, in particular, stimulated rapid antibody responses, achieving a 100% seroconversion by day 14 and high titers of hemagglutination inhibition (HI) antibodies against both the recombinant HA antigen and the H5N6-20053 virus. CONCLUSIONS The ISA78VG&Quil-A combination is an ideal adjuvant for enhancing the immunogenicity of avian influenza rHA subunit vaccines, offering a promising strategy for H5 subtype vaccine development.
Collapse
Affiliation(s)
- Yanjuan He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.H.); (J.W.); (L.C.); (Y.D.); (H.C.); (X.M.); (M.L.)
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
| | - Jiaxin Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.H.); (J.W.); (L.C.); (Y.D.); (H.C.); (X.M.); (M.L.)
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
| | - Lanyan Chi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.H.); (J.W.); (L.C.); (Y.D.); (H.C.); (X.M.); (M.L.)
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
| | - Yajing Dong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.H.); (J.W.); (L.C.); (Y.D.); (H.C.); (X.M.); (M.L.)
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
| | - Huixin Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.H.); (J.W.); (L.C.); (Y.D.); (H.C.); (X.M.); (M.L.)
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
| | - Xiaocui Meng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.H.); (J.W.); (L.C.); (Y.D.); (H.C.); (X.M.); (M.L.)
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.H.); (J.W.); (L.C.); (Y.D.); (H.C.); (X.M.); (M.L.)
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.H.); (J.W.); (L.C.); (Y.D.); (H.C.); (X.M.); (M.L.)
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
| | - Huiying Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.H.); (J.W.); (L.C.); (Y.D.); (H.C.); (X.M.); (M.L.)
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
| |
Collapse
|
3
|
Peng B, Wang Y, Xie Y, Dong X, Liu W, Li D, Chen H. An overview of influenza A virus detection methods: from state-of-the-art of laboratories to point-of-care strategies. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:4496-4515. [PMID: 38946516 DOI: 10.1039/d4ay00508b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Influenza A virus (IAV), a common respiratory infectious pathogen, poses a significant risk to personal health and public health safety due to rapid mutation and wide host range. To better prevent and treat IAV, comprehensive measures are needed for early and rapid screening and detection of IAV. Although traditional laboratory-based techniques are accurate, they are often time-consuming and not always feasible in emergency or resource-limited areas. In contrast, emerging point-of-care strategies provide faster results but may compromise sensitivity and specificity. Here, this review critically evaluates various detection methods for IAV from established laboratory-based procedures to innovative rapid diagnosis. By analyzing the recent research progress, we aim to address significant gaps in understanding the effectiveness, practicality, and applicability of these methods in different scenarios, which could provide information for healthcare strategies, guide public health response measures, and ultimately strengthen patient care in the face of the ongoing threat of IAV. Through a detailed comparison of diagnostic models, this review can provide a reliable reference for rapid, accurate and efficient detection of IAV, and to contribute to the diagnosis, treatment, prevention, and control of IAV.
Collapse
Affiliation(s)
- Bin Peng
- Guangzhou Huashang Vocational College, Guangzhou, 510000, China
| | - Yaqi Wang
- Guangzhou Institute for Food Inspection, Guangzhou, 510000, China
| | - Yueliang Xie
- Guangdong Agriculture Industry Business Polytechnic College, Guangzhou, 510000, China
| | - Xiangyan Dong
- State Key Laboratory of Chemical Oncogenomics, Institute of Biomedical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China.
| | - Wen Liu
- Guangdong Agriculture Industry Business Polytechnic College, Guangzhou, 510000, China
| | - Dan Li
- College of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, China
| | - Hui Chen
- State Key Laboratory of Chemical Oncogenomics, Institute of Biomedical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China.
| |
Collapse
|
4
|
Alizadeh M, Raj S, Shojadoost B, Matsuyama-Kato A, Boodhoo N, Abdelaziz K, Sharif S. In ovo administration of retinoic acid enhances cell-mediated immune responses against an inactivated H9N2 avian influenza virus vaccine. Vaccine 2023; 41:7281-7289. [PMID: 37923694 DOI: 10.1016/j.vaccine.2023.10.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 10/15/2023] [Accepted: 10/22/2023] [Indexed: 11/07/2023]
Abstract
The H9N2 subtype avian influenza virus (AIV) is a low pathogenic AIV that infects avian species and lead to huge economical losses in the poultry industry. The unique immunomodulatory properties of Retinoic acid (RA), an active component of vitamin A, highlights its potential to enhance chicken's resistance to infectious diseases and perhaps vaccine-induced immunity. Therefore, the present study evaluated the effects of in ovo supplementation of RA on the immunogenicity and protective efficacy of an inactivated avian influenza virus vaccine. On embryonic day 18, eggs were inoculated with either 90 μmol RA/200 μL/egg or diluent into the amniotic sac. On days 7 and 21 post-hatch, birds were vaccinated with 15 μg of β-propiolactone (BPL) inactivated H9N2 virus via the intramuscular route. One group received BPL in combination with an adjuvant, while the other group received saline solution and served as a non-vaccinated control group. Serum samples were collected on days 7, 14, 21, 28, 35, and 42 post-primary vaccination (ppv) for antibody analysis. On day 24 ppv, spleens were collected, and splenocytes were isolated to analyze cytokine expression, interferon gamma (IFN-γ) production, and cell population. On day 28 ppv, birds in all groups were infected with H9N2 virus and oral and cloacal swabs were collected for TCID50 (50 % Tissue Culture Infectious Dose) assay up to day 7 post-infection. The results demonstrated that in ovo administration of RA did not significantly enhance the AIV vaccine-induced antibody response against H9N2 virus compared to the group that received the vaccine alone. However, RA supplementation enhanced the frequency of macrophages (KUL01+), expression of inflammatory cytokines and production of IFN-γ by splenocytes. In addition, RA administration reduced oral shedding of AIV on day 5 post-infection. In conclusion, these findings suggest that RA can be supplemented in ovo to enhance AIV vaccine efficacy against LPAIV.
Collapse
Affiliation(s)
- Mohammadali Alizadeh
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada.
| | - Sugandha Raj
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada.
| | | | - Ayumi Matsuyama-Kato
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada.
| | - Nitish Boodhoo
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada.
| | - Khaled Abdelaziz
- Animal and Veterinary Sciences Department, Clemson University, Clemson, SC 29634, USA.
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada.
| |
Collapse
|
5
|
Batty CJ, Pena ES, Amouzougan EA, Moore KM, Ainslie KM, Bachelder EM. Humoral Response to the Acetalated Dextran M2e Vaccine is Enhanced by Antigen Surface Conjugation. Bioconjug Chem 2023; 34:1447-1458. [PMID: 37458383 PMCID: PMC11654056 DOI: 10.1021/acs.bioconjchem.3c00223] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
The influenza A virus causes substantial morbidity and mortality worldwide every year and poses a constant threat of an emergent pandemic. Seasonal influenza vaccination strategies fail to provide complete protection against infection due to antigenic drift and shift. A universal vaccine targeting a conserved influenza epitope could substantially improve current vaccination strategies. The ectodomain of the matrix 2 protein (M2e) of influenza is a highly conserved epitope between virus strains but is also poorly immunogenic. Administration of M2e and the immunostimulatory stimulator of interferon genes (STING) agonist 3'3'-cyclic guanosine-adenosine monophosphate (cGAMP) encapsulated in microparticles made of acetalated dextran (Ace-DEX) has previously been shown to be effective for increasing the immunogenicity of M2e, primarily through T-cell-mediated responses. Here, the immunogenicity of Ace-DEX MPs delivering M2e was further improved by conjugating the M2e peptide to the particle surface in an effort to affect B-cell responses more directly. Conjugated or encapsulated M2e co-administered with Ace-DEX MPs containing cGAMP were used to vaccinate mice, and it was shown that two or three vaccinations could fully protect against a lethal influenza challenge, while only the surface-conjugated antigen constructs could provide some protection against lethal challenge with only one vaccination. Additionally, the use of a reducible linker augmented the T-cell response to the antigen. These results show the utility of conjugating M2e to the surface of a particle carrier to increase its immunogenicity for use as the antigen in a universal influenza vaccine.
Collapse
Affiliation(s)
- Cole J. Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Erik S. Pena
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, NC, USA
| | - Eva A. Amouzougan
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kathryn M. Moore
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, NC, USA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
Wu CY, Kao SE, Tseng YC, Lin YP, Hou JT, Wu LY, Chiu S, Ma CA, Hsiao PW, Hsiao J, Chen JR. Pilot-scale production of inactivated monoglycosylated split H 1N 1 influenza virus vaccine provides cross-strain protection against influenza viruses. Antiviral Res 2023; 216:105640. [PMID: 37263355 DOI: 10.1016/j.antiviral.2023.105640] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/03/2023]
Abstract
Influenza epidemics and pandemics caused by newly emerging virus strains highlight an urgent need to develop a universal vaccine against viruses. Previously, a monoglycosylated X-181mg vaccine demonstrated that the HA possessing a single N-acetylglucosamine at each N-glycosylation site is superior to confer broader protection in mice than conventional vaccines. However, the greatest challenge in conducting clinical trials is the need to develop robust manufacturing processes capable of producing vaccines at the pilot scale with the desired stability, potency, and efficacy. Whether the monoglycosylated virus vaccine platform can be applied to the new vaccine strain in a timely manner and whether the mass-produced vaccine has the proper immunogenicity to induce cross-protective immunity remains unclear. Here, we show that a pilot-scale manufacturing process produced a monoglycosylated A/Brisbane/02/2018(H1N1) virus vaccine (IVR-190mg) with a single glycan at each glycosylation site of HA and NA. Compared with the fully glycosylated virus vaccine (IVR-190fg), the IVR-190mg provided broader cross-protection in mice against a wide range of H1N1 variants. The enhanced antibody responses induced by IVR-190mg immunization include higher hemagglutination-inhibition titers, higher neutralization activity, more anti-HA head domain, more anti-HA stem antibodies, higher neuraminidase activity inhibition titers, and notably, higher antibody-dependent cellular cytotoxicity. Additionally, the IVR-190mg also induced a more balanced Th1/Th2 response and elicited broader splenic CD4+ and CD8+ T-cell responses than IVR-190fg. This study demonstrated that IVR-190mg produced using a pilot-scale manufacturing process elicits comprehensive cross-strain immune responses that have great potential to substantially mitigate the need for yearly reformulation of strain-specific inactivated vaccines.
Collapse
Affiliation(s)
| | - Shao-En Kao
- RuenHuei Biopharmaceuticals Inc. Taipei, Taiwan
| | | | - Yu-Po Lin
- RuenHuei Biopharmaceuticals Inc. Taipei, Taiwan
| | - Jen-Tzu Hou
- RuenHuei Biopharmaceuticals Inc. Taipei, Taiwan
| | - Li-Yang Wu
- RuenHuei Biopharmaceuticals Inc. Taipei, Taiwan
| | - Sharon Chiu
- RuenHuei Biopharmaceuticals Inc. Taipei, Taiwan
| | - Che Alex Ma
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Pei-Wen Hsiao
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Jane Hsiao
- RuenHuei Biopharmaceuticals Inc. Taipei, Taiwan; OPKO Health Inc. Miami, Florida, USA
| | | |
Collapse
|
7
|
Varghese PM, Kishore U, Rajkumari R. Innate and adaptive immune responses against Influenza A Virus: Immune evasion and vaccination strategies. Immunobiology 2022; 227:152279. [DOI: 10.1016/j.imbio.2022.152279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022]
|
8
|
A Deep Learning Approach for Predicting Antigenic Variation of Influenza A H3N2. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:9997669. [PMID: 34697557 PMCID: PMC8541863 DOI: 10.1155/2021/9997669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 09/07/2021] [Accepted: 09/22/2021] [Indexed: 11/17/2022]
Abstract
Modeling antigenic variation in influenza (flu) virus A H3N2 using amino acid sequences is a promising approach for improving the prediction accuracy of immune efficacy of vaccines and increasing the efficiency of vaccine screening. Antigenic drift and antigenic jump/shift, which arise from the accumulation of mutations with small or moderate effects and from a major, abrupt change with large effects on the surface antigen hemagglutinin (HA), respectively, are two types of antigenic variation that facilitate immune evasion of flu virus A and make it challenging to predict the antigenic properties of new viral strains. Despite considerable progress in modeling antigenic variation based on the amino acid sequences, few studies focus on the deep learning framework which could be most suitable to be applied to this task. Here, we propose a novel deep learning approach that incorporates a convolutional neural network (CNN) and bidirectional long-short-term memory (BLSTM) neural network to predict antigenic variation. In this approach, CNN extracts the complex local contexts of amino acids while the BLSTM neural network captures the long-distance sequence information. When compared to the existing methods, our deep learning approach achieves the overall highest prediction performance on the validation dataset, and more encouragingly, it achieves prediction agreements of 99.20% and 96.46% for the strains in the forthcoming year and in the next two years included in an existing set of chronological amino acid sequences, respectively. These results indicate that our deep learning approach is promising to be applied to antigenic variation prediction of flu virus A H3N2.
Collapse
|
9
|
Bull MB, Cohen CA, Leung NH, Valkenburg SA. Universally Immune: How Infection Permissive Next Generation Influenza Vaccines May Affect Population Immunity and Viral Spread. Viruses 2021; 13:1779. [PMID: 34578360 PMCID: PMC8472936 DOI: 10.3390/v13091779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 12/24/2022] Open
Abstract
Next generation influenza vaccines that target conserved epitopes are becoming a clinical reality but still have challenges to overcome. Universal next generation vaccines are considered a vital tool to combat future pandemic viruses and have the potential to vastly improve long-term protection against seasonal influenza viruses. Key vaccine strategies include HA-stem and T cell activating vaccines; however, they could have unintended effects for virus adaptation as they recognise the virus after cell entry and do not directly block infection. This may lead to immune pressure on residual viruses. The potential for immune escape is already evident, for both the HA stem and T cell epitopes, and mosaic approaches for pre-emptive immune priming may be needed to circumvent key variants. Live attenuated influenza vaccines have not been immunogenic enough to boost T cells in adults with established prior immunity. Therefore, viral vectors or peptide approaches are key to harnessing T cell responses. A plethora of viral vector vaccines and routes of administration may be needed for next generation vaccine strategies that require repeated long-term administration to overcome vector immunity and increase our arsenal against diverse influenza viruses.
Collapse
Affiliation(s)
- Maireid B. Bull
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, China; (M.B.B.); (C.A.C.)
| | - Carolyn A. Cohen
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, China; (M.B.B.); (C.A.C.)
| | - Nancy H.L. Leung
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Hong Kong, China;
| | - Sophie A. Valkenburg
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, China; (M.B.B.); (C.A.C.)
| |
Collapse
|
10
|
Venkataraman S, Hefferon K, Makhzoum A, Abouhaidar M. Combating Human Viral Diseases: Will Plant-Based Vaccines Be the Answer? Vaccines (Basel) 2021; 9:vaccines9070761. [PMID: 34358177 PMCID: PMC8310141 DOI: 10.3390/vaccines9070761] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 12/28/2022] Open
Abstract
Molecular pharming or the technology of application of plants and plant cell culture to manufacture high-value recombinant proteins has progressed a long way over the last three decades. Whether generated in transgenic plants by stable expression or in plant virus-based transient expression systems, biopharmaceuticals have been produced to combat several human viral diseases that have impacted the world in pandemic proportions. Plants have been variously employed in expressing a host of viral antigens as well as monoclonal antibodies. Many of these biopharmaceuticals have shown great promise in animal models and several of them have performed successfully in clinical trials. The current review elaborates the strategies and successes achieved in generating plant-derived vaccines to target several virus-induced health concerns including highly communicable infectious viral diseases. Importantly, plant-made biopharmaceuticals against hepatitis B virus (HBV), hepatitis C virus (HCV), the cancer-causing virus human papillomavirus (HPV), human immunodeficiency virus (HIV), influenza virus, zika virus, and the emerging respiratory virus, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) have been discussed. The use of plant virus-derived nanoparticles (VNPs) and virus-like particles (VLPs) in generating plant-based vaccines are extensively addressed. The review closes with a critical look at the caveats of plant-based molecular pharming and future prospects towards further advancements in this technology. The use of biopharmed viral vaccines in human medicine and as part of emergency response vaccines and therapeutics in humans looks promising for the near future.
Collapse
Affiliation(s)
- Srividhya Venkataraman
- Virology Laboratory, Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; (K.H.); (M.A.)
- Correspondence:
| | - Kathleen Hefferon
- Virology Laboratory, Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; (K.H.); (M.A.)
| | - Abdullah Makhzoum
- Department of Biological Sciences & Biotechnology, Botswana International University of Science & Technology, Palapye, Botswana;
| | - Mounir Abouhaidar
- Virology Laboratory, Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; (K.H.); (M.A.)
| |
Collapse
|
11
|
Mima KA, Katorkina EI, Katorkin SA, Tsybanov SZ, Malogolovkin AS. [In silico prediction of B- and T-cell epitopes in the CD2v protein of african swine fever virus (African swine fever virus, Asfivirus, Asfarviridae).]. Vopr Virusol 2021; 65:103-112. [PMID: 32515566 DOI: 10.36233/0507-4088-2020-65-2-103-112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 01/29/2020] [Indexed: 11/05/2022]
Abstract
INTRODUCTION African swine fever virus (ASF) is a large DNA virus that is the only member of the Asfarviridae family. The spread of the ASF virus in the territory of the Russian Federation, Eastern Europe and China indicates the ineffectiveness of existing methods of combating the disease and reinforces the urgent need to create effective vaccines. One of the most significant antigens required for the formation of immune protection against ASF is a serotype-specific CD2v protein. THE PURPOSE OF THE STUDY This study presents the results of immuno-informatics on the identification of B- and T-cell epitopes for the CD2v protein of the ASF virus using in silico prediction methods. MATERIAL AND METHODS The primary sequence of the CD2v protein of the ASFV virus strain Georgia 2007/1 (IDFR682468) was analyzed in silico by programs BCPred, NetCTLpan, VaxiJen, PVS and Epitope Conservancy Analysis. RESULTS Using the BCPred and VaxiJen programs, 4 major B-cell immunogenic epitopes were identified. Analysis of the secretory region of ASF virus CD2v protein in NetCTLpan revealed 5 T-cell epitopes from the 32nd to the 197th position of amino acids that cross-link from the 1st to the 13th allele of the MHC-I of pig Discussion. This study presents the results in silico prediction to identify B- and T-cell epitopes of ASF virus CD2v protein. The soluble region of the CD2v protein can be included in the recombinant polyepitope vaccine against African swine fever. CONCLUSION B- and T-cell epitopes in the secretory region of the CD2v protein (from 17 to 204 aa) of ASF virus were identified by in silico prediction. An analysis of the conservatism of the identified B- and T-cell epitopes allowed us to develop a map of the distribution of immune epitopes in the CD2v protein sequence.
Collapse
Affiliation(s)
- K A Mima
- Federal Research Center for Virology and Microbiology, Vladimir region, Volginskiy, 601125, Russia
| | - E I Katorkina
- Federal Research Center for Virology and Microbiology, Vladimir region, Volginskiy, 601125, Russia
| | - S A Katorkin
- Federal Research Center for Virology and Microbiology, Vladimir region, Volginskiy, 601125, Russia
| | - S Z Tsybanov
- Federal Research Center for Virology and Microbiology, Vladimir region, Volginskiy, 601125, Russia
| | - A S Malogolovkin
- Federal Research Center for Virology and Microbiology, Vladimir region, Volginskiy, 601125, Russia
| |
Collapse
|
12
|
Mara K, Dai M, Brice AM, Alexander MR, Tribolet L, Layton DS, Bean AGD. Investigating the Interaction between Negative Strand RNA Viruses and Their Hosts for Enhanced Vaccine Development and Production. Vaccines (Basel) 2021; 9:vaccines9010059. [PMID: 33477334 PMCID: PMC7830660 DOI: 10.3390/vaccines9010059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 01/13/2021] [Indexed: 11/30/2022] Open
Abstract
The current pandemic has highlighted the ever-increasing risk of human to human spread of zoonotic pathogens. A number of medically-relevant zoonotic pathogens are negative-strand RNA viruses (NSVs). NSVs are derived from different virus families. Examples like Ebola are known for causing severe symptoms and high mortality rates. Some, like influenza, are known for their ease of person-to-person transmission and lack of pre-existing immunity, enabling rapid spread across many countries around the globe. Containment of outbreaks of NSVs can be difficult owing to their unpredictability and the absence of effective control measures, such as vaccines and antiviral therapeutics. In addition, there remains a lack of essential knowledge of the host–pathogen response that are induced by NSVs, particularly of the immune responses that provide protection. Vaccines are the most effective method for preventing infectious diseases. In fact, in the event of a pandemic, appropriate vaccine design and speed of vaccine supply is the most critical factor in protecting the population, as vaccination is the only sustainable defense. Vaccines need to be safe, efficient, and cost-effective, which is influenced by our understanding of the host–pathogen interface. Additionally, some of the major challenges of vaccines are the establishment of a long-lasting immunity offering cross protection to emerging strains. Although many NSVs are controlled through immunisations, for some, vaccine design has failed or efficacy has proven unreliable. The key behind designing a successful vaccine is understanding the host–pathogen interaction and the host immune response towards NSVs. In this paper, we review the recent research in vaccine design against NSVs and explore the immune responses induced by these viruses. The generation of a robust and integrated approach to development capability and vaccine manufacture can collaboratively support the management of outbreaking NSV disease health risks.
Collapse
|
13
|
HA1 (Hemagglutinin) quantitation for influenza A H1N1 and H3N2 high yield reassortant vaccine candidate seed viruses by RP-UPLC. Vaccine 2020; 39:545-553. [PMID: 33341306 DOI: 10.1016/j.vaccine.2020.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/20/2020] [Accepted: 12/01/2020] [Indexed: 11/23/2022]
Abstract
The only effective measure to decrease morbidity and mortality caused by the influenza virus in the human population is worldwide vaccination. Vaccination produces neutralizing antibodies that target the HA1 subunit of the HA (hemagglutinin) protein and are strain specific. The effectiveness of new influenza vaccines are linked to two factors, the correct prediction of the circulating strains in the population in a particular season and the concentration of the HA1 protein in the vaccine formulation. With the advent of the licensing of quadrivalent vaccines, pharmaceutical manufacturers are under considerable pressure due to time constraints and dedicated resources to deliver 194-198 million doses (2020-2021 U.S. market) of vaccine. Considering the valuable resources needed to produce the influenza vaccine in a timely manner, the efficient quantitation of the HA1 protein (the main component in the influenza vaccine) is required. Currently the only method approved by regulatory agencies for quantitation of the HA antigen in vaccines is the single radial immunodiffusion assay (SRID), an antibody dependent assay that is not time efficient. Time efficient methods that are antibody independent e.g. reverse phase-high performance liquid chromatography (RP-HPLC) or size exclusion-HPLC (SE-HPLC) are available. An improved method implementing reverse phase-ultra performance liquid chromatography (RP-UPLC) has been developed to quantitate the HA1 protein antigen present in the high yield reassortant vaccine seed viruses from influenza A H1N1 and H3N2 subtypes harvested from inoculated embryonated chicken eggs. This method differentiates between high yield and lower yielding reassortants in order to select the best vaccine candidate seed virus with the highest growth 'in ovo'. This direct capability to monitor the HA1 concentration of potential reassortant seed viruses and to choose the best yielding HA influenza reassortant when faced with multiple viral seed candidates provides a major advantage on the industrial scale to the influenza vaccine process.
Collapse
|
14
|
Bhatta TR, Ryt-Hansen P, Nielsen JP, Larsen LE, Larsen I, Chamings A, Goecke NB, Alexandersen S. Infection Dynamics of Swine Influenza Virus in a Danish Pig Herd Reveals Recurrent Infections with Different Variants of the H1N2 Swine Influenza A Virus Subtype. Viruses 2020; 12:v12091013. [PMID: 32927910 PMCID: PMC7551734 DOI: 10.3390/v12091013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022] Open
Abstract
Influenza A virus (IAV) in swine, so-called swine influenza A virus (swIAV), causes respiratory illness in pigs around the globe. In Danish pig herds, a H1N2 subtype named H1N2dk is one of the main circulating swIAV. In this cohort study, the infection dynamic of swIAV was evaluated in a Danish pig herd by sampling and PCR testing of pigs from two weeks of age until slaughter at 22 weeks of age. In addition, next generation sequencing (NGS) was used to identify and characterize the complete genome of swIAV circulating in the herd, and to examine the antigenic variability in the antigenic sites of the virus hemagglutinin (HA) and neuraminidase (NA) proteins. Overall, 76.6% of the pigs became PCR positive for swIAV during the study, with the highest prevalence at four weeks of age. Detailed analysis of the virus sequences obtained showed that the majority of mutations occurred at antigenic sites in the HA and NA proteins of the virus. At least two different H1N2 variants were found to be circulating in the herd; one H1N2 variant was circulating at the sow and nursery sites, while another H1N2 variant was circulating at the finisher site. Furthermore, it was demonstrated that individual pigs had recurrent swIAV infections with the two different H1N2 variants, but re-infection with the same H1N2 variant was also observed. Better understandings of the epidemiology, genetic and antigenic diversity of swIAV may help to design better health interventions for the prevention and control of swIAV infections in the herds.
Collapse
Affiliation(s)
- Tarka Raj Bhatta
- Geelong Centre for Emerging Infectious Diseases, Geelong, VIC 3220, Australia;
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark; (P.R.-H.); (J.P.N.); (L.E.L.); (I.L.); (N.B.G.)
- Correspondence: (T.R.B.); (S.A.); Tel.: +61-0-452199095 (T.R.B.); +61-0-342159635 (S.A.)
| | - Pia Ryt-Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark; (P.R.-H.); (J.P.N.); (L.E.L.); (I.L.); (N.B.G.)
| | - Jens Peter Nielsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark; (P.R.-H.); (J.P.N.); (L.E.L.); (I.L.); (N.B.G.)
| | - Lars Erik Larsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark; (P.R.-H.); (J.P.N.); (L.E.L.); (I.L.); (N.B.G.)
| | - Inge Larsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark; (P.R.-H.); (J.P.N.); (L.E.L.); (I.L.); (N.B.G.)
| | - Anthony Chamings
- Geelong Centre for Emerging Infectious Diseases, Geelong, VIC 3220, Australia;
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Nicole B. Goecke
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark; (P.R.-H.); (J.P.N.); (L.E.L.); (I.L.); (N.B.G.)
- Division for Diagnostics & Scientific Advice, National Veterinary Institute, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Soren Alexandersen
- Geelong Centre for Emerging Infectious Diseases, Geelong, VIC 3220, Australia;
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Barwon Health, University Hospital Geelong, Geelong, VIC 3220, Australia
- Correspondence: (T.R.B.); (S.A.); Tel.: +61-0-452199095 (T.R.B.); +61-0-342159635 (S.A.)
| |
Collapse
|
15
|
Normal modes analysis and surface electrostatics of haemagglutinin proteins as fingerprints for high pathogenic type A influenza viruses. BMC Bioinformatics 2020; 21:354. [PMID: 32838732 PMCID: PMC7445075 DOI: 10.1186/s12859-020-03563-w] [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] [Indexed: 01/22/2023] Open
Abstract
Background Type A influenza viruses circulate and spread among wild birds and mostly consist of low pathogenic strains. However, fast genome variation timely results in the insurgence of high pathogenic strains, which when infecting poultry birds may cause a million deaths and strong commercial damage. More importantly, the host shift may concern these viruses and sustained human-to-human transmission may result in a dangerous pandemic outbreak. Therefore, fingerprints specific to either low or high pathogenic strains may represent a very important tool for global surveillance. Results We combined Normal Modes Analysis and surface electrostatic analysis of a mixed strain dataset of influenza A virus haemagglutinins from high and low pathogenic strains in order to infer specific fingerprints. Normal Modes Analysis sorted the strains in two different, homogeneous clusters; sorting was independent of clades and specific instead to high vs low pathogenicity. A deeper analysis of fluctuations and flexibility regions unveiled a special role for the 110-helix region. Specific sorting was confirmed by surface electrostatics analysis, which further allowed to focus on regions and mechanisms possibly crucial to the low-to-high transition. Conclusions Evidence from previous work demonstrated that changes in surface electrostatics are associated with the evolution and spreading of avian influenza A virus clades, and seemingly involved also in the avian to mammalian host shift. This work shows that a combination of electrostatics and Normal Modes Analysis can also identify fingerprints specific to high and low pathogenicity. The possibility to predict which specific mutations may result in a shift to high pathogenicity may help in surveillance and vaccine development.
Collapse
|
16
|
Cueno ME, Iguchi K, Suemitsu K, Hirano M, Hanzawa K, Isoda T, Ueno M, Iguchi R, Otani A, Imai K. Structural insights into the potential changes in receptor binding site found in the 1998-2018 influenza B/Yamagata hemagglutinin: A putative correlation between receptor binding site structural variability and seasonal infection. J Mol Graph Model 2020; 97:107580. [PMID: 32193088 DOI: 10.1016/j.jmgm.2020.107580] [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: 12/20/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/09/2022]
Abstract
Influenza B virus has two distinct lineages (Victoria and Yamagata) and are associated with seasonal influenza epidemics that cause respiratory illness. Influenza B hemagglutinin (HA) is a major surface glycoprotein with the receptor-binding site (RBS) primarily involved in viral pathogenesis. Generally, influenza B exclusively infects the human population which would insinuate that the structural variability of the influenza B HA RBS rarely changes. However, to our knowledge, the potential impact of variations in the influenza B HA RBS structural variability was not fully elucidated. Throughout this study, we generated models from the transitioning (evolving viral lineage) 1998-2018 influenza B/Yamagata HA, verified the quality of each HA model, performed HA RBS structural variability measurements, superimposed varying HA models for comparison, and designed a phylogenetic tree network for further analyses. We found that measurements of the transitioning HA RBS structural variability were generally maintained and, similarly, measurements of the altered (years that differed from the evolving viral lineage, specifically 2003, 2007, 2017) HA RBS structural variability differed from the transitioning HA RBS. Moreover, we observed that the altered HA RBS structural variability favored the formation of a putative Y202-H191 hydrogen bond which we postulate may increase structural stability, thereby, allowing for a winter infection of the virus. Furthermore, we established that changes in HA RBS structural variability does not influence viral evolution, but putatively seasonal infection.
Collapse
Affiliation(s)
- Marni E Cueno
- Department of Microbiology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan; Immersion Physics Class, Department of Science, Tokyo Gakugei University International Secondary School, Tokyo, 178-0063, Japan; Immersion Biology Class, Department of Science, Tokyo Gakugei University International Secondary School, Tokyo, 178-0063, Japan.
| | - Kanako Iguchi
- Immersion Physics Class, Department of Science, Tokyo Gakugei University International Secondary School, Tokyo, 178-0063, Japan
| | - Kanta Suemitsu
- Immersion Physics Class, Department of Science, Tokyo Gakugei University International Secondary School, Tokyo, 178-0063, Japan
| | - Marina Hirano
- Immersion Physics Class, Department of Science, Tokyo Gakugei University International Secondary School, Tokyo, 178-0063, Japan
| | - Kosei Hanzawa
- Immersion Physics Class, Department of Science, Tokyo Gakugei University International Secondary School, Tokyo, 178-0063, Japan
| | - Takemasa Isoda
- Immersion Biology Class, Department of Science, Tokyo Gakugei University International Secondary School, Tokyo, 178-0063, Japan
| | - Miu Ueno
- Immersion Biology Class, Department of Science, Tokyo Gakugei University International Secondary School, Tokyo, 178-0063, Japan
| | - Rinako Iguchi
- Immersion Biology Class, Department of Science, Tokyo Gakugei University International Secondary School, Tokyo, 178-0063, Japan
| | - Aoi Otani
- Immersion Biology Class, Department of Science, Tokyo Gakugei University International Secondary School, Tokyo, 178-0063, Japan
| | - Kenichi Imai
- Department of Microbiology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| |
Collapse
|
17
|
Ford-Siltz LA, Wales S, Tohma K, Gao Y, Parra GI. Genotype-Specific Neutralization of Norovirus Is Mediated by Antibodies Against the Protruding Domain of the Major Capsid Protein. J Infect Dis 2020; 225:1205-1214. [DOI: 10.1093/infdis/jiaa116] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/24/2020] [Indexed: 01/26/2023] Open
Abstract
Abstract
Human noroviruses are the most common viral agents of acute gastroenteritis. Recently, human intestinal enteroids were shown to be permissive for norovirus infection. We tested their suitability as a system to study norovirus neutralization. Hyperimmune sera raised against virus-like particles (VLPs) representing different genotypes showed highly specific neutralization activity against GII.4 and GII.6 noroviruses. Carbohydrate blocking assays and neutralization exhibited similar patterns in antibody responses. Notably, sera produced against chimeric VLPs that presented swapped structural shell and protruding (P) domains, from different genotypes showed that neutralization is primarily mediated by antibodies mapping to the P domain of the norovirus capsid protein. This study provides empirical information on the antigenic differences among genotypes as measured by neutralization, which could guide vaccine design.
Collapse
Affiliation(s)
- Lauren A Ford-Siltz
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Samantha Wales
- Center for Food Safety and Applied Nutrition, Food and Drug Administration, Laurel, Maryland, USA
| | - Kentaro Tohma
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yamei Gao
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gabriel I Parra
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
18
|
The Kaposi's Sarcoma-Associated Herpesvirus (KSHV) gH/gL Complex Is the Predominant Neutralizing Antigenic Determinant in KSHV-Infected Individuals. Viruses 2020; 12:v12030256. [PMID: 32111001 PMCID: PMC7150787 DOI: 10.3390/v12030256] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/14/2020] [Accepted: 02/23/2020] [Indexed: 12/24/2022] Open
Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi’s sarcoma (KS), one of the most prevalent cancers of people living with HIV/AIDS in sub-Saharan Africa. The seroprevalence for KSHV is high in the region, and no prophylactic vaccine against the virus is available. In this study, we characterized the antigenic targets of KSHV-specific neutralizing antibodies (nAbs) in asymptomatic KSHV-infected individuals and KS patients with high nAbs titers. We quantified the extent to which various KSHV envelope glycoproteins (gB, ORF28, ORF68, gH, gL, gM, gN and gpK8.1) adsorbed/removed KSHV-specific nAbs from the plasma of infected individuals. Our study revealed that plasma from a majority of KSHV neutralizers recognizes multiple viral glycoproteins. Moreover, the breadth of nAbs responses against these viral glycoproteins varies among endemic KS, epidemic KS and asymptomatic KSHV-infected individuals. Importantly, among the KSHV glycoproteins, the gH/gL complex, but neither gH nor gL alone, showed the highest adsorption of KSHV-specific nAbs. This activity was detected in 80% of the KSHV-infected individuals regardless of their KS status. The findings suggest that the gH/gL complex is the predominant antigenic determinant of KSHV-specific nAbs. Therefore, gH/gL is a potential target for development of KSHV prophylactic vaccines.
Collapse
|
19
|
Smatti MK, Nasrallah GK, Al Thani AA, Yassine HM. Measuring influenza hemagglutinin (HA) stem-specific antibody-dependent cellular cytotoxicity (ADCC) in human sera using novel stabilized stem nanoparticle probes. Vaccine 2019; 38:815-821. [PMID: 31735504 DOI: 10.1016/j.vaccine.2019.10.093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/26/2019] [Accepted: 10/29/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Generating vaccine that confers a complete protection is a major goal in designing a universal influenza vaccine. Currently, there is a considerable interest in the broadly neutralizing antibodies (bnAb) targeting the conserved HA stem region. These antibodies have been shown to activate cellular immune responses, such as ADCC, in addition to their neutralization activity. We had previously demonstrated that immunization with H1-based stabilized stem (SS) nanoparticles (np) protects against heterosubtypic lethal H5N1 challenge, despite the absence of detectable neutralizing activity. Utilizing these novel SS probes to develop an ADCC assay would help in understanding the mechanism of action of stem-specific antibodies, as well as evaluating future influenza vaccines. OBJECTIVES To develop a new protocol to assess the ADCC activity mediated by stem-directed antibodies in human sera using novel SS np probes. STUDY DESIGN Human sera samples were screened for binding and ADCC activities to different influenza group 1 SS probes (H1, H2, and H5) using trimeric SS or multivalent SS-np (n = 8 trimers) formats. RESULTS Initial screening revealed 63% (57/90) seroprevalence of anti-HA (H1) stem-epitope antibodies, as determined by the differential binding to HA SS and its corresponding epitope-mutant (Ile45Arg/Thr49Arg) probe. Using equimolar amounts, the multivalent presentation of HA SS on np induced significantly higher ADCC activity compared to the monovalent (trimer) SS probes (2-6 fold increase). Further, ADCC activity was similarly reported against different group 1 influenza subtypes: H1, H2, and H5. Importantly, ADCC was mediated mainly by antibodies targeting the bnAb-epitope on the HA stem. CONCLUSION We report on an assay to measure stem-specific ADCC activity using SS np probes. Our results indicate high prevalence of HA-stem antibodies with cross-reactive ADCC activity. Such assay could be utilized in the assessment of next generation influenza vaccines.
Collapse
Affiliation(s)
- Maria K Smatti
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Gheyath K Nasrallah
- Biomedical Research Center, Qatar University, Doha, Qatar; Biomedical Sciences Program, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Asmaa A Al Thani
- Biomedical Research Center, Qatar University, Doha, Qatar; Biomedical Sciences Program, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Hadi M Yassine
- Biomedical Research Center, Qatar University, Doha, Qatar; Biomedical Sciences Program, College of Health Sciences, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
20
|
Ning T, Nie J, Huang W, Li C, Li X, Liu Q, Zhao H, Wang Y. Antigenic Drift of Influenza A(H7N9) Virus Hemagglutinin. J Infect Dis 2019; 219:19-25. [PMID: 29982588 DOI: 10.1093/infdis/jiy408] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/28/2018] [Indexed: 01/03/2023] Open
Abstract
Background Since the emergence of influenza A(H7N9) virus in 2013, there have been 5 waves of influenza A(H7N9) epidemics in China. However, evolution of the hemagglutinin (HA) protein antigenicity has not been systematically investigated. Methods To better understand how antigenic drift in HA proteins of influenza (A)H7N9 virus occurs, 902 influenza A(H7N9) virus HA protein sequences from a public database were retrieved and analyzed. Fifty-three mutants with single amino acid substitutions in HA protein were introduced into pseudoviruses, and their antigenic characteristics were analyzed using pseudovirus-based assays. Results The frequencies of 9 mutations incrementally increased over the past 5 years, with mutations identified at multiple sites. While mean neutralization titers of most variants remained unchanged, 3 mutations, A143V, A143T, and R148K, displayed a median 4-fold lower susceptibility to neutralization by antisera against influenza A/Anhui/1/2013(H7N9) virus. Notably, A143V and A143T were located outside the previously reported antigenic sites. The most dominant variant (A143V/R148K) in the most recent season constituted 74.11% of all mutations and demonstrated a 10-fold reduction in its reactivity to influenza A/Anhui/1/2013(H7N9) virus antisera. Importantly, compared with the DNA construct without the corresponding HA protein mutation, DNA vaccine encoding the A143V/R148K mutant induced a 5-fold increase in the neutralizing activity against this circulating virus. Conclusions An appropriate vaccine strain should be considered in response to increasing antigenic drift in influenza A(H7N9) virus HA protein.
Collapse
Affiliation(s)
- Tingting Ning
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, Beijing, China.,Graduate School of Peking Union Medical College, Beijing, China
| | - Jianhui Nie
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, Beijing, China
| | - Weijin Huang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, Beijing, China
| | - Changgui Li
- Division of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Xuguang Li
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Canada, Ottawa, Canada
| | - Qiang Liu
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, Beijing, China
| | - Hui Zhao
- Division of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Youchun Wang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, Beijing, China.,Graduate School of Peking Union Medical College, Beijing, China
| |
Collapse
|
21
|
Mutsvunguma LZ, Rodriguez E, Escalante GM, Muniraju M, Williams JC, Warden C, Qin H, Wang J, Wu X, Barasa A, Mulama DH, Mwangi W, Ogembo JG. Identification of multiple potent neutralizing and non-neutralizing antibodies against Epstein-Barr virus gp350 protein with potential for clinical application and as reagents for mapping immunodominant epitopes. Virology 2019; 536:1-15. [PMID: 31377598 PMCID: PMC6733660 DOI: 10.1016/j.virol.2019.07.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/29/2019] [Accepted: 07/29/2019] [Indexed: 12/20/2022]
Abstract
Prevention of Epstein-Barr virus (EBV) infection has focused on generating neutralizing antibodies (nAbs) targeting the major envelope glycoprotein gp350/220 (gp350). In this study, we generated 23 hybridomas producing gp350-specific antibodies. We compared the candidate gp350-specific antibodies to the well-characterized nAb 72A1 by: (1) testing their ability to detect gp350 using enzyme-linked immunosorbent assay, flow cytometry, and immunoblot; (2) sequencing their heavy and light chain complementarity-determining regions (CDRs); (3) measuring the ability of each monoclonal antibody (mAb) to neutralize EBV infection in vitro; and (4) mapping the gp350 amino acids bound by the mAbs using competitive cell and linear peptide binding assays. We performed sequence analysis to identify 15 mAbs with CDR regions unique from those of murine 72A1 (m72A1). We observed antigen binding competition between biotinylated m72A1, serially diluted unlabeled gp350 nAbs (HB1, HB5, HB11, HB20), and our recently humanized 72A1, but not gp350 non-nAb (HB17) or anti-KSHV gH/gL antibody.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/isolation & purification
- Antibodies, Monoclonal/pharmacology
- Antibodies, Neutralizing/biosynthesis
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/isolation & purification
- Antibodies, Neutralizing/pharmacology
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/chemistry
- Antibodies, Viral/isolation & purification
- Antibodies, Viral/pharmacology
- B-Lymphocytes/immunology
- B-Lymphocytes/virology
- Binding Sites, Antibody
- Binding, Competitive
- Cell Line, Tumor
- Complementarity Determining Regions/chemistry
- Complementarity Determining Regions/immunology
- Enzyme-Linked Immunosorbent Assay
- Epithelial Cells/immunology
- Epithelial Cells/virology
- Epstein-Barr Virus Infections/immunology
- Epstein-Barr Virus Infections/prevention & control
- Epstein-Barr Virus Infections/virology
- Herpesvirus 4, Human/drug effects
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/immunology
- Humans
- Hybridomas/chemistry
- Hybridomas/immunology
- Immunodominant Epitopes/chemistry
- Immunodominant Epitopes/immunology
- Mice
- Protein Binding
- Sequence Alignment
- Sequence Homology, Amino Acid
- Viral Matrix Proteins/chemistry
- Viral Matrix Proteins/immunology
Collapse
Affiliation(s)
- Lorraine Z Mutsvunguma
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Esther Rodriguez
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Gabriela M Escalante
- Irell & Manella Graduate School of Biological Sciences of City of Hope, Duarte, CA, USA
| | - Murali Muniraju
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - John C Williams
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Charles Warden
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Hanjun Qin
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Jinhui Wang
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Anne Barasa
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, USA; Department of Human Pathology, University of Nairobi, Nairobi, Kenya
| | - David H Mulama
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, USA; Department of Biological Sciences, Masinde Muliro University of Science and Technology, Kakamega, Kenya
| | - Waithaka Mwangi
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Javier Gordon Ogembo
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, USA.
| |
Collapse
|
22
|
Gou X, Wu X, Shi Y, Zhang K, Huang J. A systematic review and meta-analysis of cross-reactivity of antibodies induced by H7 influenza vaccine. Hum Vaccin Immunother 2019; 16:286-294. [PMID: 31419167 PMCID: PMC7062429 DOI: 10.1080/21645515.2019.1649551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Inoculation with vaccine is the major intervention currently used to prevent influenza infections. However, it will be a challenge to produce and implement a new vaccine when a novel highly pathogenic influenza virus emerges in humans as significant infections. H7 subtype influenza viruses have similar epitopes on hemagglutinin, which can induce cross-reactive antibodies. In this study, a meta-analysis of the cross-reactivity of antibodies induced by one H7 subtype influenza vaccine against other H7 subtypes was performed. Database search was conducted in PubMed, Cochrane Library, EMBASE, MEDLINE, Chinese Biological Medicine Database (CBM), and Wanfang. A total of 9 articles comprising 811 human subjects were included in this meta-analysis. All assessed H7 influenza vaccines induced vaccine strain-specific protective antibodies [seroconversion rate (SCR) = 0.74, 95% CI (0.65, 0.82); seroprotection rate (SPR) = 0.81, 95% CI (0.78, 0.83)]. All H7 influenza virus monovalent vaccines exhibited cross-reactivity tested by hemagglutinin inhibition test (HI), microneutralization test (MN) and immunosorbent assay (ELISA) to other H7 subtype viruses. H7N1, H7N3, H7N7, and H7N9 vaccines elicited cross-reactive antibodies against other H7 subtype influenza viruses [SCR = 0.66, 95% CI (0.50, 0.82); SPR = 0.79, 95% CI (0.67, 0.91)]. The pooled SCR (95%CI) of cross-reactivity of H7N1 and H7N3 vaccines were 0.88 (0.85, 0.91) and 0.40 (0.26, 0.54), respectively. The consolidated SPR (95%CI) of H7N1 and H7N7 vaccines were 0.89 (0.86, 0.92) and 0.93 (0.81, 1.06). All H7 vaccines induced cross-reactive antibodies against H7N9 viruses [SCR = 0.69, 95% CI (0.52, 0.86); SPR = 0.85, 95% CI (0.76, 0.94)]. H7 vaccines can be used to limit influenza infection when a new highly pathogenic H7 virus appears.
Collapse
Affiliation(s)
- Xiaoqin Gou
- Medical Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaoxue Wu
- Medical Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yu Shi
- Medical Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ke Zhang
- Medical Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Junqiong Huang
- Medical Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
23
|
Mutations in Influenza A Virus Neuraminidase and Hemagglutinin Confer Resistance against a Broadly Neutralizing Hemagglutinin Stem Antibody. J Virol 2019; 93:JVI.01639-18. [PMID: 30381484 DOI: 10.1128/jvi.01639-18] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/22/2018] [Indexed: 11/20/2022] Open
Abstract
Influenza A virus (IAV), a major cause of human morbidity and mortality, continuously evolves in response to selective pressures. Stem-directed, broadly neutralizing antibodies (sBnAbs) targeting the influenza virus hemagglutinin (HA) are a promising therapeutic strategy, but neutralization escape mutants can develop. We used an integrated approach combining viral passaging, deep sequencing, and protein structural analyses to define escape mutations and mechanisms of neutralization escape in vitro for the F10 sBnAb. IAV was propagated with escalating concentrations of F10 over serial passages in cultured cells to select for escape mutations. Viral sequence analysis revealed three mutations in HA and one in neuraminidase (NA). Introduction of these specific mutations into IAV through reverse genetics confirmed their roles in resistance to F10. Structural analyses revealed that the selected HA mutations (S123G, N460S, and N203V) are away from the F10 epitope but may indirectly impact influenza virus receptor binding, endosomal fusion, or budding. The NA mutation E329K, which was previously identified to be associated with antibody escape, affects the active site of NA, highlighting the importance of the balance between HA and NA function for viral survival. Thus, whole-genome population sequencing enables the identification of viral resistance mutations responding to antibody-induced selective pressure.IMPORTANCE Influenza A virus is a public health threat for which currently available vaccines are not always effective. Broadly neutralizing antibodies that bind to the highly conserved stem region of the influenza virus hemagglutinin (HA) can neutralize many influenza virus strains. To understand how influenza virus can become resistant or escape such antibodies, we propagated influenza A virus in vitro with escalating concentrations of antibody and analyzed viral populations by whole-genome sequencing. We identified HA mutations near and distal to the antibody binding epitope that conferred resistance to antibody neutralization. Additionally, we identified a neuraminidase (NA) mutation that allowed the virus to grow in the presence of high concentrations of the antibody. Virus carrying dual mutations in HA and NA also grew under high antibody concentrations. We show that NA mutations mediate the escape of neutralization by antibodies against HA, highlighting the importance of a balance between HA and NA for optimal virus function.
Collapse
|
24
|
Sicca F, Neppelenbroek S, Huckriede A. Effector mechanisms of influenza-specific antibodies: neutralization and beyond. Expert Rev Vaccines 2018; 17:785-795. [PMID: 30145912 DOI: 10.1080/14760584.2018.1516553] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Antibodies directed against influenza virus execute their protective function by exploiting a variety of effector mechanisms. Neutralizing antibodies have been thoroughly studied because of their pivotal role in preventing influenza virus infection and their presence in host serum is correlated with protection. Influenza antibodies can also exploit non-neutralizing effector mechanisms, which until recently have been largely overlooked. AREAS COVERED Here, we discuss the antibody response to influenza virus in its entire breadth. Neutralizing antibodies mostly target variable epitopes on influenza surface proteins and interfere with virus binding, fusion, or egress. Non-neutralizing antibodies instead usually target conserved epitopes which can be located on surface as well as internal proteins. They drive viral clearance via interaction of their Fc region with components of the innate immune system such as immune effector cells (e.g. NK cells, macrophages) or the complement system. EXPERT COMMENTARY Recent research has unraveled that influenza-specific antibodies target multiple proteins and make use of diverse effector mechanisms. Often these antibodies are cross-reactive among virus strains of the same subtype or even between subtypes. As such they are induced early in life and are boosted by regular encounters with virus or vaccine. Designing strategies to optimally exploit these pre-existing antibodies may represent the key for the development of new broadly protective influenza vaccines.
Collapse
Affiliation(s)
- Federica Sicca
- a Department of Medical Microbiology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Sam Neppelenbroek
- a Department of Medical Microbiology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Anke Huckriede
- a Department of Medical Microbiology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| |
Collapse
|
25
|
Guo C, Zhang H, Xie X, Liu Y, Sun L, Li H, Yu P, Hu H, Sun J, Li Y, Feng Q, Zhao X, Liang D, Wang Z, Hu J. H1N1 influenza virus epitopes classified by monoclonal antibodies. Exp Ther Med 2018; 16:2001-2007. [PMID: 30186431 PMCID: PMC6122413 DOI: 10.3892/etm.2018.6429] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 06/22/2018] [Indexed: 12/26/2022] Open
Abstract
Epitopes serve an important role in influenza infection. It may be useful to screen universal influenza virus vaccines, analyzing the epitopes of multiple subtypes of the hemagglutinin (HA) protein. A total of 40 monoclonal antibodies (mAbs) previously obtained from flu virus HA antigens (development and characterization of 40 mAbs generated using H1N1 influenza virus split vaccines were previously published) were used to detect and classify mAbs into distinct flu virus sub-categories using the ELISA method. Following this, the common continuous amino acid sequences were identified by multiple sequence alignment analysis with the GenBank database and DNAMAN software, for use in predicting the epitopes of the HA protein. Synthesized peptides of these common sequences were prepared, and used to verify and determine the predicted linear epitopes through localization and distribution analyses. With these methods, nine HA linear epitopes distributed among different strains of influenza virus were identified, which included three from influenza A, four from 2009 H1N1 and seasonal influenza, and two from H1. The present study showed that considering a combination of the antigen-antibody reaction specificity, variation in the influenza virus HA protein and linear epitopes may present a useful approach for designing effective multi-epitope vaccines. Furthermore, the study aimed to clarify the cause and pathogenic mechanism of influenza virus HA-induced flu, and presents a novel idea for identifying the epitopes of other pathogenic microorganisms.
Collapse
Affiliation(s)
- Chunyan Guo
- Central Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Haixiang Zhang
- Central Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Xin Xie
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Yang Liu
- Central Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Lijun Sun
- Central Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Huijin Li
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Pengbo Yu
- Center of Shaanxi Provincial Disease Control and Prevention, Institute of Viral Diseases, Xi'an, Shaanxi 710052, P.R. China
| | - Hanyu Hu
- Central Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Jingying Sun
- Central Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Yuan Li
- Central Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Qing Feng
- Central Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Xiangrong Zhao
- Central Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Daoyan Liang
- Central Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Zhen Wang
- Central Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Jun Hu
- Central Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| |
Collapse
|
26
|
Astill J, Alkie T, Yitbarek A, Taha-Abdelaziz K, Bavananthasivam J, Nagy É, Petrik JJ, Sharif S. Examination of the effects of virus inactivation methods on the induction of antibody- and cell-mediated immune responses against whole inactivated H9N2 avian influenza virus vaccines in chickens. Vaccine 2018; 36:3908-3916. [PMID: 29853199 DOI: 10.1016/j.vaccine.2018.05.093] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/20/2018] [Accepted: 05/21/2018] [Indexed: 11/16/2022]
Abstract
Several types of avian influenza virus (AIV) vaccines exist, including live-attenuated, vectored, and whole inactivated virus (WIV) vaccines. Inactivated vaccines offer some advantages compared to other types of vaccines, including ease of production and lack of ability to revert to a virulent state. However, WIV are poorly immunogenic, especially when these vaccines are delivered to mucosal surfaces. There are several factors that contribute to the immunogenicity of vaccines, one of which is the method used to inactivate viruses. Several methods exist for producing influenza WIVs, including formaldehyde, a chemical that affects protein structures leading to virus inactivation. Other methods include treatment with beta-propiolactone (BPL) and the application of gamma radiation, both of which have less effects on protein structures compared to formaldehyde, and instead alter nucleic acids in the virion. Here, we sought to determine the effect of the above inactivation methods on immunogenicity of AIV vaccines. To this end, chickens were vaccinated with three different H9N2 WIVs using formaldehyde, BPL, and gamma radiation for inactivation. In addition to administering these three WIVs alone as vaccines, we also included CpG ODN 2007, a synthetic ligand recognized by Toll-like receptor (TLR)21 in chickens, as an adjuvant for each WIV. Subsequently, antibody- and cell-mediated immune responses were measured following vaccination. Antibody-mediated immune responses were increased in chickens that received the BPL and Gamma WIVs compared to the formaldehyde WIV. CpG ODN 2007 was found to significantly increase antibody responses for each WIV compared to WIV alone. Furthermore, we observed the presence of cell-mediated immune responses in chickens that received the BPL WIV combined with CpG ODN 2007. Based on these results, the BPL WIV + CpG ODN 2007 combination was the most effective vaccine at inducing adaptive immune responses against H9N2 AIV. Future studies should characterize mucosal adaptive immune responses to these vaccines.
Collapse
Affiliation(s)
- Jake Astill
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Tamiru Alkie
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; Department of Biology, Wilfred Laurier University, Waterloo, ON N2L 3C5, Canada(1)
| | - Alexander Yitbarek
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Khaled Taha-Abdelaziz
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; Pathology Department, Faculty of Veterinary Medicine, Beni-Suef University, Al Shamlah, 62511 Beni-Suef, Egypt
| | - Jegarubee Bavananthasivam
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Éva Nagy
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - James John Petrik
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
27
|
Alghamdi AN, Mahfouz ME, Hamdi FA, Al Aboud D, Al-Laylah TZ, Alotaibi MI, Al-Thomali KWA, Abdel-Moneim AS. Sequence analysis of haemagglutinin and neuraminidase of H1N1 strain from a patient coinfected with Mycobacterium tuberculosis. Mol Cell Probes 2017; 34:59-63. [PMID: 28499541 DOI: 10.1016/j.mcp.2017.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/20/2017] [Accepted: 05/08/2017] [Indexed: 12/01/2022]
Abstract
The 2009 H1N1 pandemic (H1N1pdm09) was associated with a considerable influenza-related morbidity and mortality. Among the complications, Mycobacterial tuberculosis was recorded as a coinfection with influenza in rare cases. The full-length sequences of the viral haemagglutinin and neuraminidase of H1N1pdm09 influenza A virus were analyzed from a recently infected patient. The patient was chronically infected with Mycobacterium tuberculosis. Molecular modelling and in-silico docking of the virus, and other selected strains with the drug oseltamivir were conducted and compared. Sequence analysis of the viral haemagglutinin revealed it to be closely related to the 6B.1 clade, with high identity to the circulating H1N1pdm09 strains, and confirmed that the virus still harbouring high affinity to the α-2,6-sialic acid human receptor. The viral neuraminidase showed high identity to the neuraminidase of the recently circulating strains of the virus with no evidence of the development of oseltamivir-resistant mutants. Regular monitoring of the circulating strains is recommended to screen for a possible emergence of drug-resistant strains.
Collapse
Affiliation(s)
- Ahmed N Alghamdi
- College of Medicine, Taif University, Al-Taif 21944, Saudi Arabia
| | - Mohammad E Mahfouz
- College of Medicine, Taif University, Al-Taif 21944, Saudi Arabia; King Faisal Hospital, Al-Taif, Saudi Arabia
| | | | | | | | | | | | - Ahmed S Abdel-Moneim
- College of Medicine, Taif University, Al-Taif 21944, Saudi Arabia; Virology Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef 62511, Egypt.
| |
Collapse
|
28
|
Łęga T, Weiher P, Obuchowski M, Nidzworski D. Presenting Influenza A M2e Antigen on Recombinant Spores of Bacillus subtilis. PLoS One 2016; 11:e0167225. [PMID: 27902762 PMCID: PMC5130239 DOI: 10.1371/journal.pone.0167225] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 11/10/2016] [Indexed: 11/19/2022] Open
Abstract
Effective vaccination against influenza virus infection is a serious problem mainly due to antigenic variability of the virus. Among many of investigated antigens, the extracellular domain of the M2 protein (M2e) features high homology in all strains of influenza A viruses and antibodies against M2e and is protective in animal models; this makes it a potential candidate for generation of a universal influenza vaccine. However, due to the low immunogenicity of the M2e, formulation of a vaccine based on this antigen requires some modification to induce effective immune responses. In this work we evaluated the possible use of Bacillus subtilis spores as a carrier of the Influenza A M2e antigen in mucosal vaccination. A tandem repeat of 4 consensus sequences coding for human-avian-swine-human M2e (M2eH-A-S-H) peptide was fused to spore coat proteins and stably exposed on the spore surface, as demonstrated by the immunostaining of intact, recombinant spores. Oral immunization of mice with recombinant endospores carrying M2eH-A-S-H elicited specific antibody production without the addition of adjuvants. Bacillus subtilis endospores can serve as influenza antigen carriers. Recombinant spores constructed in this work showed low immunogenicity although were able to induce antibody production. The System of influenza antigen administration presented in this work is attractive mainly due to the omitting time-consuming and cost-intensive immunogen production and purification. Therefore modification should be made to increase the immunogenicity of the presented system.
Collapse
Affiliation(s)
- Tomasz Łęga
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Paulina Weiher
- Department of Recombinant Vaccine, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Michał Obuchowski
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology UG-GUMed, Medical University of Gdańsk, Gdańsk, Poland
| | - Dawid Nidzworski
- Department of Recombinant Vaccine, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
29
|
Hsieh CF, Chen YL, Lin CF, Ho JY, Huang CH, Chiu CH, Hsieh PW, Horng JT. An extract from Taxodium distichum targets hemagglutinin- and neuraminidase-related activities of influenza virus in vitro. Sci Rep 2016; 6:36015. [PMID: 27796330 PMCID: PMC5086851 DOI: 10.1038/srep36015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 10/07/2016] [Indexed: 12/11/2022] Open
Abstract
Influenza virus remains an emerging virus and causes pandemics with high levels of fatality. After screening different plant extracts with potential anti-influenza activity, a water extract of Taxodium distichum stems (TDSWex) showed excellent activity against influenza viruses. The EC50 of TDSWex was 0.051 ± 0.024 mg/mL against influenza virus A/WSN/33. TDSWex had excellent antiviral efficacy against various strains of human influenza A and B viruses, particularly oseltamivir-resistant clinical isolates and a swine-origin influenza strain. We observed that the synthesis of viral RNA and protein were inhibited in the presence of TDSWex. The results of the time-of-addition assay suggested that TDSWex inhibited viral entry and budding. In the hemagglutination inhibition assay, TDSWex inhibited the hemagglutination of red blood cells, implying that the extract targeted hemagglutin-related functions such as viral entry. In the attachment and penetration assay, TDSWex showed antiviral activity with EC50s of 0.045 ± 0.026 and 0.012 ± 0.003 mg/mL, respectively. In addition, TDSWex blocked neuraminidase activity. We conclude that TDSWex has bimodal activities against both hemagglutinin and neuraminidase during viral replication.
Collapse
Affiliation(s)
- Chung-Fan Hsieh
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Li Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chwan-Fwu Lin
- Department of Cosmetic Science, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Jin-Yuan Ho
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Hsun Huang
- Department of Cosmetic Science, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Cheng-Hsun Chiu
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Pei-Wen Hsieh
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jim-Tong Horng
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.,Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
30
|
Antanasijevic A, Hafeman NJ, Tundup S, Kingsley C, Mishra RK, Rong L, Manicassamy B, Wardrop D, Caffrey M. Stabilization and Improvement of a Promising Influenza Antiviral: Making a PAIN PAINless. ACS Infect Dis 2016; 2:608-615. [PMID: 27759373 DOI: 10.1021/acsinfecdis.6b00046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The viral envelope protein hemagglutinin (HA) plays a critical role in influenza entry and thus is an attractive target for novel therapeutics. The small molecule tert-butylhydroquinone (TBHQ) has previously been shown to bind to HA and inhibit HA-mediated entry with low micromolar potency. However, enthusiasm for the use of TBHQ has diminished due to the compound's antioxidant properties. In this work we show that the antioxidant properties of TBHQ are not responsible for the inhibition of HA-mediated entry. In addition, we have performed a structure-activity relationship (SAR) analysis of TBHQ derivatives. We find that the most promising compound, 3-tert-butyl-4-methoxyphenol, exhibits enhanced potency (IC50 = 0.6 μM), decreased toxicity (CC50 = 340 μM), and increased stability (t1/2 > 48 h). Finally, we have characterized the binding properties of 3-tert-butyl-4-methoxyphenol using NMR and molecular dynamics to guide future efforts for chemical optimization.
Collapse
Affiliation(s)
- Aleksandar Antanasijevic
- Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago , 900 South Ashland Avenue, Chicago, Illinois 60607, United States
| | - Nicholas J Hafeman
- Department of Chemistry, University of Illinois at Chicago , 845 West Taylor Street, Chicago, Illinois 60607, United States
| | - Smanla Tundup
- Department of Microbiology and Immunology, University of Chicago , 920 East 58th Street, Chicago, Illinois 60637, United States
| | - Carolyn Kingsley
- Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago , 900 South Ashland Avenue, Chicago, Illinois 60607, United States
| | - Rama K Mishra
- Center for Molecular Innovation and Drug Discovery, Northwestern University , 2135 Sheridan Road, Evanston, Illinois 60208, United States
| | - Lijun Rong
- Department of Microbiology & Immunology, University of Illinois at Chicago , 835 South Wolcott, Chicago, Illinois 60612, United States
| | - Balaji Manicassamy
- Department of Microbiology and Immunology, University of Chicago , 920 East 58th Street, Chicago, Illinois 60637, United States
| | - Duncan Wardrop
- Department of Chemistry, University of Illinois at Chicago , 845 West Taylor Street, Chicago, Illinois 60607, United States
| | - Michael Caffrey
- Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago , 900 South Ashland Avenue, Chicago, Illinois 60607, United States
| |
Collapse
|
31
|
Shembekar N, Mallajosyula VVA, Malik A, Saini A, Varadarajan R, Gupta SK. Neutralization and Binding Profile of Monoclonal Antibodies Generated Against Influenza A H1N1 Viruses. Monoclon Antib Immunodiagn Immunother 2016; 35:191-8. [PMID: 27463230 DOI: 10.1089/mab.2016.0015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Monoclonal antibodies (MAbs) provide scope for the development of better therapeutics and diagnostic tools. Herein, we describe the binding and neutralization profile(s) for a panel of murine MAbs generated against influenza A H1N1 viruses elicited by immunization with pandemic H1 recombinant hemagglutinin (rHA)/whole virus or seasonal H1 rHA. Neutralizing MAbs, MA-2070 and MA-M, were obtained after pandemic A/California/07/2009 (H1N1) virus/rHA immunization(s). Both MAbs reacted specifically with rHA from A/California/07/2009 and A/England/195/2009 in ELISA. MA-2070 bound rHA of A/California/07/2009 with high affinity (KD = 51.36 ± 9.20 nM) and exhibited potent in vitro neutralization (IC50 = 2.50 μg/mL). MA-2070 bound within the stem domain of HA. MA-M exhibited both hemagglutination inhibition (HI, 1.50 μg/mL) and in vitro neutralization (IC50 = 0.66 μg/mL) activity against the pandemic A/California/07/2009 virus and showed higher binding affinity (KD = 9.80 ± 0.67 nM) than MA-2070. MAb, MA-H generated against the seasonal A/Solomon Islands/03/2006 (H1N1) rHA binds within the head domain and bound the seasonal H1N1 (A/Solomon Islands/03/2006 and A/New Caledonia/20/1990) rHAs with high affinity (KD; 0.72-8.23 nM). MA-H showed high HI (2.50 μg/mL) and in vitro neutralization (IC50 = 2.61 μg/mL) activity against the A/Solomon Islands/03/2006 virus. All 3 MAbs failed to react in ELISA with rHA from various strains of H2N2, H3N2, H5N1, H7N9, and influenza virus B, suggesting their specificity for either pandemic or seasonal H1N1 influenza virus. The MAbs reported here may be useful in developing diagnostic assays.
Collapse
Affiliation(s)
- Nachiket Shembekar
- 1 Reproductive Cell Biology Laboratory, National Institute of Immunology , New Delhi, India
| | | | - Ankita Malik
- 1 Reproductive Cell Biology Laboratory, National Institute of Immunology , New Delhi, India
| | - Ashok Saini
- 1 Reproductive Cell Biology Laboratory, National Institute of Immunology , New Delhi, India
| | | | - Satish Kumar Gupta
- 1 Reproductive Cell Biology Laboratory, National Institute of Immunology , New Delhi, India
| |
Collapse
|
32
|
Influenza virus neuraminidase (NA): a target for antivirals and vaccines. Arch Virol 2016; 161:2087-94. [PMID: 27255748 DOI: 10.1007/s00705-016-2907-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/22/2016] [Indexed: 10/21/2022]
Abstract
Influenza, the most common infectious disease, poses a great threat to human health because of its highly contagious nature and fast transmissibility, often leading to high morbidity and mortality. Effective vaccination strategies may aid in the prevention and control of recurring epidemics and pandemics associated with this infectious disease. However, antigenic shifts and drifts are major concerns with influenza virus, requiring effective global monitoring and updating of vaccines. Current vaccines are standardized primarily based on the amount of hemagglutinin, a major surface antigen, which chiefly constitutes these preparations along with the varying amounts of neuraminidase (NA). Anti-influenza drugs targeting the active site of NA have been in use for more than a decade now. However, NA has not been approved as an effective antigenic component of the influenza vaccine because of standardization issues. Although some studies have suggested that NA antibodies are able to reduce the severity of the disease and induce a long-term and cross-protective immunity, a few major scientific issues need to be addressed prior to launching NA-based vaccines. Interestingly, an increasing number of studies have shown NA to be a promising target for future influenza vaccines. This review is an attempt to consolidate studies that reflect the strength of NA as a suitable vaccine target. The studies discussed in this article highlight NA as a potential influenza vaccine candidate and support taking the process of developing NA vaccines to the next stage.
Collapse
|
33
|
Terajima M, Co MDT, Cruz J, Ennis FA. High Antibody-Dependent Cellular Cytotoxicity Antibody Titers to H5N1 and H7N9 Avian Influenza A Viruses in Healthy US Adults and Older Children. J Infect Dis 2015; 212:1052-60. [PMID: 25795791 PMCID: PMC4668882 DOI: 10.1093/infdis/jiv181] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/10/2015] [Indexed: 11/12/2022] Open
Abstract
Human influenza is a highly contagious acute respiratory illness that is responsible for significant morbidity and excess mortality worldwide. In addition to neutralizing antibodies, there are antibodies that bind to influenza virus-infected cells and mediate lysis of the infected cells by natural killer (NK) cells (antibody-dependent cellular cytotoxicity [ADCC]) or complement (complement-dependent lysis [CDL]). We analyzed sera obtained from 16 healthy adults (18-63 years of age), 52 children (2-17 years of age), and 10 infants (0.75-1 year of age) in the United States, who were unlikely to have been exposed to the avian H7N9 subtype of influenza A virus, by ADCC and CDL assays. As expected, none of these sera had detectable levels of hemagglutination-inhibiting antibodies against the H7N9 virus, but we unexpectedly found high titers of ADCC antibodies to the H7N9 subtype virus in all sera from adults and children aged ≥8 years.
Collapse
Affiliation(s)
| | | | - John Cruz
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester
| | - Francis A. Ennis
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester
| |
Collapse
|
34
|
Streatfield SJ, Kushnir N, Yusibov V. Plant-produced candidate countermeasures against emerging and reemerging infections and bioterror agents. PLANT BIOTECHNOLOGY JOURNAL 2015; 13:1136-59. [PMID: 26387510 PMCID: PMC7167919 DOI: 10.1111/pbi.12475] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 08/06/2015] [Accepted: 08/19/2015] [Indexed: 05/20/2023]
Abstract
Despite progress in the prevention and treatment of infectious diseases, they continue to present a major threat to public health. The frequency of emerging and reemerging infections and the risk of bioterrorism warrant significant efforts towards the development of prophylactic and therapeutic countermeasures. Vaccines are the mainstay of infectious disease prophylaxis. Traditional vaccines, however, are failing to satisfy the global demand because of limited scalability of production systems, long production timelines and product safety concerns. Subunit vaccines are a highly promising alternative to traditional vaccines. Subunit vaccines, as well as monoclonal antibodies and other therapeutic proteins, can be produced in heterologous expression systems based on bacteria, yeast, insect cells or mammalian cells, in shorter times and at higher quantities, and are efficacious and safe. However, current recombinant systems have certain limitations associated with production capacity and cost. Plants are emerging as a promising platform for recombinant protein production due to time and cost efficiency, scalability, lack of harboured mammalian pathogens and possession of the machinery for eukaryotic post-translational protein modification. So far, a variety of subunit vaccines, monoclonal antibodies and therapeutic proteins (antivirals) have been produced in plants as candidate countermeasures against emerging, reemerging and bioterrorism-related infections. Many of these have been extensively evaluated in animal models and some have shown safety and immunogenicity in clinical trials. Here, we overview ongoing efforts to producing such plant-based countermeasures.
Collapse
Affiliation(s)
| | - Natasha Kushnir
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| |
Collapse
|
35
|
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
|
36
|
Suzuki Y. Selecting vaccine strains for H3N2 human influenza A virus. Meta Gene 2015; 4:64-72. [PMID: 25893173 PMCID: PMC4392175 DOI: 10.1016/j.mgene.2015.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 02/17/2015] [Accepted: 03/20/2015] [Indexed: 12/23/2022] Open
Abstract
H3N2 human influenza A virus causes epidemics of influenza mainly in the winter season in temperate regions. Since the antigenicity of this virus evolves rapidly, several attempts have been made to predict the major amino acid sequence of hemagglutinin 1 (HA1) in the target season of vaccination. However, the usefulness of predicted sequence was unclear because its relationship to the antigenicity was unknown. Here the antigenic model for estimating the degree of antigenic difference (antigenic distance) between amino acid sequences of HA1 was integrated into the process of selecting vaccine strains for H3N2 human influenza A virus. When the effectiveness of a potential vaccine strain for a target season was evaluated retrospectively using the average antigenic distance between the strain and the epidemic viruses sampled in the target season, the most effective vaccine strain was identified mostly in the season one year before the target season (pre-target season). Effectiveness of actual vaccines appeared to be lower than that of the strains randomly chosen in the pre-target season on average. It was recommended to replace the vaccine strain for every target season with the strain having the smallest average antigenic distance to the others in the pre-target season. The procedure of selecting vaccine strains for future epidemic seasons described in the present study was implemented in the influenza virus forecasting system (INFLUCAST) (http://www.nsc.nagoya-cu.ac.jp/~yossuzuk/influcast.html).
Collapse
Affiliation(s)
- Yoshiyuki Suzuki
- Graduate School of Natural Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
37
|
Moattari A, Dehghani B, Khodadad N, Tavakoli F. In Silico Functional and Structural Characterization of H1N1 Influenza A Viruses Hemagglutinin, 2010-2013, Shiraz, Iran. Acta Biotheor 2015; 63:183-202. [PMID: 25963671 DOI: 10.1007/s10441-015-9260-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 05/06/2015] [Indexed: 12/27/2022]
Abstract
Hemagglutinin (HA) is a major virulence factor of influenza viruses and plays an important role in viral pathogenesis. Analysis of amino acid changes, epitopes' regions, glycosylation and phosphorylation sites have greatly contributed to the development of new generations of vaccine. The hemagglutinins of 10 selected isolates, 8 of 2010 and 2 of 2013 samples were sequenced and analyzed by several bioinformatic softwares and the results were compared with those of 3 vaccine isolates. The study detected several amino acid changes related to altered epitopes' sites, modification sites and physico-chemical properties. The results showed some conserved modification sites in HA structure. This study is the first analytical research on isolates obtained from Shiraz, Iran, and our results can be used to better understand the genetic diversity and antigenic variations in Iranian and Asian H1N1 pathogenic strains.
Collapse
Affiliation(s)
- Afagh Moattari
- Influenza Research Center, Department of Bacteriology and Virology, Shiraz University of Medical Sciences, 71348-45794, Shiraz, Iran,
| | | | | | | |
Collapse
|
38
|
An anti-H5N1 influenza virus FcDART antibody is a highly efficacious therapeutic agent and prophylactic against H5N1 influenza virus infection. J Virol 2015; 89:4549-61. [PMID: 25673719 DOI: 10.1128/jvi.00078-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Highly pathogenic H5N1 avian influenza viruses are associated with severe disease in humans and continue to be a pandemic threat. While vaccines are available, other approaches are required for patients that typically respond poorly to vaccination, such as the elderly and the immunocompromised. To produce a therapeutic agent that is highly efficacious at low doses and is broadly specific against antigenically drifted H5N1 influenza viruses, we developed two neutralizing monoclonal antibodies and combined them into a single bispecific Fc fusion protein (the Fc dual-affinity retargeting [FcDART] molecule). In mice, a single therapeutic or prophylactic dose of either monoclonal antibody at 2.5 mg/kg of body weight provided 100% protection against challenge with A/Vietnam/1203/04 (H5N1) or the antigenically drifted strain A/Whooper swan/Mongolia/244/05 (H5N1). In ferrets, a single 1-mg/kg prophylactic dose provided 100% protection against A/Vietnam/1203/04 challenge. FcDART was also effective, as a single 2.5-mg/kg therapeutic or prophylactic dose in mice provided 100% protection against A/Vietnam/1203/04 challenge. Antibodies bound to conformational epitopes in antigenic sites on the globular head of the hemagglutinin protein, on the basis of analysis of mutants with antibody escape mutations. While it was possible to generate escape mutants in vitro, they were neutralized by the antibodies in vivo, as mice infected with escape mutants were 100% protected after only a single therapeutic dose of the antibody used to generate the escape mutant in vitro. In summary, we have combined the antigen specificities of two highly efficacious anti-H5N1 influenza virus antibodies into a bispecific FcDART molecule, which represents a strategy to produce broadly neutralizing antibodies that are effective against antigenically diverse influenza viruses. IMPORTANCE Highly pathogenic H5N1 avian influenza viruses are associated with severe disease in humans and are a pandemic threat. A vaccine is available, but other approaches are required for patients that typically respond poorly to vaccination, such as the elderly and the immunocompromised. The variability of the virus means that such an approach must be broad spectrum. To achieve this, we developed two antibodies that neutralize H5N1 influenza viruses. In mice, these antibodies provided complete protection against a spectrum of H5N1 influenza viruses at a single low dose. We then combined the two antibodies into a single molecule, FcDART, which combined the broad-spectrum activity and protective efficacy of both antibodies. This treatment provides a novel and effective therapeutic agent or prophylactic with activity against highly pathogenic H5N1 avian influenza viruses.
Collapse
|
39
|
|
40
|
Comparative structural analysis of haemagglutinin proteins from type A influenza viruses: conserved and variable features. BMC Bioinformatics 2014; 15:363. [PMID: 25492298 PMCID: PMC4265342 DOI: 10.1186/s12859-014-0363-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 10/28/2014] [Indexed: 02/07/2023] Open
Abstract
Background Genome variation is very high in influenza A viruses. However, viral evolution and spreading is strongly influenced by immunogenic features and capacity to bind host cells, depending in turn on the two major capsidic proteins. Therefore, such viruses are classified based on haemagglutinin and neuraminidase types, e.g. H5N1. Current analyses of viral evolution are based on serological and primary sequence comparison; however, comparative structural analysis of capsidic proteins can provide functional insights on surface regions possibly crucial to antigenicity and cell binding. Results We performed extensive structural comparison of influenza virus haemagglutinins and of their domains and subregions to investigate type- and/or domain-specific variation. We found that structural closeness and primary sequence similarity are not always tightly related; moreover, type-specific features could be inferred when comparing surface properties of haemagglutinin subregions, monomers and trimers, in terms of electrostatics and hydropathy. Focusing on H5N1, we found that variation at the receptor binding domain surface intriguingly relates to branching of still circulating clades from those ones that are no longer circulating. Conclusions Evidence from this work suggests that integrating phylogenetic and serological analyses by extensive structural comparison can help in understanding the ‘functional evolution’ of viral surface determinants. In particular, variation in electrostatic and hydropathy patches can provide molecular evolution markers: intriguing surface charge redistribution characterizing the haemagglutinin receptor binding domains from circulating H5N1 clades 2 and 7 might have contributed to antigenic escape hence to their evolutionary success and spreading. Electronic supplementary material The online version of this article (doi:10.1186/s12859-014-0363-5) contains supplementary material, which is available to authorized users.
Collapse
|
41
|
Yusibov V, Kushnir N, Streatfield SJ. Advances and challenges in the development and production of effective plant-based influenza vaccines. Expert Rev Vaccines 2014; 14:519-35. [PMID: 25487788 DOI: 10.1586/14760584.2015.989988] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Influenza infections continue to present a major threat to public health. Traditional modes of influenza vaccine manufacturing are failing to satisfy the global demand because of limited scalability and long production timelines. In contrast, subunit vaccines (SUVs) can be produced in heterologous expression systems in shorter times and at higher quantities. Plants are emerging as a promising platform for SUV production due to time efficiency, scalability, lack of harbored mammalian pathogens and possession of the machinery for eukaryotic post-translational protein modifications. So far, several organizations have utilized plant-based transient expression systems to produce SUVs against influenza, including vaccines based on virus-like particles. Plant-produced influenza SUV candidates have been extensively evaluated in animal models and some have shown safety and immunogenicity in clinical trials. Here, the authors review ongoing efforts and challenges to producing influenza SUV candidates in plants and discuss the likelihood of bringing these products to the market.
Collapse
Affiliation(s)
- Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, 9 Innovation Way, Suite 200, Newark, DE 19711, USA
| | | | | |
Collapse
|
42
|
Relating influenza virus membrane fusion kinetics to stoichiometry of neutralizing antibodies at the single-particle level. Proc Natl Acad Sci U S A 2014; 111:E5143-8. [PMID: 25404330 DOI: 10.1073/pnas.1411755111] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The ability of antibodies binding the influenza hemagglutinin (HA) protein to neutralize viral infectivity is of key importance in the design of next-generation vaccines and for prophylactic and therapeutic use. The two antibodies CR6261 and CR8020 have recently been shown to efficiently neutralize influenza A infection by binding to and inhibiting the influenza A HA protein that is responsible for membrane fusion in the early steps of viral infection. Here, we use single-particle fluorescence microscopy to correlate the number of antibodies or antibody fragments (Fab) bound to an individual virion with the capacity of the same virus particle to undergo membrane fusion. To this end, individual, infectious virus particles bound by fluorescently labeled antibodies/Fab are visualized as they fuse to a planar, supported lipid bilayer. The fluorescence intensity arising from the virus-bound antibodies/Fab is used to determine the number of molecules attached to viral HA while a fluorescent marker in the viral membrane is used to simultaneously obtain kinetic information on the fusion process. We experimentally determine that the stoichiometry required for fusion inhibition by both antibody and Fab leaves large numbers of unbound HA epitopes on the viral surface. Kinetic measurements of the fusion process reveal that those few particles capable of fusion at high antibody/Fab coverage display significantly slower hemifusion kinetics. Overall, our results support a membrane fusion mechanism requiring the stochastic, coordinated action of multiple HA trimers and a model of fusion inhibition by stem-binding antibodies through disruption of this coordinated action.
Collapse
|
43
|
Shoji Y, Prokhnevsky A, Leffet B, Vetter N, Tottey S, Satinover S, Musiychuk K, Shamloul M, Norikane J, Jones RM, Chichester JA, Green BJ, Streatfield SJ, Yusibov V. Immunogenicity of H1N1 influenza virus-like particles produced in Nicotiana benthamiana. Hum Vaccin Immunother 2014; 11:118-23. [PMID: 25483524 PMCID: PMC4514423 DOI: 10.4161/hv.34365] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 07/09/2014] [Indexed: 12/15/2022] Open
Abstract
The H1N1 influenza pandemic of 2009 stimulated interest in developing safe and effective subunit influenza vaccines using rapid and cost-effective recombinant technologies that can avoid dependence on hens' eggs supply and live viruses for production. Among alternative approaches to subunit vaccine development, virus-like particles (VLPs) represent an attractive strategy due to their safety and immunogenicity. Previously, we have produced a recombinant monomeric hemagglutinin (HA) protein derived from the A/California/04/09 (H1N1) strain of influenza virus in a plant-based transient expression system and demonstrated immunogenicity and safety of this monomeric HA in animal models and human volunteers. In an effort to produce higher potency influenza vaccine in plants, we have designed and generated enveloped VLPs using the ectodomain of HA from the A/California/04/09 strain and heterologous sequences. The resulting H1 HA VLPs (HAC-VLPs) elicited robust hemagglutination inhibition antibody responses in mice at doses lower than 1 µg in the presence or absence of Alhydrogel adjuvant. These results suggest enhanced immunogenicity of recombinant HA in the form of an enveloped VLP over soluble antigen.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Aluminum Hydroxide/administration & dosage
- Animals
- Antibodies, Viral/blood
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Antigens, Viral/metabolism
- Hemagglutination Inhibition Tests
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/metabolism
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza Vaccines/isolation & purification
- Mice, Inbred BALB C
- Plants, Genetically Modified/genetics
- Plants, Genetically Modified/metabolism
- Nicotiana/genetics
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/isolation & purification
Collapse
Affiliation(s)
- Yoko Shoji
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Alex Prokhnevsky
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Brett Leffet
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Nancy Vetter
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Stephen Tottey
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Shama Satinover
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | | | - Moneim Shamloul
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Joey Norikane
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - R Mark Jones
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | | | - Brian J Green
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | | | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| |
Collapse
|
44
|
Trombetta CM, Perini D, Mather S, Temperton N, Montomoli E. Overview of Serological Techniques for Influenza Vaccine Evaluation: Past, Present and Future. Vaccines (Basel) 2014; 2:707-34. [PMID: 26344888 PMCID: PMC4494249 DOI: 10.3390/vaccines2040707] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 07/25/2014] [Accepted: 09/22/2014] [Indexed: 12/12/2022] Open
Abstract
Serological techniques commonly used to quantify influenza-specific antibodies include the Haemagglutination Inhibition (HI), Single Radial Haemolysis (SRH) and Virus Neutralization (VN) assays. HI and SRH are established and reproducible techniques, whereas VN is more demanding. Every new influenza vaccine needs to fulfil the strict criteria issued by the European Medicines Agency (EMA) in order to be licensed. These criteria currently apply exclusively to SRH and HI assays and refer to two different target groups-healthy adults and the elderly, but other vaccine recipient age groups have not been considered (i.e., children). The purpose of this timely review is to highlight the current scenario on correlates of protection concerning influenza vaccines and underline the need to revise the criteria and assays currently in use. In addition to SRH and HI assays, the technical advantages provided by other techniques such as the VN assay, pseudotype-based neutralization assay, neuraminidase and cell-mediated immunity assays need to be considered and regulated via EMA criteria, considering the many significant advantages that they could offer for the development of effective vaccines.
Collapse
Affiliation(s)
- Claudia Maria Trombetta
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro, 53100 Siena, Italy.
| | - Daniele Perini
- VisMederi srl, Enterprise in Life Sciences, Via Fiorentina 1, 53100 Siena, Italy.
| | - Stuart Mather
- 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.
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro, 53100 Siena, Italy.
- VisMederi srl, Enterprise in Life Sciences, Via Fiorentina 1, 53100 Siena, Italy.
| |
Collapse
|
45
|
Suzuki Y. Predictability of antigenic evolution for H3N2 human influenza A virus. Genes Genet Syst 2014; 88:225-32. [PMID: 24463525 DOI: 10.1266/ggs.88.225] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Influenza A virus continues to pose a threat to public health. Since this virus can evolve escape mutants rapidly, it is desirable to predict the antigenic evolution for developing effective vaccines. Although empirical methods have been proposed and reported to predict the antigenic evolution more or less accurately, they did not provide much insight into the effects of unobserved mutations and the mechanisms of antigenic evolution. Here a theoretical method was introduced to predict the antigenic evolution of H3N2 human influenza A virus by evaluating de novo mutations through estimating the antigenic distance. The antigenic distance defined with the hemagglutination inhibition (HI) titer was estimated with antigenic models taking into account the volume, isoelectric point, relative solvent accessibility, and distances from receptor-binding sites (RBS) and N-linked glycosylation sites (NGS) for amino acids in hemagglutinin 1 (HA1). When the best model with the optimized parameter values was used to predict the antigenic evolution for the dominant strains, the prediction accuracy was relatively low. However, there appeared to be an overall tendency that the amino acid sites with larger potential net effect on antigenicity were more likely to evolve and the amino acid changes with larger potential effect were more likely to take place, suggesting that natural selection may operate to enhance the antigenic evolution of H3N2 human influenza A virus.
Collapse
|
46
|
Sun X, Cao W, Pappas C, Liu F, Katz JM, Tumpey TM. Effect of receptor binding specificity on the immunogenicity and protective efficacy of influenza virus A H1 vaccines. Virology 2014; 464-465:156-165. [PMID: 25078114 DOI: 10.1016/j.virol.2014.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 06/17/2014] [Accepted: 07/04/2014] [Indexed: 10/25/2022]
Abstract
The biological basis for the poor immunogenicity of unadjuvanted avian influenza A virus vaccines in mammals is not well understood. Here, we mutated the hemagglutinin (HA) of two H1N1 virus vaccines to determine whether virus receptor binding specificity contributes to the low immunogenicity of avian influenza virus vaccines. Mutations were introduced into the HA of an avian influenza virus, A/Duck/New York/15024-21/96 (Dk/96) which switched the binding preference from α2,3- to α2,6-linked sialic acid (SA). A switch in receptor specificity of the human A/South Carolina/1/18 (SC/18) virus generated a mutant virus with α2,3 SA (avian) binding preference. Inactivated vaccines were generated and administered to mice and ferrets intramuscularly. We found that the vaccines with human receptor binding preference induced slightly higher antibody titers and cell-mediated immune responses compared to their isogenic viruses with avian receptor binding specificity. Upon challenge with DK/96 or SC18 virus, differences in lung virus titers between the vaccine groups with different receptor-binding specificities were minimal. Overall, our data suggest that receptor binding specificity contributes only marginally to the immunogenicity of avian influenza vaccines and that other factors may also be involved.
Collapse
Affiliation(s)
- Xiangjie Sun
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunology and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS: G16, Atlanta, GA 30333, United States
| | - Weiping Cao
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunology and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS: G16, Atlanta, GA 30333, United States
| | - Claudia Pappas
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunology and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS: G16, Atlanta, GA 30333, United States
| | - Feng Liu
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunology and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS: G16, Atlanta, GA 30333, United States
| | - Jacqueline M Katz
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunology and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS: G16, Atlanta, GA 30333, United States
| | - Terrence M Tumpey
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunology and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS: G16, Atlanta, GA 30333, United States.
| |
Collapse
|
47
|
Discordant correlation between serological assays observed when measuring heterosubtypic responses against avian influenza H5 and H7 viruses in unexposed individuals. BIOMED RESEARCH INTERNATIONAL 2014; 2014:231365. [PMID: 25013769 PMCID: PMC4071775 DOI: 10.1155/2014/231365] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/11/2014] [Accepted: 05/11/2014] [Indexed: 11/21/2022]
Abstract
The human population is constantly exposed to multiple influenza A subtypes 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 HA protein are of importance since they have been shown to correlate with protective immunity. Serological techniques, detecting these responses, play a critical role for influenza surveillance, vaccine development, and assessment. As the recent human pandemics and avian influenza outbreaks 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 preexisting heterosubtypic immunity in populations. In this study, 68 serum samples collected from the Italian population between 1992 and 2007 were found to be positive for antibodies against H5N1 as determined by single radial hemolysis (SRH), but most were negative when evaluated using haemagglutination inhibition (HI) and microneutralisation (MN) assays. As a result of these discordant serological findings, the increased sensitivity of lentiviral pseudotypes was exploited in pseudotype-based neutralisation (pp-NT) assays and the results obtained provide further insight into the complex nature of humoral immunity against influenza A viruses.
Collapse
|
48
|
Avnir Y, Tallarico AS, Zhu Q, Bennett AS, Connelly G, Sheehan J, Sui J, Fahmy A, Huang CY, Cadwell G, Bankston LA, McGuire AT, Stamatatos L, Wagner G, Liddington RC, Marasco WA. Molecular signatures of hemagglutinin stem-directed heterosubtypic human neutralizing antibodies against influenza A viruses. PLoS Pathog 2014; 10:e1004103. [PMID: 24788925 PMCID: PMC4006906 DOI: 10.1371/journal.ppat.1004103] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Accepted: 03/15/2014] [Indexed: 12/28/2022] Open
Abstract
Recent studies have shown high usage of the IGHV1-69 germline immunoglobulin gene for influenza hemagglutinin stem-directed broadly-neutralizing antibodies (HV1-69-sBnAbs). Here we show that a major structural solution for these HV1-69-sBnAbs is achieved through a critical triad comprising two CDR-H2 loop anchor residues (a hydrophobic residue at position 53 (Ile or Met) and Phe54), and CDR-H3-Tyr at positions 98±1; together with distinctive V-segment CDR amino acid substitutions that occur in positions sparse in AID/polymerase-η recognition motifs. A semi-synthetic IGHV1-69 phage-display library screen designed to investigate AID/polη restrictions resulted in the isolation of HV1-69-sBnAbs that featured a distinctive Ile52Ser mutation in the CDR-H2 loop, a universal CDR-H3 Tyr at position 98 or 99, and required as little as two additional substitutions for heterosubtypic neutralizing activity. The functional importance of the Ile52Ser mutation was confirmed by mutagenesis and by BCR studies. Structural modeling suggests that substitution of a small amino acid at position 52 (or 52a) facilitates the insertion of CDR-H2 Phe54 and CDR-H3-Tyr into adjacent pockets on the stem. These results support the concept that activation and expansion of a defined subset of IGHV1-69-encoded B cells to produce potent HV1-69-sBnAbs does not necessarily require a heavily diversified V-segment acquired through recycling/reentry into the germinal center; rather, the incorporation of distinctive amino acid substitutions by Phase 2 long-patch error-prone repair of AID-induced mutations or by random non-AID SHM events may be sufficient. We propose that these routes of B cell maturation should be further investigated and exploited as a pathway for HV1-69-sBnAb elicitation by vaccination. The quest for universal influenza vaccine has gained wide interest with the discovery of human neutralizing antibodies that are able to variably cross neutralize and protect against different influenza strains, subtypes, groups and lineages. These antibodies, which bind to a highly conserved epitope in the hemagglutinin stem, are often encoded by rearranged IGHV1-69 germline genes that alone make contact with HA and prevent virus entry and emergence of escape mutants. Our study was undertaken to gain an understanding of what structural requirements enable a rearranged IGHV1-69 Ab to become a potent cross-neutralizing antibody. We found that in addition to a critical amino acid triad consisting of a pair of anchor residues in CDR-H2 and a properly positioned CDR-H3 Tyr, distinctive V-segment substitutions that arise in positions that are distinct from phase I AID somatic hypermutation (SHM) hotspot motifs are often required. As few as two V-segment SHM can fulfill this role which appears to facilitate the optimal binding of CDR-H2 Phe54 and CHR-H3-Tyr into adjacent hydrophobic pockets in the HA stem. These studies provide new information on the SHM requirements for IGHV1-69-encoded B cells to produce HV1-69-sBnAbs and suggest that there may exist alternative routes to their elicitation by vaccination.
Collapse
Affiliation(s)
- Yuval Avnir
- Department of Cancer Immunology & AIDS, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aimee S Tallarico
- Department of Cancer Immunology & AIDS, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Quan Zhu
- Department of Cancer Immunology & AIDS, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Andrew S Bennett
- Department of Cancer Immunology & AIDS, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Gene Connelly
- Department of Cancer Immunology & AIDS, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jared Sheehan
- Department of Cancer Immunology & AIDS, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jianhua Sui
- Department of Cancer Immunology & AIDS, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Amr Fahmy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Chiung-yu Huang
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Greg Cadwell
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Laurie A Bankston
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Andrew T McGuire
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Leonidas Stamatatos
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Gerhard Wagner
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Robert C Liddington
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Wayne A Marasco
- Department of Cancer Immunology & AIDS, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
49
|
Tate MD, Job ER, Deng YM, Gunalan V, Maurer-Stroh S, Reading PC. Playing hide and seek: how glycosylation of the influenza virus hemagglutinin can modulate the immune response to infection. Viruses 2014; 6:1294-316. [PMID: 24638204 PMCID: PMC3970151 DOI: 10.3390/v6031294] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 03/03/2014] [Accepted: 03/07/2014] [Indexed: 12/22/2022] Open
Abstract
Seasonal influenza A viruses (IAV) originate from pandemic IAV and have undergone changes in antigenic structure, including addition of glycans to the hemagglutinin (HA) glycoprotein. The viral HA is the major target recognized by neutralizing antibodies and glycans have been proposed to shield antigenic sites on HA, thereby promoting virus survival in the face of widespread vaccination and/or infection. However, addition of glycans can also interfere with the receptor binding properties of HA and this must be compensated for by additional mutations, creating a fitness barrier to accumulation of glycosylation sites. In addition, glycans on HA are also recognized by phylogenetically ancient lectins of the innate immune system and the benefit provided by evasion of humoral immunity is balanced by attenuation of infection. Therefore, a fine balance must exist regarding the optimal pattern of HA glycosylation to offset competing pressures associated with recognition by innate defenses, evasion of humoral immunity and maintenance of virus fitness. In this review, we examine HA glycosylation patterns of IAV associated with pandemic and seasonal influenza and discuss recent advancements in our understanding of interactions between IAV glycans and components of innate and adaptive immunity.
Collapse
Affiliation(s)
- Michelle D Tate
- Centre for Innate Immunity and Infectious Diseases, Monash Institute of Medical Research, Monash University, Clayton, Victoria, 3168, Australia.
| | - Emma R Job
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria 3010, Australia.
| | - Yi-Mo Deng
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, at the Peter Doherty Institute for Infection and Immunity, Victoria 3010, Australia.
| | - Vithiagaran Gunalan
- Bioinformatics Institute, Agency for Science, Technology and Research, 138671, Singapore.
| | - Sebastian Maurer-Stroh
- Bioinformatics Institute, Agency for Science, Technology and Research, 138671, Singapore.
| | - Patrick C Reading
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria 3010, Australia.
| |
Collapse
|
50
|
Abstract
Influenza A and B viruses are highly contagious respiratory pathogens with a considerable medical and socioeconomical burden and known pandemic potential. Current influenza vaccines require annual updating and provide only partial protection in some risk groups. Due to the global spread of viruses with resistance to the M2 proton channel inhibitor amantadine or the neuraminidase inhibitor oseltamivir, novel antiviral agents with an original mode of action are urgently needed. We here focus on emerging options to interfere with the influenza virus entry process, which consists of the following steps: attachment of the viral hemagglutinin to the sialylated host cell receptors, endocytosis, M2-mediated uncoating, low pH-induced membrane fusion, and, finally, import of the viral ribonucleoprotein into the nucleus. We review the current functional and structural insights in the viral and cellular components of this entry process, and the diverse antiviral strategies that are being explored. This encompasses small molecule inhibitors as well as macromolecules such as therapeutic antibodies. There is optimism that at least some of these innovative concepts to block influenza virus entry will proceed from the proof of concept to a more advanced stage. Special attention is therefore given to the challenging issues of influenza virus (sub)type-dependent activity or potential drug resistance.
Collapse
Affiliation(s)
| | - Lieve Naesens
- Rega Institute for Medical ResearchKU LeuvenLeuvenBelgium
| |
Collapse
|