1
|
Mackin SR, Sariol A, Diamond MS. Antibody-mediated control mechanisms of viral infections. Immunol Rev 2024; 328:205-220. [PMID: 39162394 PMCID: PMC11661935 DOI: 10.1111/imr.13383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Antibodies generated after vaccination or natural pathogen exposure are essential mediators of protection against many infections. Most studies with viruses have focused on antibody neutralization, in which protection is conferred by the fragment antigen binding region (Fab) through targeting of different steps in the viral lifecycle including attachment, internalization, fusion, and egress. Beyond neutralization, the fragment crystallizable (Fc) region of antibodies can integrate innate and adaptive immune responses by engaging complement components and distinct Fc gamma receptors (FcγR) on different host immune cells. In this review, we discuss recent advances in our understanding of antibody neutralization and Fc effector functions, and the assays used to measure them. Additionally, we describe the contexts in which these mechanisms are associated with protection against viruses and highlight how Fc-FcγR interactions can improve the potency of antibody-based therapies.
Collapse
Affiliation(s)
- Samantha R. Mackin
- Department of Medicine, Washington University School of Medicine, MO 63110, USA
- Department of Pathology & Immunology and Center for Genome Sciences, Lab & Genomic Medicine, Washington University School of Medicine, MO 63110, USA
| | - Alan Sariol
- Department of Medicine, Washington University School of Medicine, MO 63110, USA
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, MO 63110, USA
- Department of Pathology & Immunology and Center for Genome Sciences, Lab & Genomic Medicine, Washington University School of Medicine, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
- Andrew M. and Jane M. Bursky the Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
2
|
Myers ML, Gallagher JR, Woolfork DD, Khorrami ND, Park WB, Maldonado-Puga S, Bohrnsen E, Schwarz BH, Alves DA, Bock KW, Dearborn AD, Harris AK. Structure-guided assembly of an influenza spike nanobicelle vaccine provides pan H1 intranasal protection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613335. [PMID: 39372767 PMCID: PMC11451756 DOI: 10.1101/2024.09.16.613335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Development of intranasal vaccines for respiratory viruses has gained popularity. However, currently only a live-attenuated influenza vaccine is FDA-approved for intranasal administration. Here, we focused on influenza virus as it circulates seasonally, has pandemic potential, and has vaccine formulations that present hemagglutinin (HA) in different structural arrangements. These display differences have not been correlated with induction of pan-H1 antibodies or shown to provide intranasal protection. Using electron microscopy, biochemistry and animal studies, we identified HA complexes arranged as lipid discs with multiple trimeric HAs displayed along the perimeter, termed spike nanobicelles (SNB). We utilized a structure-guided approach to synthesize in vitro assembled spiked nanobicelles (IA-SNB) from a classical 1934 H1N1 influenza virus. IA-SNBs elicited pan-H1 antibodies and provided protection against antigenically divergent H1N1 viruses via intranasal immunizations. Viral glycoprotein spikes displayed as SNBs could aid in combating antigenic variation and provide innovative intranasal vaccines to aid universal influenza vaccine development.
Collapse
Affiliation(s)
- Mallory L. Myers
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - John R. Gallagher
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - De’Marcus D. Woolfork
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - Noah D. Khorrami
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - William B. Park
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - Samantha Maldonado-Puga
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - Eric Bohrnsen
- Protein Chemistry Section, Research and Technologies Branch, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, USA 59840
| | - Benjamin H. Schwarz
- Protein Chemistry Section, Research and Technologies Branch, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, USA 59840
| | - Derron A. Alves
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Room BN25, Bethesda, MD, USA 20892
| | - Kevin W. Bock
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Room BN25, Bethesda, MD, USA 20892
| | - Altaira D. Dearborn
- Structural Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6531, Bethesda, MD, USA 20892
| | - Audray K. Harris
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| |
Collapse
|
3
|
Kosik I, Da Silva Santos J, Angel M, Hu Z, Holly J, Gibbs JS, Gill T, Kosikova M, Li T, Bakhache W, Dolan PT, Xie H, Andrews SF, Gillespie RA, Kanekiyo M, McDermott AB, Pierson TC, Yewdell JW. C1q enables influenza hemagglutinin stem binding antibodies to block viral attachment and broadens the antibody escape repertoire. Sci Immunol 2024; 9:eadj9534. [PMID: 38517951 DOI: 10.1126/sciimmunol.adj9534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 02/14/2024] [Indexed: 03/24/2024]
Abstract
Antigenic drift, the gradual accumulation of amino acid substitutions in the influenza virus hemagglutinin (HA) receptor protein, enables viral immune evasion. Antibodies (Abs) specific for the drift-resistant HA stem region are a promising universal influenza vaccine target. Although anti-stem Abs are not believed to block viral attachment, here we show that complement component 1q (C1q), a 460-kilodalton protein with six Ab Fc-binding domains, confers attachment inhibition to anti-stem Abs and enhances their fusion and neuraminidase inhibition. As a result, virus neutralization activity in vitro is boosted up to 30-fold, and in vivo protection from influenza PR8 infection in mice is enhanced. These effects reflect increased steric hindrance and not increased Ab avidity. C1q greatly expands the anti-stem Ab viral escape repertoire to include residues throughout the HA, some of which cause antigenic alterations in the globular region or modulate HA receptor avidity. We also show that C1q enhances the neutralization activity of non-receptor binding domain anti-SARS-CoV-2 spike Abs, an effect dependent on spike density on the virion surface. These findings demonstrate that C1q can greatly expand Ab function and thereby contribute to viral evolution and immune escape.
Collapse
Affiliation(s)
- Ivan Kosik
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Jefferson Da Silva Santos
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Mathew Angel
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Zhe Hu
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Jaroslav Holly
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - James S Gibbs
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Tanner Gill
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Martina Kosikova
- Laboratory of Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Tiansheng Li
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - William Bakhache
- Quantitative Virology and Evolution Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Patrick T Dolan
- Quantitative Virology and Evolution Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Hang Xie
- Laboratory of Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Sarah F Andrews
- Vaccine Immunology Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rebecca A Gillespie
- Molecular Immunoengineering Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Masaru Kanekiyo
- Molecular Immunoengineering Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Adrian B McDermott
- Vaccine Immunology Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Theodore C Pierson
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Jonathan W Yewdell
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| |
Collapse
|
4
|
Mellors J, Carroll M. Direct enhancement of viral neutralising antibody potency by the complement system: a largely forgotten phenomenon. Cell Mol Life Sci 2024; 81:22. [PMID: 38200235 PMCID: PMC10781860 DOI: 10.1007/s00018-023-05074-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 01/12/2024]
Abstract
Neutralisation assays are commonly used to assess vaccine-induced and naturally acquired immune responses; identify correlates of protection; and inform important decisions on the screening, development, and use of therapeutic antibodies. Neutralisation assays are useful tools that provide the gold standard for measuring the potency of neutralising antibodies, but they are not without limitations. Common methods such as the heat-inactivation of plasma samples prior to neutralisation assays, or the use of anticoagulants such as EDTA for blood collection, can inactivate the complement system. Even in non-heat-inactivated samples, the levels of complement activity can vary between samples. This can significantly impact the conclusions regarding neutralising antibody potency. Restoration of the complement system in these samples can be achieved using an exogenous source of plasma with preserved complement activity or with purified complement proteins. This can significantly enhance the neutralisation titres for some antibodies depending on characteristics such as antibody isotype and the epitope they bind, enable neutralisation with otherwise non-neutralising antibodies, and demonstrate a better relationship between in vitro and in vivo findings. In this review, we discuss the evidence for complement-mediated enhancement of antibody neutralisation against a range of viruses, explore the potential mechanisms which underpin this enhancement, highlight current gaps in the literature, and provide a brief summary of considerations for adopting this approach in future research applications.
Collapse
Affiliation(s)
- Jack Mellors
- Centre for Human Genetics and the Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Miles Carroll
- Centre for Human Genetics and the Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
5
|
Mosquera-Sulbaran JA, Pedreañez A, Hernandez-Fonseca JP, Hernandez-Fonseca H. Angiotensin II and dengue. Arch Virol 2023; 168:191. [PMID: 37368044 DOI: 10.1007/s00705-023-05814-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 05/09/2023] [Indexed: 06/28/2023]
Abstract
Dengue is a disease caused by a flavivirus that is transmitted principally by the bite of an Aedes aegypti mosquito and represents a major public-health problem. Many studies have been carried out to identify soluble factors that are involved in the pathogenesis of this infection. Cytokines, soluble factors, and oxidative stress have been reported to be involved in the development of severe disease. Angiotensin II (Ang II) is a hormone with the ability to induce the production of cytokines and soluble factors related to the inflammatory processes and coagulation disorders observed in dengue. However, a direct involvement of Ang II in this disease has not been demonstrated. This review primarily summarizes the pathophysiology of dengue, the role of Ang II in various diseases, and reports that are highly suggestive of the involvement of this hormone in dengue.
Collapse
Affiliation(s)
- Jesus A Mosquera-Sulbaran
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, 4001-A, Venezuela.
| | - Adriana Pedreañez
- Cátedra de Inmunología, Escuela de Bioanálisis, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Juan Pablo Hernandez-Fonseca
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, 4001-A, Venezuela
- Servicio de Microscopia Electronica del Centro Nacional de Biotecnologia (CNB- CSIC) Madrid, Madrid, España
| | - Hugo Hernandez-Fonseca
- Department of Anatomy, Physiology and Pharmacology, School of Veterinary Medicine, Saint George's University, True Blue, West Indies, Grenada
| |
Collapse
|
6
|
Beirag N, Varghese PM, Neto MM, Al Aiyan A, Khan HA, Qablan M, Shamji MH, Sim RB, Temperton N, Kishore U. Complement Activation-Independent Attenuation of SARS-CoV-2 Infection by C1q and C4b-Binding Protein. Viruses 2023; 15:1269. [PMID: 37376569 DOI: 10.3390/v15061269] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
The complement system is a key component of the innate immune response to viruses and proinflammatory events. Exaggerated complement activation has been attributed to the induction of a cytokine storm in severe SARS-CoV-2 infection. However, there is also an argument for the protective role of complement proteins, given their local synthesis or activation at the site of viral infection. This study investigated the complement activation-independent role of C1q and C4b-binding protein (C4BP) against SARS-CoV-2 infection. The interactions of C1q, its recombinant globular heads, and C4BP with the SARS-CoV-2 spike and receptor binding domain (RBD) were examined using direct ELISA. In addition, RT-qPCR was used to evaluate the modulatory effect of these complement proteins on the SARS-CoV-2-mediated immune response. Cell binding and luciferase-based viral entry assays were utilised to assess the effects of C1q, its recombinant globular heads, and C4BP on SARS-CoV-2 cell entry. C1q and C4BP bound directly to SARS-CoV-2 pseudotype particles via the RBD domain of the spike protein. C1q via its globular heads and C4BP were found to reduce binding as well as viral transduction of SARS-CoV-2 spike protein expressing lentiviral pseudotypes into transfected A549 cells expressing human ACE2 and TMPRSS2. Furthermore, the treatment of the SARS-CoV-2 spike, envelope, nucleoprotein, and membrane protein expressing alphaviral pseudotypes with C1q, its recombinant globular heads, or C4BP triggered a reduction in mRNA levels of proinflammatory cytokines and chemokines such as IL-1β, IL-8, IL-6, TNF-α, IFN-α, and RANTES (as well as NF-κB) in A549 cells expressing human ACE2 and TMPRSS2. In addition, C1q and C4BP treatment also reduced SARS-CoV-2 pseudotype infection-mediated NF-κB activation in A549 cells expressing human ACE2 and TMPRSS2. C1q and C4BP are synthesised primarily by hepatocytes; however, they are also produced by macrophages, and alveolar type II cells, respectively, locally at the pulmonary site. These findings support the notion that the locally produced C1q and C4BP can be protective against SARS-CoV-2 infection in a complement activation-independent manner, offering immune resistance by inhibiting virus binding to target host cells and attenuating the infection-associated inflammatory response.
Collapse
Affiliation(s)
- Nazar Beirag
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
| | - Praveen M Varghese
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Martin Mayora Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent and Greenwich, Kent ME4 4TB, UK
| | - Ahmad Al Aiyan
- Department of Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Haseeb A Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 4545, Saudi Arabia
| | - Moneeb Qablan
- Department of Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Mohamed H Shamji
- Immunomodulation and Tolerance Group, Department of Allergy and Clinical Immunology, National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
| | - Robert B Sim
- MRC Immunochemistry Unit, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent and Greenwich, Kent ME4 4TB, UK
| | - Uday Kishore
- Department of Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
7
|
Zhou J, Qiao ML, Jahejo AR, Han XY, Wang P, Wang Y, Ren JL, Niu S, Zhao YJ, Zhang D, Bi YH, Wang QH, Si LL, Fan RW, Shang GJ, Tian WX. Effect of Avian Influenza Virus subtype H9N2 on the expression of complement-associated genes in chicken erythrocytes. Br Poult Sci 2023:1-9. [PMID: 36939295 DOI: 10.1080/00071668.2023.2191308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
The H9N2 subtype avian influenza virus can infect both chickens and humans. Previous studies have reported a role for erythrocytes in immunity. However, the role of H9N2 against chicken erythrocytes and the presence of complement-related genes in erythrocytes has not been studied. This research investigated the effect of H9N2 on complement-associated gene expression in chicken erythrocytes. The expression of complement-associated genes (C1s, C1q, C2, C3, C3ar1, C4, C4a, C5, C5ar1, C7, CD93 and CFD) was detected by reverse transcription-polymerase chain reaction (RT-PCR). Quantitative Real-Time PCR (qRT-PCR) was used to analyse the differential expression of complement-associated genes in chicken erythrocytes at 0 h, 2 h, 6 h and 10 h after the interaction between H9N2 virus and chicken erythrocytes in vitro and 3, 7 and 14 d after H9N2 virus nasal infection of chicks. Expression levels of C1q, C4, C1s, C2, C3, C5, C7 and CD93 were significantly up-regulated at 2 h and significantly down-regulated at 10 h. Gene expression levels of C1q, C3ar1, C4a, CFD and C5ar1 were seen to be different at each time point. The expression levels of C1q, C4, C1s, C2, C3, C5, C7, CFD, C3ar1, C4a and C5ar1 were significantly up-regulated at 7 d and the gene expression of levels of C3, CD93 and C5ar1 were seen to be different at each time point. The results confirmed that all the complement-associated genes were expressed in chicken erythrocytes and showed the H9N2 virus interaction with chicken erythrocytes and subsequent regulation of chicken erythrocyte complement-associated genes expression. This study reported, for the first time, the relationship between H9N2 and complement system of chicken erythrocytes, which will provide a foundation for further research into the prevention and control of H9N2 infection.
Collapse
Affiliation(s)
- J Zhou
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - M L Qiao
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - A R Jahejo
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - X Y Han
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - P Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Y Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - J L Ren
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - S Niu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Y J Zhao
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - D Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Y H Bi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), Chinese Academy of Sciences, Beijing, China
| | - Q H Wang
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - L L Si
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - R W Fan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - G J Shang
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - W X Tian
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| |
Collapse
|
8
|
Anti-SARS-Cov-2 S-RBD IgG Formed after BNT162b2 Vaccination Can Bind C1q and Activate Complement. J Immunol Res 2022; 2022:7263740. [PMID: 36573216 PMCID: PMC9789906 DOI: 10.1155/2022/7263740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 11/19/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Background The ability of vaccine-induced antibodies to bind C1q could affect pathogen neutralization. In this study, we investigated C1q binding and subsequent complement activation by anti-spike (S) protein receptor-binding domain (RBD) specific antibodies produced following vaccination with either the mRNA vaccine BNT162b2 or the inactivated vaccine BBIBP-CorV. Methods Serum samples were collected in the period of July 2021-March 2022. Participants' demographic data, type of vaccine, date of vaccination, as well as adverse effects of the vaccine were recorded. The serum samples were incubated with S protein RBD-coated plates. Levels of human IgG, IgA, IgM, C1q, and mannose-binding lectin (MBL) that were bound to the plate, as well as formed C3d, and C5b-9 were compared between different groups of participants. Results A total of 151 samples were collected from vaccinated (n = 116) and nonvaccinated (n = 35) participants. Participants who received either one or two doses of BNT162b2 formed higher levels of anti-RBD IgG and IgA than participants who received BBIBP-CorV. The anti-RBD IgG formed following either vaccine bound C1q, but significantly more C1q binding was observed in participants who received BNT162b2. Subsequently, C5b-9 formation was significantly higher in participants who received BNT162b2, while no significant difference in C5b-9 formation was found between the nonvaccinated and BBIBP-CorV groups. The formation of C5b-9 was strongly correlated to C1q binding and not to MBL binding, additionally, the ratio of formed C5b-9/bound C1q was significantly higher in the BNT162b2 group. Conclusion Anti-RBD IgG formed following vaccination can bind C1q with subsequent complement activation, and the degree of terminal complement pathway activation differed between vaccines, which could play a role in the protection offered by COVID-19 vaccines. Further investigation into the correlation between vaccine protection and vaccine-induced antibodies' ability to activate complement is required.
Collapse
|
9
|
Varghese PM, Kishore U, Rajkumari R. Human C1q Regulates Influenza A Virus Infection and Inflammatory Response via Its Globular Domain. Int J Mol Sci 2022; 23:3045. [PMID: 35328462 PMCID: PMC8949502 DOI: 10.3390/ijms23063045] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 01/27/2023] Open
Abstract
The Influenza A virus (IAV) is a severe respiratory pathogen. C1q is the first subcomponent of the complement system's classical pathway. C1q is composed of 18 polypeptide chains. Each of these chains contains a collagen-like region located at the N terminus, and a C-terminal globular head region organized as a heterotrimeric structure (ghA, ghB and ghC). This study was aimed at investigating the complement activation-independent modulation by C1q and its individual recombinant globular heads against IAV infection. The interaction of C1q and its recombinant globular heads with IAV and its purified glycoproteins was examined using direct ELISA and far-Western blotting analysis. The effect of the complement proteins on IAV replication kinetics and immune modulation was assessed by qPCR. The IAV entry inhibitory properties of C1q and its recombinant globular heads were confirmed using cell binding and luciferase reporter assays. C1q bound IAV virions via HA, NA and M1 IAV proteins, and suppressed replication in H1N1, while promoting replication in H3N2-infected A549 cells. C1q treatment further triggered an anti-inflammatory response in H1N1 and pro-inflammatory response in H3N2-infected cells as evident from differential expression of TNF-α, NF-κB, IFN-α, IFN-β, IL-6, IL-12 and RANTES. Furthermore, C1q treatment was found to reduce luciferase reporter activity of MDCK cells transfected with H1N1 pseudotyped lentiviral particles, indicative of an entry inhibitory role of C1q against infectivity of IAV. These data appear to demonstrate the complement-independent subtype specific modulation of IAV infection by locally produced C1q.
Collapse
Affiliation(s)
- Praveen M. Varghese
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, London UB8 3PH, UK;
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, London UB8 3PH, UK;
| | - Reena Rajkumari
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| |
Collapse
|
10
|
Zolotarova O, Fesenko A, Holubka O, Radchenko L, Bortz E, Budzanivska I, Mironenko A. Genotypic Variants of Pandemic H1N1 Influenza A Viruses Isolated from Severe Acute Respiratory Infections in Ukraine during the 2015/16 Influenza Season. Viruses 2021; 13:2125. [PMID: 34834932 PMCID: PMC8619959 DOI: 10.3390/v13112125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/05/2021] [Accepted: 10/15/2021] [Indexed: 01/15/2023] Open
Abstract
Human type A influenza viruses A(H1N1)pdm09 have caused seasonal epidemics of influenza since the 2009-2010 pandemic. A(H1N1)pdm09 viruses had a leading role in the severe epidemic season of 2015/16 in the Northern Hemisphere and caused a high incidence of acute respiratory infection (ARI) in Ukraine. Serious complications of influenza-associated severe ARI (SARI) were observed in the very young and individuals at increased risk, and 391 fatal cases occurred in the 2015/16 epidemic season. We analyzed the genetic changes in the genomes of A(H1N1)pdm09 influenza viruses isolated from SARI cases in Ukraine during the 2015/16 season. The viral hemagglutinin (HA) fell in H1 group 6B.1 for all but four isolates, with known mutations affecting glycosylation, the Sa antigenic site (S162N in all 6B.1 isolates), or virulence (D222G/N in two isolates). Other mutations occurred in antigenic site Ca (A141P and S236P), and a subgroup of four strains were in group 6B.2, with potential alterations to antigenicity in A(H1N1)pdm09 viruses circulating in 2015/16 in Ukraine. A cluster of Ukrainian isolates exhibited novel D2E and N48S mutations in the RNA binding domain, and E125D in the effector domain, of immune evasion nonstructural protein 1 (NS1). The diverse spectrum of amino-acid substitutions in HA, NS1, and other viral proteins including nucleoprotein (NP) and the polymerase complex suggested the concurrent circulation of multiple lineages of A(H1N1)pdm09 influenza viruses in the human population in Ukraine, a country with low vaccination coverage, complicating public health measures against influenza.
Collapse
Affiliation(s)
- Oksana Zolotarova
- Educational Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine;
| | - Anna Fesenko
- Gromashevsky L.V. Institute of Epidemiology and Infectious Diseases, National Academy of Medical Sciences of Ukraine, 03680 Kyiv, Ukraine; (A.F.); (O.H.); (L.R.); (A.M.)
| | - Olga Holubka
- Gromashevsky L.V. Institute of Epidemiology and Infectious Diseases, National Academy of Medical Sciences of Ukraine, 03680 Kyiv, Ukraine; (A.F.); (O.H.); (L.R.); (A.M.)
| | - Larysa Radchenko
- Gromashevsky L.V. Institute of Epidemiology and Infectious Diseases, National Academy of Medical Sciences of Ukraine, 03680 Kyiv, Ukraine; (A.F.); (O.H.); (L.R.); (A.M.)
| | - Eric Bortz
- Department of Biological Sciences, University of Alaska, 3211 Providence Dr., Anchorage, AK 99508, USA;
| | - Iryna Budzanivska
- Educational Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine;
| | - Alla Mironenko
- Gromashevsky L.V. Institute of Epidemiology and Infectious Diseases, National Academy of Medical Sciences of Ukraine, 03680 Kyiv, Ukraine; (A.F.); (O.H.); (L.R.); (A.M.)
| |
Collapse
|
11
|
Matrix Protein 2 Extracellular Domain-Specific Monoclonal Antibodies Are an Effective and Potentially Universal Treatment for Influenza A. J Virol 2021; 95:JVI.01027-20. [PMID: 33268521 PMCID: PMC8092830 DOI: 10.1128/jvi.01027-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Influenza virus infection causes significant morbidity and mortality worldwide. Humans fail to make a universally protective memory immune response to influenza A. Hemagglutinin and Neuraminidase undergo antigenic drift and shift, resulting in new influenza A strains to which humans are naive. Seasonal vaccines are often ineffective and escape mutants have been reported to all treatments for influenza A. In the absence of a universal influenza A vaccine or treatment, influenza A will remain a significant threat to human health. The extracellular domain of the M2-ion channel (M2e) is an ideal antigenic target for a universal therapeutic agent, as it is highly conserved across influenza A serotypes, has a low mutation rate, and is essential for viral entry and replication. Previous M2e-specific monoclonal antibodies (M2e-MAbs) show protective potential against influenza A, however, they are either strain specific or have limited efficacy. We generated seven murine M2e-MAbs and utilized in vitro and in vivo assays to validate the specificity of our novel M2e-MAbs and to explore the universality of their protective potential. Our data shows our M2e-MAbs bind to M2e peptide, HEK cells expressing the M2 channel, as well as, influenza virions and MDCK-ATL cells infected with influenza viruses of multiple serotypes. Our antibodies significantly protect highly influenza A virus susceptible BALB/c mice from lethal challenge with H1N1 A/PR/8/34, pH1N1 A/CA/07/2009, H5N1 A/Vietnam/1203/2004, and H7N9 A/Anhui/1/2013 by improving survival rates and weight loss. Based on these results, at least four of our seven M2e-MAbs show strong potential as universal influenza A treatments.IMPORTANCE Despite a seasonal vaccine and multiple therapeutic treatments, Influenza A remains a significant threat to human health. The biggest obstacle is producing a vaccine or treatment for influenza A is their universality or efficacy against not only seasonal variances in the influenza virus, but also against all human, avian, and swine serotypes and, therefore, potential pandemic strains. M2e has huge potential as a target for a vaccine or treatment against influenza A. It is the most conserved external protein on the virus. Antibodies against M2e have made it to clinical trials, but not succeeded. Here, we describe novel M2e antibodies produced in mice that are not only protective at low doses, but that we extensively test to determine their universality and found to be cross protective against all strains tested. Additionally, our work begins to elucidate the critical role of isotype for an influenza A monoclonal antibody therapeutic.
Collapse
|
12
|
Arthur CM, Chonat S, Fasano R, Yee MEM, Josephson CD, Roback JD, Stowell SR. Examining the Role of Complement in Predicting, Preventing, and Treating Hemolytic Transfusion Reactions. Transfus Med Rev 2019; 33:217-224. [PMID: 31679762 PMCID: PMC7147990 DOI: 10.1016/j.tmrv.2019.09.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/24/2022]
Abstract
Red blood cell (RBC) transfusion is a critical component of optimal management for a broad range of conditions. Regardless of the indication, pretransfusion testing is required to appropriately match RBC donors and recipients to provide immunologically compatible blood. Although this approach is effective in the vast majority of situations, occasionally, patients will inadvertently receive an incompatible RBC transfusion, which can result in a hemolytic transfusion reaction (HTR). In addition, patients with life-threatening anemia and a complex alloantibody profile, which precludes rapid procurement of compatible RBCs, may also receive incompatible RBCs, placing them at risk for an HTR. Despite the rarity of these clinical situations, when incompatible blood transfusion results in an HTR, the consequences can be devastating. In this review, we will explore the challenges associated with actively preventing and treating acute HTRs following incompatible RBC transfusion. In doing so, we will focus primarily on the role of complement, not only as a key player in HTRs, but also as a potential target for the prevention and treatment of HTRs.
Collapse
Affiliation(s)
- Connie M Arthur
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Satheesh Chonat
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA
| | - Ross Fasano
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA; Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA
| | - Marianne E M Yee
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA; Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA
| | - Cassandra D Josephson
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA; Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA
| | - John D Roback
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA
| | - Sean R Stowell
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA.
| |
Collapse
|
13
|
Kurtovic L, Boyle MJ, Opi DH, Kennedy AT, Tham WH, Reiling L, Chan JA, Beeson JG. Complement in malaria immunity and vaccines. Immunol Rev 2019; 293:38-56. [PMID: 31556468 PMCID: PMC6972673 DOI: 10.1111/imr.12802] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022]
Abstract
Developing efficacious vaccines for human malaria caused by Plasmodium falciparum is a major global health priority, although this has proven to be immensely challenging over the decades. One major hindrance is the incomplete understanding of specific immune responses that confer protection against disease and/or infection. While antibodies to play a crucial role in malaria immunity, the functional mechanisms of these antibodies remain unclear as most research has primarily focused on the direct inhibitory or neutralizing activity of antibodies. Recently, there is a growing body of evidence that antibodies can also mediate effector functions through activating the complement system against multiple developmental stages of the parasite life cycle. These antibody‐complement interactions can have detrimental consequences to parasite function and viability, and have been significantly associated with protection against clinical malaria in naturally acquired immunity, and emerging findings suggest these mechanisms could contribute to vaccine‐induced immunity. In order to develop highly efficacious vaccines, strategies are needed that prioritize the induction of antibodies with enhanced functional activity, including the ability to activate complement. Here we review the role of complement in acquired immunity to malaria, and provide insights into how this knowledge could be used to harness complement in malaria vaccine development.
Collapse
Affiliation(s)
- Liriye Kurtovic
- Burnet Institute, Melbourne, Vic., Australia.,Central Clinical School, Monash University, Melbourne, Vic., Australia
| | | | | | - Alexander T Kennedy
- Walter and Eliza Hall Institute, Melbourne, Vic., Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - Wai-Hong Tham
- Walter and Eliza Hall Institute, Melbourne, Vic., Australia
| | | | - Jo-Anne Chan
- Burnet Institute, Melbourne, Vic., Australia.,Central Clinical School, Monash University, Melbourne, Vic., Australia
| | - James G Beeson
- Burnet Institute, Melbourne, Vic., Australia.,Central Clinical School, Monash University, Melbourne, Vic., Australia.,Department of Microbiology, Monash University, Clayton, Vic., Australia.,Department of Medicine, The University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
14
|
Long-Lasting Mucosal and Systemic Immunity against Influenza A Virus Is Significantly Prolonged and Protective by Nasal Whole Influenza Immunization with Mucosal Adjuvant N3 and DNA-Plasmid Expressing Flagellin in Aging In- and Outbred Mice. Vaccines (Basel) 2019; 7:vaccines7030064. [PMID: 31315253 PMCID: PMC6789645 DOI: 10.3390/vaccines7030064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 12/17/2022] Open
Abstract
Background: Vaccination is commonly used to prevent and control influenza infection in humans. However, improvements in the ease of delivery and strength of immunogenicity could markedly improve herd immunity. The aim of this pre-clinical study is to test the potential improvements to existing intranasal delivery of formalin-inactivated whole Influenza A vaccines (WIV) by formulation with a cationic lipid-based adjuvant (N3). Additionally, we combined WIV and N3 with a DNA-encoded TLR5 agonist secreted flagellin (pFliC(-gly)) as an adjuvant, as this adjuvant has previously been shown to improve the effectiveness of plasmid-encoded DNA antigens. Methods: Outbred and inbred mouse strains were intranasally immunized with unadjuvanted WIV A/H1N1/SI 2006 or WIV that was formulated with N3 alone. Additional groups were immunized with WIV and N3 adjuvant combined with pFliC(-gly). Homo and heterotypic humoral anti-WIV immune responses were assayed from serum and lung by ELISA and hemagglutination inhibition assay. Homo and heterotypic cellular immune responses to WIV and Influenza A NP were also determined. Results: WIV combined with N3 lipid adjuvant the pFliC(-gly) significantly increased homotypic influenza specific serum antibody responses (>200-fold), increased the IgG2 responses, indicating a mixed Th1/Th2-type immunity, and increased the HAI-titer (>100-fold). Enhanced cell-mediated IFNγ secreting influenza directed CD4+ and CD8+ T cell responses (>40-fold) to homotypic and heterosubtypic influenza A virus and peptides. Long-term and protective immunity was obtained. Conclusions: These results indicate that inactivated influenza virus that was formulated with N3 cationic adjuvant significantly enhanced broad systemic and mucosal influenza specific immune responses. These responses were broadened and further increased by incorporating DNA plasmids encoding FliC from S. typhimurum as an adjuvant providing long lasting protection against heterologous Influenza A/H1N1/CA09pdm virus challenge.
Collapse
|
15
|
Wani SA, Sahu AR, Khan RIN, Pandey A, Saxena S, Hosamani N, Malla WA, Chaudhary D, Kanchan S, Sah V, Rajak KK, Muthuchelvan D, Mishra B, Tiwari AK, Sahoo AP, Sajjanar B, Singh YP, Gandham RK, Mishra BP, Singh RK. Contrasting Gene Expression Profiles of Monocytes and Lymphocytes From Peste-Des-Petits-Ruminants Virus Infected Goats. Front Immunol 2019; 10:1463. [PMID: 31333643 PMCID: PMC6624447 DOI: 10.3389/fimmu.2019.01463] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/10/2019] [Indexed: 01/06/2023] Open
Abstract
In this study, transcriptome analysis of PPRV infected PBMC subsets-T helper cells, T cytotoxic cells, monocytes, and B lymphocytes was done to delineate their role in host response. PPRV was found to infect lymphocytes and not monocytes. The established receptor for PPRV-SLAM was found downregulated in lymphocytes and non-differentially expressed in monocytes. A profound deviation in the global gene expression profile with a large number of unique upregulated genes (851) and downregulated genes (605) was observed in monocytes in comparison to lymphocytes. ISGs-ISG15, Mx1, Mx2, RSAD2, IFIT3, and IFIT5 that play a role in antiviral response and the genes for viral sensors-MDA5, LGP2, and RIG1, were found to be upregulated in lymphocytes and downregulated in monocytes. The transcription factors-IRF-7 and STAT-1 that regulate expression of most of the ISGs were found activated in lymphocytes and not in monocytes. Interferon signaling pathway and RIG1 like receptor signaling pathway were found activated in lymphocytes and not in monocytes. This contrast in gene expression profiles and signaling pathways indicated the predominant role of lymphocytes in generating the antiviral response against PPRV in goats, thus, giving us new insights into host response to PPRV.
Collapse
Affiliation(s)
- Sajad Ahmad Wani
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute (IVRI), Bareilly, India.,Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, OH, United States
| | - Amit Ranjan Sahu
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute (IVRI), Bareilly, India.,Genomics and Computational Biology, DBT-National Institute of Animal Biotechnology, Hyderabad, India
| | - Raja Ishaq Nabi Khan
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute (IVRI), Bareilly, India
| | - Aruna Pandey
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute (IVRI), Bareilly, India
| | - Shikha Saxena
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute (IVRI), Bareilly, India
| | - Neelima Hosamani
- Genomics and Computational Biology, DBT-National Institute of Animal Biotechnology, Hyderabad, India
| | - Waseem Akram Malla
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute (IVRI), Bareilly, India
| | - Dheeraj Chaudhary
- Division of Virology, ICAR-Indian Veterinary Research Institute (IVRI), Mukteswar, India
| | - Sonam Kanchan
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute (IVRI), Bareilly, India
| | - Vaishali Sah
- Division of Animal Genetics and Breeding, ICAR-Indian Veterinary Research Institute (IVRI), Bareilly, India
| | - Kaushal Kishor Rajak
- Division of Biological Products, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - D Muthuchelvan
- Division of Virology, ICAR-Indian Veterinary Research Institute (IVRI), Mukteswar, India
| | - Bina Mishra
- Division of Biological Products, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Ashok Kumar Tiwari
- Division of Biological Standardization, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Aditya P Sahoo
- ICAR- Directorate of Foot and Mouth Disease, Mukteswar, India
| | - Basavaraj Sajjanar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute (IVRI), Bareilly, India
| | - Yash Pal Singh
- ARIS Cell, ICAR-Indian Veterinary Research Institute (IVRI), Bareilly, India
| | - Ravi Kumar Gandham
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute (IVRI), Bareilly, India.,Genomics and Computational Biology, DBT-National Institute of Animal Biotechnology, Hyderabad, India
| | - Bishnu Prasad Mishra
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute (IVRI), Bareilly, India
| | - Raj Kumar Singh
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute (IVRI), Bareilly, India
| |
Collapse
|
16
|
Kurtovic L, Agius PA, Feng G, Drew DR, Ubillos I, Sacarlal J, Aponte JJ, Fowkes FJI, Dobaño C, Beeson JG. Induction and decay of functional complement-fixing antibodies by the RTS,S malaria vaccine in children, and a negative impact of malaria exposure. BMC Med 2019; 17:45. [PMID: 30798787 PMCID: PMC6388494 DOI: 10.1186/s12916-019-1277-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/31/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Leading malaria vaccine, RTS,S, is based on the circumsporozoite protein (CSP) of sporozoites. RTS,S confers partial protection against malaria in children, but efficacy wanes relatively quickly after primary immunization. Vaccine efficacy has some association with anti-CSP IgG; however, it is unclear how these antibodies function, and how functional antibodies are induced and maintained over time. Recent studies identified antibody-complement interactions as a potentially important immune mechanism against sporozoites. Here, we investigated whether RTS,S vaccine-induced antibodies could function by interacting with complement. METHODS Serum samples were selected from children in a phase IIb trial of RTS,S/AS02A conducted at two study sites of high and low malaria transmission intensity in Manhiça, Mozambique. Samples following primary immunization and 5-year post-immunization follow-up time points were included. Vaccine-induced antibodies were characterized by isotype, subclass, and epitope specificity, and tested for the ability to fix and activate complement. We additionally developed statistical methods to model the decay and determinants of functional antibodies after vaccination. RESULTS RTS,S vaccination induced anti-CSP antibodies that were mostly IgG1, with some IgG3, IgG2, and IgM. Complement-fixing antibodies were effectively induced by vaccination, and targeted the central repeat and C-terminal regions of CSP. Higher levels of complement-fixing antibodies were associated with IgG that equally recognized both the central repeat and C-terminal regions of CSP. Older age and higher malaria exposure were significantly associated with a poorer induction of functional antibodies. There was a marked decay in functional complement-fixing antibodies within months after vaccination, as well as decays in IgG subclasses and IgM. Statistical modeling suggested the decay in complement-fixing antibodies was mostly attributed to the waning of anti-CSP IgG1, and to a lesser extent IgG3. CONCLUSIONS We demonstrate for the first time that RTS,S can induce complement-fixing antibodies in young malaria-exposed children. The short-lived nature of functional responses mirrors the declining vaccine efficacy of RTS,S over time. The negative influence of age and malaria exposure on functional antibodies has implications for understanding vaccine efficacy in different settings. These findings provide insights into the mechanisms and longevity of vaccine-induced immunity that will help inform the future development of highly efficacious and long-lasting malaria vaccines.
Collapse
Affiliation(s)
- Liriye Kurtovic
- Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Paul A Agius
- Burnet Institute, Melbourne, Australia.,Department of Epidemiology and Preventative Medicine, Monash University, Melbourne, Australia
| | | | | | - Itziar Ubillos
- ISGlobal, Hospital Clínic Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Jahit Sacarlal
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique.,Faculdade de Medicina, Universidade Eduardo Mondlane (UEM), Maputo, Mozambique
| | - John J Aponte
- ISGlobal, Hospital Clínic Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| | - Freya J I Fowkes
- Burnet Institute, Melbourne, Australia.,Department of Epidemiology and Preventative Medicine, Monash University, Melbourne, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| | - James G Beeson
- Burnet Institute, Melbourne, Australia. .,Department of Immunology and Pathology, Monash University, Melbourne, Australia. .,Department of Microbiology, Monash University, Clayton, Australia. .,Department of Medicine, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
17
|
Sato K, Takahashi Y, Adachi Y, Asanuma H, Ato M, Tashiro M, Itamura S. Efficient protection of mice from influenza A/H1N1pdm09 virus challenge infection via high avidity serum antibodies induced by booster immunizations with inactivated whole virus vaccine. Heliyon 2019; 5:e01113. [PMID: 30623129 PMCID: PMC6319303 DOI: 10.1016/j.heliyon.2018.e01113] [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] [Received: 10/24/2018] [Revised: 12/27/2018] [Accepted: 12/27/2018] [Indexed: 12/17/2022] Open
Abstract
The immunogenicities of inactivated whole and split virus vaccines derived from influenza A/H1N1pdm09 virus were compared in a mouse model. We demonstrated the unique properties of whole virus vaccine boosters on the serum memory antibody response in mice. Consistent with previous studies, booster immunization with either whole or split virus vaccines of A/H1N1pdm09 virus produced comparable titers of serum antibodies with hemagglutination inhibition and virus-neutralizing activities. However, superior protection against the challenge infection was unexpectedly observed in mice primed and boosted with whole virus vaccines compared with those treated with split virus vaccines, despite similar levels of antibody titers in each group. Immune serum antibodies were shown to be primarily responsible for this protection via passive transfer experiments of immune serum antibodies to naive recipient mice. Moreover, this protection correlated with elevated affinity maturation of the antibodies. Thus, booster immunization with whole virus vaccines elicited a robust serum antibody response with high avidity to the virus, which was not measurable via conventional serological assays.
Collapse
Affiliation(s)
- Kayoko Sato
- Influenza Virus Research Center, National Institute of Infectious Diseases, Musashimurayama-shi, Tokyo, Japan
| | - Yoshimasa Takahashi
- Department of Immunology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Yu Adachi
- Department of Immunology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Hideki Asanuma
- Influenza Virus Research Center, National Institute of Infectious Diseases, Musashimurayama-shi, Tokyo, Japan
| | - Manabu Ato
- Department of Immunology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Masato Tashiro
- Influenza Virus Research Center, National Institute of Infectious Diseases, Musashimurayama-shi, Tokyo, Japan
| | - Shigeyuki Itamura
- Influenza Virus Research Center, National Institute of Infectious Diseases, Musashimurayama-shi, Tokyo, Japan
| |
Collapse
|
18
|
Rowell J, Lo CY, Price GE, Misplon JA, Epstein SL, Garcia M. Conventional influenza vaccines influence the performance of a universal influenza vaccine in mice. Vaccine 2017; 36:1008-1015. [PMID: 29249542 DOI: 10.1016/j.vaccine.2017.11.065] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/03/2017] [Accepted: 11/22/2017] [Indexed: 01/10/2023]
Abstract
Universal influenza vaccines are designed to protect against diverse strains of influenza virus. Preclinical testing of new vaccine candidates is usually done in naïve animals, despite intended use in the human population with its varied immune history including responses to previous vaccinations. As an approach more relevant to human use, we tested a candidate universal influenza vaccine in mice with a history of conventional vaccination. Female BALB/c mice were given two intramuscular doses of inactivated influenza vaccine (IIV) or diphtheria and tetanus toxoids vaccine (DT), one month apart. Another group was given two intranasal doses of live attenuated influenza virus (LAIV). One month after the second dose, mice were given the universal influenza vaccine: recombinant adenoviruses expressing influenza A nucleoprotein (A/NP) and matrix 2 (M2) (A/NP + M2-rAd). Immune responses to universal vaccine antigens A/NP and M2 were assessed by ELISA and interferon-γ ELISPOT. Protection was tested by challenge with mouse-adapted A/FM/1/47 (H1N1) and monitoring for weight loss and survival. Universal vaccine performance was enhanced, inhibited or unaffected by particular prior vaccinations. Mice given Afluria IIV and LAIV had greater antibody and T-cell response to A/NP than mice without prior vaccination, providing examples of enhanced A/NP + M2-rAd performance. Though Fluvirin IIV partially inhibited, the universal vaccine still provided considerable protection unlike conventional vaccination. Fluzone IIV and DT had no effect on A/NP + M2-rAd performance. Thus our results demonstrate that universal vaccine candidate A/NP + M2-rAd was at least partially effective in mice with diverse prior histories. However, the degree of protection and nature of the immune responses may be affected by a history of conventional vaccination and suggests that performance in humans would be influenced by immune history.
Collapse
Affiliation(s)
- Janelle Rowell
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993-0002, USA
| | - Chia-Yun Lo
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993-0002, USA
| | - Graeme E Price
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993-0002, USA
| | - Julia A Misplon
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993-0002, USA
| | - Suzanne L Epstein
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993-0002, USA.
| | - Mayra Garcia
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993-0002, USA
| |
Collapse
|
19
|
Corti D, Cameroni E, Guarino B, Kallewaard NL, Zhu Q, Lanzavecchia A. Tackling influenza with broadly neutralizing antibodies. Curr Opin Virol 2017; 24:60-69. [PMID: 28527859 PMCID: PMC7102826 DOI: 10.1016/j.coviro.2017.03.002] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/02/2017] [Accepted: 03/09/2017] [Indexed: 11/28/2022]
Abstract
Monoclonal antibodies have revolutionized the treatment of several human diseases, including cancer, autoimmunity and inflammatory conditions and represent a new frontier for the treatment of infectious diseases. In the last decade, new methods have allowed the efficient interrogation of the human antibody repertoire from influenza immune individuals and the isolation of several monoclonal antibodies capable of dealing with the high variability of influenza viruses. Here, we will provide a comprehensive overview of the specificity, antiviral and immunological mechanisms of action and development into the clinic of broadly reactive monoclonal antibodies against influenza A and B viruses.
Collapse
Affiliation(s)
| | | | | | | | - Qing Zhu
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA
| | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| |
Collapse
|
20
|
Broadly Reactive Anti-Respiratory Syncytial Virus G Antibodies from Exposed Individuals Effectively Inhibit Infection of Primary Airway Epithelial Cells. J Virol 2017; 91:JVI.02357-16. [PMID: 28275185 DOI: 10.1128/jvi.02357-16] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/27/2017] [Indexed: 11/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) causes severe respiratory disease in young children. Antibodies specific for the RSV prefusion F protein have guided RSV vaccine research, and in human serum, these antibodies contribute to >90% of the neutralization response; however, detailed insight into the composition of the human B cell repertoire against RSV is still largely unknown. In order to study the B cell repertoire of three healthy donors for specificity against RSV, CD27+ memory B cells were isolated and immortalized using BCL6 and Bcl-xL. Of the circulating memory B cells, 0.35% recognized RSV-A2-infected cells, of which 59% were IgA-expressing cells and 41% were IgG-expressing cells. When we generated monoclonal B cells selected for high binding to RSV-infected cells, 44.5% of IgG-expressing B cells and 56% of IgA-expressing B cells reacted to the F protein, while, unexpectedly, 41.5% of IgG-expressing B cells and 44% of IgA expressing B cells reacted to the G protein. Analysis of the G-specific antibodies revealed that 4 different domains on the G protein were recognized. These epitopes predicted cross-reactivity between RSV strain A (RSV-A) and RSV-B and matched the potency of antibodies to neutralize RSV in HEp-2 cells and in primary epithelial cell cultures. G-specific antibodies were also able to induce antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis of RSV-A2-infected cells. However, these processes did not seem to depend on a specific epitope. In conclusion, healthy adults harbor a diverse repertoire of RSV glycoprotein-specific antibodies with a broad range of effector functions that likely play an important role in antiviral immunity.IMPORTANCE Human RSV remains the most common cause of severe lower respiratory tract disease in premature babies, young infants, the elderly, and immunocompromised patients and plays an important role in asthma exacerbations. In developing countries, RSV lower respiratory tract disease has a high mortality. Without an effective vaccine, only passive immunization with palivizumab is approved for prophylactic treatment. However, highly potent RSV-specific monoclonal antibodies could potentially serve as a therapeutic treatment and contribute to disease control and mortality reduction. In addition, these antibodies could guide further vaccine development. In this study, we isolated and characterized several novel antibodies directed at the RSV G protein. This information can add to our understanding and treatment of RSV disease.
Collapse
|
21
|
Zeng W, Tan ACL, Horrocks K, Jackson DC. A lipidated form of the extracellular domain of influenza M2 protein as a self-adjuvanting vaccine candidate. Vaccine 2015; 33:3526-32. [PMID: 26049002 DOI: 10.1016/j.vaccine.2015.05.053] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 05/18/2015] [Accepted: 05/21/2015] [Indexed: 12/15/2022]
Abstract
The highly conserved extracellular domain of Matrix protein 2 (M2e) of influenza A virus has been previously investigated as a potential target for an universal influenza vaccine. In this study we prepared four lipopeptide influenza vaccine candidates in which the TLR2 agonist S-[2,3-bis(palmitoyloxy)propyl] cysteine, (Pam2Cys) was attached to either the N- or C-terminus of the M2e consensus sequence SLLTEVETPIRNEWGCRCNDSSDP and its analogue sequence with the two cysteine residues replaced with serine residues. The results of animal study show that each of these lipopeptides induced strong M2e-specific antibody responses in the absence of extraneous T helper cell epitope(s) which are normally incorporated in the previous studies or addition of extraneous adjuvant and that these antibodies are protective against lethal challenge with influenza virus. Comparison of different routes of inoculation demonstrated that intranasal administration of M2e lipopeptide induced higher titers of IgA and IgG2b antibodies in the bronchoalveolar lavage than did subcutaneous vaccination and was better at mitigating the severity of viral challenge. Finally, we show that anti-M2e antibody specificities absent from the antibody repertoire elicited by a commercially available influenza vaccine and by virus infection can be introduced by immunization with M2e-lipopeptide and boosted by viral challenge. Immunization with this lipidated form of the M2e epitope therefore offers a means of using a widely conserved epitope to generate protective antibodies which are not otherwise induced.
Collapse
Affiliation(s)
- Weiguang Zeng
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne 3010, Victoria, Australia.
| | - Amabel C L Tan
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne 3010, Victoria, Australia
| | - Kylie Horrocks
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne 3010, Victoria, Australia
| | - David C Jackson
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne 3010, Victoria, Australia
| |
Collapse
|
22
|
Boyle MJ, Reiling L, Feng G, Langer C, Osier FH, Aspeling-Jones H, Cheng YS, Stubbs J, Tetteh KKA, Conway DJ, McCarthy JS, Muller I, Marsh K, Anders RF, Beeson JG. Human antibodies fix complement to inhibit Plasmodium falciparum invasion of erythrocytes and are associated with protection against malaria. Immunity 2015; 42:580-90. [PMID: 25786180 PMCID: PMC4372259 DOI: 10.1016/j.immuni.2015.02.012] [Citation(s) in RCA: 226] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 12/01/2014] [Accepted: 02/23/2015] [Indexed: 11/26/2022]
Abstract
Antibodies play major roles in immunity to malaria; however, a limited understanding of mechanisms mediating protection is a major barrier to vaccine development. We have demonstrated that acquired human anti-malarial antibodies promote complement deposition on the merozoite to mediate inhibition of erythrocyte invasion through C1q fixation and activation of the classical complement pathway. Antibody-mediated complement-dependent (Ab-C′) inhibition was the predominant invasion-inhibitory activity of human antibodies; most antibodies were non-inhibitory without complement. Inhibitory activity was mediated predominately via C1q fixation, and merozoite surface proteins 1 and 2 were identified as major targets. Complement fixation by antibodies was very strongly associated with protection from both clinical malaria and high-density parasitemia in a prospective longitudinal study of children. Ab-C′ inhibitory activity could be induced by human immunization with a candidate merozoite surface-protein vaccine. Our findings demonstrate that human anti-malarial antibodies have evolved to function by fixing complement for potent invasion-inhibitory activity and protective immunity. Antibodies function with complement to inhibit P. falciparum replication Antibodies fix C1q to block invasion and lyse merozoites Complement-fixing antibodies are strongly associated with immunity in children Antibody-complement inhibition can be induced by human vaccination
Collapse
Affiliation(s)
- Michelle J Boyle
- The Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC 3004, Australia; Department of Medical Biology, University of Melbourne, Royal Parade, Melbourne, VIC 3010, Australia
| | - Linda Reiling
- The Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC 3004, Australia
| | - Gaoqian Feng
- The Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC 3004, Australia
| | - Christine Langer
- The Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC 3004, Australia
| | - Faith H Osier
- Centre for Geographic Medicine Research, Kenya Medical Research Institute, Coast, PO Box 230, 80108 Kilifi, Kenya
| | | | - Yik Sheng Cheng
- The Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC 3004, Australia; Department of Medical Biology, University of Melbourne, Royal Parade, Melbourne, VIC 3010, Australia
| | - Janine Stubbs
- The Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC 3004, Australia
| | - Kevin K A Tetteh
- London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E7HT, UK
| | - David J Conway
- London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E7HT, UK
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, University of Queensland, 300 Herston Road, Herston, QLD 4006, Australia
| | - Ivo Muller
- Walter and Eliza Hall Institute, Royal Parade, Melbourne, VIC 3050, Australia
| | - Kevin Marsh
- Centre for Geographic Medicine Research, Kenya Medical Research Institute, Coast, PO Box 230, 80108 Kilifi, Kenya
| | - Robin F Anders
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - James G Beeson
- The Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC 3004, Australia; Department of Microbiology, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
23
|
Ambati A, Valentini D, Montomoli E, Lapini G, Biuso F, Wenschuh H, Magalhaes I, Maeurer M. H1N1 viral proteome peptide microarray predicts individuals at risk for H1N1 infection and segregates infection versus Pandemrix(®) vaccination. Immunology 2015; 145:357-66. [PMID: 25639813 PMCID: PMC4479535 DOI: 10.1111/imm.12448] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/02/2015] [Accepted: 01/26/2015] [Indexed: 12/30/2022] Open
Abstract
A high content peptide microarray containing the entire influenza A virus [A/California/08/2009(H1N1)] proteome and haemagglutinin proteins from 12 other influenza A subtypes, including the haemagglutinin from the [A/South Carolina/1/1918(H1N1)] strain, was used to gauge serum IgG epitope signatures before and after Pandemrix® vaccination or H1N1 infection in a Swedish cohort during the pandemic influenza season 2009. A very narrow pattern of pandemic flu-specific IgG epitope recognition was observed in the serum from individuals who later contracted H1N1 infection. Moreover, the pandemic influenza infection generated IgG reactivity to two adjacent epitopes of the neuraminidase protein. The differential serum IgG recognition was focused on haemagglutinin 1 (H1) and restricted to classical antigenic sites (Cb) in both the vaccinated controls and individuals with flu infections. We further identified a novel epitope VEPGDKITFEATGNL on the Ca antigenic site (251–265) of the pandemic flu haemagglutinin, which was exclusively recognized in serum from individuals with previous vaccinations and never in serum from individuals with H1N1 infection (confirmed by RNA PCR analysis from nasal swabs). This epitope was mapped to the receptor-binding domain of the influenza haemagglutinin and could serve as a correlate of immune protection in the context of pandemic flu. The study shows that unbiased epitope mapping using peptide microarray technology leads to the identification of biologically and clinically relevant target structures. Most significantly an H1N1 infection induced a different footprint of IgG epitope recognition patterns compared with the pandemic H1N1 vaccine.
Collapse
Affiliation(s)
- Aditya Ambati
- Therapeutic Immunology Unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - Davide Valentini
- Therapeutic Immunology Unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Guilia Lapini
- VisMederi srl, Enterprise in Life Science, Siena, Italy
| | | | | | - Isabelle Magalhaes
- Therapeutic Immunology Unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - Markus Maeurer
- Therapeutic Immunology Unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
24
|
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
|
25
|
Seasonal influenza vaccination is the strongest correlate of cross-reactive antibody responses in migratory bird handlers. mBio 2014; 5:e02107. [PMID: 25491354 PMCID: PMC4324241 DOI: 10.1128/mbio.02107-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Avian species are reservoirs of influenza A viruses and could harbor viruses with significant pandemic potential. We examined the antibody and cellular immune responses to influenza A viruses in field or laboratory workers with a spectrum of occupational exposure to avian species for evidence of zoonotic infections. We measured the seroprevalence and T cell responses among 95 individuals with various types and degrees of prior field or laboratory occupational exposure to wild North American avian species using whole blood samples collected in 2010. Plasma samples were tested using endpoint enzyme-linked immunosorbent assay (ELISA) and hemagglutination (HA) inhibition (HAI) assays to subtypes H3, H4, H5, H6, H7, H8, and H12 proteins. Detectable antibodies were found against influenza HA antigens in 77% of individuals, while 65% of individuals tested had measurable T cell responses (gamma interferon [IFN-γ] enzyme-linked immunosorbent spot assay [ELISPOT]) to multiple HA antigens of avian origin. To begin defining the observed antibody specificities, Spearman rank correlation analysis showed that ELISA responses, which measure both head- and stalk-binding antibodies, do not predict HAI reactivities, which measure primarily head-binding antibodies. This result suggests that ELISA titers can report cross-reactivity based on the levels of non-head-binding responses. However, the strongest positive correlate of HA-specific ELISA antibody titers was receipt of seasonal influenza virus vaccination. Occupational exposure was largely uncorrelated with serological measures, with the exception of individuals exposed to poultry, who had higher levels of H7-specific antibodies than non-poultry-exposed individuals. While the cohort had antibody and T cell reactivity to a broad range of influenza viruses, only occupational exposure to poultry was associated with a significant difference in antibody levels to a specific subtype (H7). There was no evidence that T cell assays provided greater specificity for the detection of zoonotic infection. However, influenza vaccination appears to promote cross-reactive antibodies and may provide enhanced protection to novel influenza viruses. IMPORTANCE Annual vaccinations are necessary to ameliorate influenza disease due to drifted viral variants that emerge in the population. Major shifts in the antigenicity of influenza viruses can result in immunologically distinct viruses that can cause more severe disease in humans. Historically, genetic reassortment between avian, swine, or human influenza viruses has caused influenza pandemics in humans several times in the last century. Therefore, it is important to design vaccines to elicit broad protective responses to influenza infections. Because avian influenza viruses have an important role in emerging infections, we tested whether occupational exposure to birds can elicit immune responses to avian influenza viruses in humans. Instead of a specific occupational exposure, the strongest association of enhanced cross-reactive antibody responses was receipt of seasonal influenza vaccination. Therefore, individuals with preexisting immune responses to seasonal human influenza viruses have substantial cross-reactive antibody and T cell responses that may lead to enhanced protection to novel influenza viruses.
Collapse
|
26
|
Schmeisser F, Vasudevan A, Soto J, Kumar A, Williams O, Weir JP. A monoclonal antibody-based immunoassay for measuring the potency of 2009 pandemic influenza H1N1 vaccines. Influenza Other Respir Viruses 2014; 8:587-95. [PMID: 25087462 PMCID: PMC4181825 DOI: 10.1111/irv.12272] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2014] [Indexed: 12/03/2022] Open
Abstract
Background The potency of inactivated influenza vaccines is determined using a single radial immunodiffusion (SRID) assay. This assay is relatively easy to standardize, it is not technically demanding, and it is capable of measuring the potency of several vaccine strain subtypes in a multivalent vaccine. Nevertheless, alternative methods that retain the major advantages of the SRID, but with a greater dynamic range of measurement and with reduced reagent requirements, are needed. Objectives The feasibility of an ELISA-based assay format was explored as an alternative potency assay for inactivated influenza vaccines. Methods Several murine monoclonal antibodies (mAbs), specific for the 2009 pandemic H1N1 influenza virus hemagglutinin (HA), were evaluated for their potential to capture and quantify HA antigen. Vaccine samples, obtained from four licensed influenza vaccine manufacturers, included monovalent bulk vaccine, monovalent vaccine, and trivalent vaccine. Traditional SRID potency assays were run in parallel with the mAb–ELISA potency assay using the reference antigen standard appropriate for the vaccine samples being tested. Results The results indicated that the ELISA potency assay can quantify HA over a wide range of concentrations, including vaccine at subpotent doses, and the ELISA and SRID potency values correlated well for most vaccine samples. Importantly, the assay was capable of quantifying A/California HA in a trivalent formulation. Conclusions This study demonstrates the general feasibility of the mAb approach and strongly suggests that such ELISAs have potential for continued development as an alternative method to assay the potency of inactivated influenza vaccines.
Collapse
Affiliation(s)
- Falko Schmeisser
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
27
|
Marinho CF, Azeredo EL, Torrentes-Carvalho A, Marins-Dos-Santos A, Kubelka CF, de Souza LJ, Cunha RV, de-Oliveira-Pinto LM. Down-regulation of complement receptors on the surface of host monocyte even as in vitro complement pathway blocking interferes in dengue infection. PLoS One 2014; 9:e102014. [PMID: 25061945 PMCID: PMC4111305 DOI: 10.1371/journal.pone.0102014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 06/13/2014] [Indexed: 12/16/2022] Open
Abstract
In dengue virus (DENV) infection, complement system (CS) activation appears to have protective and pathogenic effects. In severe dengue fever (DF), the levels of DENV non-structural-1 protein and of the products of complement activation, including C3a, C5a and SC5b-9, are higher before vascular leakage occurs, supporting the hypothesis that complement activation contributes to unfavourable outcomes. The clinical manifestations of DF range from asymptomatic to severe and even fatal. Here, we aimed to characterise CS by their receptors or activation product, in vivo in DF patients and in vitro by DENV-2 stimulation on monocytes. In comparison with healthy controls, DF patients showed lower expression of CR3 (CD11b), CR4 (CD11c) and, CD59 on monocytes. The DF patients who were high producers of SC5b-9 were also those that showed more pronounced bleeding or vascular leakage. Those findings encouraged us to investigate the role of CS in vitro, using monocytes isolated from healthy subjects. Prior blocking with CR3 alone (CD11b) or CR3 (CD11b/CD18) reduced viral infection, as quantified by the levels of intracellular viral antigen expression and soluble DENV non-structural viral protein. However, we found that CR3 alone (CD11b) or CR3 (CD11b/CD18) blocking did not influence major histocompatibility complex presentation neither active caspase-1 on monocytes, thus probably ruling out inflammasome-related mechanisms. Although it did impair the secretion of tumour necrosis factor alpha and interferon alpha. Our data provide strategies of blocking CR3 (CD11b) pathways could have implications for the treatment of viral infection by antiviral-related mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rivaldo Venâncio Cunha
- Department of Clinical Medicine, Universidade Federal do Mato Grosso do Sul, Campo Grande, Brazil
| | | |
Collapse
|
28
|
Mayer AE, Johnson JB, Parks GD. The neutralizing capacity of antibodies elicited by parainfluenza virus infection of African Green Monkeys is dependent on complement. Virology 2014; 460-461:23-33. [PMID: 25010267 DOI: 10.1016/j.virol.2014.05.004] [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: 02/25/2014] [Revised: 03/29/2014] [Accepted: 05/04/2014] [Indexed: 11/27/2022]
Abstract
The African Green Monkey (AGM) model was used to analyze the role of complement in neutralization of parainfluenza virus. Parainfluenza virus 5 (PIV5) and human parainfluenza virus type 2 were effectively neutralized in vitro by naïve AGM sera, but neutralizing capacity was lost by heat-inactivation. The mechanism of neutralization involved formation of massive aggregates, with no evidence of virion lysis. Following inoculation of the respiratory tract with a PIV5 vector expressing HIV gp160, AGM produced high levels of serum and tracheal antibodies against gp120 and the viral F and HN proteins. However, in the absence of complement these anti-PIV5 antibodies had very poor neutralizing capacity. Virions showed extensive deposition of IgG and C1q with post- but not pre-immune sera. These results highlight the importance of complement in the initial antibody response to parainfluenza viruses, with implications for understanding infant immune responses and design of vaccine strategies for these pediatric pathogens.
Collapse
Affiliation(s)
- Anne E Mayer
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - John B Johnson
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Griffith D Parks
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| |
Collapse
|
29
|
Keating R, Hertz T, Wehenkel M, Harris TL, Edwards BA, McClaren JL, Brown SA, Surman S, Wilson ZS, Bradley P, Hurwitz J, Chi H, Doherty PC, Thomas PG, McGargill MA. The kinase mTOR modulates the antibody response to provide cross-protective immunity to lethal infection with influenza virus. Nat Immunol 2013; 14:1266-76. [PMID: 24141387 PMCID: PMC3883080 DOI: 10.1038/ni.2741] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 09/13/2013] [Indexed: 12/13/2022]
Abstract
Highly pathogenic avian influenza viruses pose a continuing global threat. Current vaccines will not protect against newly evolved pandemic viruses. The creation of 'universal' vaccines has been unsuccessful because the immunological mechanisms that promote heterosubtypic immunity are incompletely defined. We found here that rapamycin, an immunosuppressive drug that inhibits the kinase mTOR, promoted cross-strain protection against lethal infection with influenza virus of various subtypes when administered during immunization with influenza virus subtype H3N2. Rapamycin reduced the formation of germinal centers and inhibited class switching in B cells, which yielded a unique repertoire of antibodies that mediated heterosubtypic protection. Our data established a requirement for the mTORC1 complex in B cell class switching and demonstrated that rapamycin skewed the antibody response away from high-affinity variant epitopes and targeted more conserved elements of hemagglutinin. Our findings have implications for the design of a vaccine against influenza virus.
Collapse
Affiliation(s)
- Rachael Keating
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Rimmelzwaan GF, Katz JM. Immune responses to infection with H5N1 influenza virus. Virus Res 2013; 178:44-52. [PMID: 23735534 DOI: 10.1016/j.virusres.2013.05.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 04/03/2013] [Accepted: 05/20/2013] [Indexed: 01/22/2023]
Abstract
Influenza A H5N1 viruses remain a substantial threat to global public health. In particular, the expanding genetic diversity of H5N1 viruses and the associated risk for human adaptation underscore the importance of better understanding host immune responses that may protect against disease or infection. Although much emphasis has been placed on investigating early virus-host interactions and the induction of innate immune responses, little is known of the consequent adaptive immune response to H5N1 virus infection. In this review, we describe the H5N1 virus-specific and cross-reactive antibody and T cell responses in humans and animal models. Data from limited studies suggest that although initially robust, there is substantial waning of the serum antibody responses in survivors of H5N1 virus infection. Characterization of monoclonal antibodies generated from memory B cells of survivors of H5N1 virus infection has provided an understanding of the fine specificity of the human antibody response to H5N1 virus infection and identified strategies for immunotherapy. Human T cell responses induced by infection with seasonal influenza viruses are directed to relatively conserved internal proteins and cross-react with the H5N1 subtype. A role for T cell-based heterosubtypic immunity against H5N1 viruses is suggested in animal studies. Further studies on adaptive immune responses to H5N1 virus infection in both humans and animals are needed to inform the design of optimal immunological treatment and prevention modalities.
Collapse
Affiliation(s)
- Guus F Rimmelzwaan
- Viroscience Laboratory, Erasmus Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
31
|
Yamamoto T, Mizoguchi Y, Kaneno H, Yamamoto K, Inoue Y, Kawashima H, Kase T, Shimotsuji T. Serum immunoglobulin G subclass levels and estimated clinical severity caused by possible influenza A (H1N1) pdm 2009 infection. J Infect Chemother 2013; 19:833-42. [PMID: 23467791 DOI: 10.1007/s10156-013-0570-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 02/03/2013] [Indexed: 11/27/2022]
Abstract
The clinical severity of the 2009 pandemic H1N1 influenza (H1N1 pdm09) was thought to be related to the difference between the amount of viral load and condition of the host immune response. We investigated the role of serum levels of IgG and its subclasses in clinical severity using the data from 45 child inpatients suffering from bronchitis or mild pneumonia caused by possible H1N1 pdm09 (pH1N1 pdm09) infection. After selecting parameters for serum IgG subclasses and logarithmically transformed urinary beta-2 microglobulin/creatinine (b2MG/Cr) values and admission duration, we performed path analysis using a mean covariance structure equation analysis to investigate the relationship between the clinical severity and the foregoing selected parameters. Total path analyses using a Bayesian method revealed that the estimated clinical severity caused by pH1N1 pdm09 was positively associated with maximal respiration rates, admission duration, and log urinary b2MG/Cr levels, whereas negatively associated with serum IgG, IgG1, IgG2, and IgG3 levels, duration of neuraminidase inhibitor therapy in outpatient clinics, and age. Serum IgG and its subclasses (IgG1-IgG3) reduced estimated clinical severity in children with pH1N1 pdm09 infection.
Collapse
Affiliation(s)
- Takehisa Yamamoto
- Department of Pediatrics, Minoh City Hospital, 5-7-1 Kayano, Minoh, Osaka, 562-8562, Japan,
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Benhnia MREI, Maybeno M, Blum D, Aguilar-Sino R, Matho M, Meng X, Head S, Felgner PL, Zajonc DM, Koriazova L, Kato S, Burton DR, Xiang Y, Crowe JE, Peters B, Crotty S. Unusual features of vaccinia virus extracellular virion form neutralization resistance revealed in human antibody responses to the smallpox vaccine. J Virol 2013; 87:1569-85. [PMID: 23152530 PMCID: PMC3554146 DOI: 10.1128/jvi.02152-12] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 11/07/2012] [Indexed: 11/20/2022] Open
Abstract
The extracellular virion form (EV) of vaccinia virus (VACV) is essential for viral pathogenesis and is difficult to neutralize with antibodies. Why this is the case and how the smallpox vaccine overcomes this challenge remain incompletely understood. We previously showed that high concentrations of anti-B5 antibodies are insufficient to directly neutralize EV (M. R. Benhnia, et al., J. Virol. 83:1201-1215, 2009). This allowed for at least two possible interpretations: covering the EV surface is insufficient for neutralization, or there are insufficient copies of B5 to allow anti-B5 IgG to cover the whole surface of EV and another viral receptor protein remains active. We endeavored to test these possibilities, focusing on the antibody responses elicited by immunization against smallpox. We tested whether human monoclonal antibodies (MAbs) against the three major EV antigens, B5, A33, and A56, could individually or together neutralize EV. While anti-B5 or anti-A33 (but not anti-A56) MAbs of appropriate isotypes were capable of neutralizing EV in the presence of complement, a mixture of anti-B5, anti-A33, and anti-A56 MAbs was incapable of directly neutralizing EV, even at high concentrations. This remained true when neutralizing the IHD-J strain, which lacks a functional version of the fourth and final known EV surface protein, A34. These immunological data are consistent with the possibility that viral proteins may not be the active component of the EV surface for target cell binding and infectivity. We conclude that the protection afforded by the smallpox vaccine anti-EV response is predominantly mediated not by direct neutralization but by isotype-dependent effector functions, such as complement recruitment for antibodies targeting B5 and A33.
Collapse
Affiliation(s)
| | | | - David Blum
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Rowena Aguilar-Sino
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, USA
| | - Michael Matho
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology (LIAI), La Jolla, California, USA
| | - Xiangzhi Meng
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Steven Head
- DNA Array Core Facility and Consortium for Functional Glycomics, The Scripps Research Institute, La Jolla, California, USA
| | - Philip L. Felgner
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, California, USA
| | - Dirk M. Zajonc
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology (LIAI), La Jolla, California, USA
| | | | | | - Dennis R. Burton
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, USA
| | - Yan Xiang
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, Texas, USA
| | - James E. Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | |
Collapse
|
33
|
Abstract
A fascinating aspect of viral evolution relates to the ability of viruses to escape the adaptive immune response. The widely held view has been that the great variability of viral glycoproteins would be an absolute obstacle to the development of antibody-based therapies or vaccines that could confer broad and long-lasting protection. In the past five years, new approaches have been developed to interrogate human memory B cells and plasma cells with high efficiency and to isolate several broadly neutralizing antiviral antibodies against highly variable pathogens such as HIV-1 and influenza virus. These antibodies not only provide new tools for prophylaxis and therapy for viral diseases but also identify conserved epitopes that may be used to design new vaccines capable of conferring broader protection.
Collapse
Affiliation(s)
- Davide Corti
- Institute for Research in Biomedicine IRB, 6500 Bellinzona, Switzerland.
| | | |
Collapse
|
34
|
Schmeisser F, Friedman R, Besho J, Lugovtsev V, Soto J, Wang W, Weiss C, Williams O, Xie H, Ye Z, Weir JP. Neutralizing and protective epitopes of the 2009 pandemic influenza H1N1 hemagglutinin. Influenza Other Respir Viruses 2012; 7:480-90. [PMID: 23122228 PMCID: PMC5779835 DOI: 10.1111/irv.12029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
AIMS AND METHODS To facilitate antigenic characterization of the influenza A 2009 pandemic H1N1 [A(H1N1)pdm09] hemagglutinin (HA), we generated a panel of murine monoclonal antibodies (mAbs) using as the immunogen mammalian-derived virus-like particles containing the HA of the A/California/04/2009 virus. The antibodies were specific for the A/California/04/2009 HA, and individual mAbs suitable for use in several practical applications including ELISA, immunofluorescence, and Western blot analysis were identified. RESULTS AND CONCLUSIONS As the panel of mAbs included antibodies with hemagglutination inhibition (HI) and virus neutralizing activities, this allowed identification and characterization of potentially important antigenic and neutralizing epitopes of the A/California/04/2009 HA and comparison of those epitopes with the HAs of other influenza viruses including seasonal H1N1 viruses as well as the A/South Carolina/1918 and A/New Jersey/1976 H1N1 viruses. Three mAbs with the highest HI and neutralizing titers were able to provide passive protection against virus challenge. Two other mAbs without HI or neutralizing activities were able to provide partial protection against challenge. HA epitopes recognized by the strongest neutralizing mAbs in the panel were identified by isolation and selection of virus escape mutants in the presence of individual mAbs. Cloned viruses resistant to HI and antibody neutralization were sequenced to identify mutations, and two unique mutations (D127E and G155E) were identified, both near the antigenic site Sa. Using human post-vaccination sera, however, there were no differences in HI titer between A/California/04/2009 and either escape mutant, suggesting that these single mutations were not sufficient to abrogate a protective antibody response to the vaccine.
Collapse
Affiliation(s)
- Falko Schmeisser
- Division of Viral Products, Center for Biologics Evaluations and Research, Food and Drug Administration, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Initiation and regulation of complement during hemolytic transfusion reactions. Clin Dev Immunol 2012; 2012:307093. [PMID: 23118779 PMCID: PMC3479954 DOI: 10.1155/2012/307093] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Accepted: 09/07/2012] [Indexed: 11/19/2022]
Abstract
Hemolytic transfusion reactions represent one of the most common causes of transfusion-related mortality. Although many factors influence hemolytic transfusion reactions, complement activation represents one of the most common features associated with fatality. In this paper we will focus on the role of complement in initiating and regulating hemolytic transfusion reactions and will discuss potential strategies aimed at mitigating or favorably modulating complement during incompatible red blood cell transfusions.
Collapse
|
36
|
Dong L, Liu F, Fairman J, Hong DK, Lewis DB, Monath T, Warner JF, Belser JA, Patel J, Hancock K, Katz JM, Lu X. Cationic liposome-DNA complexes (CLDC) adjuvant enhances the immunogenicity and cross-protective efficacy of a pre-pandemic influenza A H5N1 vaccine in mice. Vaccine 2011; 30:254-64. [PMID: 22085545 DOI: 10.1016/j.vaccine.2011.10.103] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 10/27/2011] [Accepted: 10/31/2011] [Indexed: 12/22/2022]
Abstract
The development of pre-pandemic influenza A H5N1 vaccines that confer both antigen-sparing and cross-clade protection are a high priority given the limited worldwide capacity for influenza vaccine production, and the antigenic and genetic heterogeneity of circulating H5N1 viruses. The inclusion of potent adjuvants in vaccine formulations may achieve both of these aims. Here we show that the addition of JVRS-100, an adjuvant consisting of cationic liposome-DNA complexes (CLDC) to a clade 1-derived H5N1 split vaccine induced significantly higher virus-specific antibody than unadjuvanted formulations, with a >30-fold dose-sparing effect and induction of increased antigen-specific CD4(+) T-cell responses in mice. All mice that received one dose of adjuvanted vaccine and subsequent H5N1 viral challenges exhibited mild illness, lower lung viral titers, undetectable spleen and brain viral titers, and 100% survival after either homologous clade 1 or heterologous clade 2 H5N1 viral challenges, whereas unadjuvanted vaccine recipients showed significantly increased weight loss, viral titers, and mortality. The protective immunity induced by JVRS-100 adjuvanted H5N1 vaccine was shown to last for over one year without significant waning. Thus, JVRS-100 adjuvanted H5N1 vaccine elicited enhanced humoral and T-cell responses, dose-sparing, and cross-clade protection in mice. CLDC holds promise as an adjuvant for human pre-pandemic inactivated H5N1 vaccines.
Collapse
Affiliation(s)
- Libo Dong
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Popov J, Kapanen AI, Turner C, Ng R, Tucker C, Chiu G, Klasa R, Bally MB, Chikh G. Multivalent rituximab lipid nanoparticles as improved lymphoma therapies: indirect mechanisms of action and in vivo activity. Nanomedicine (Lond) 2011; 6:1575-91. [PMID: 22011314 DOI: 10.2217/nnm.11.50] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
AIMS The activity of therapeutic antibodies can be enhanced by creating multivalent constructs, such as antibody lipid nanoparticles (LNPs). Here, we examine differences between rituximab (Ritux) and Ritux-LNPs in terms of their indirect mechanisms of action: complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC). MATERIALS & METHODS We employed two mantle-cell lymphoma cell lines, Z138 and JVM2, which exhibit different in vivo sensitivities to Ritux along with variable expression levels of cell-surface proteins that regulate ADCC and CDC. RESULTS In both cell lines, CDC and ADCC were found to be significantly enhanced after treatment with Ritux-LNPs compared with Ritux. In vivo efficacy studies, however, suggested that the therapeutic activities of Ritux and Ritux-LNPs were equivalent, which was subsequently explained in part by pharmacokinetic studies indicating rapid elimination of Ritux-LNP. CONCLUSION Although indirect and direct mechanisms of multivalent Ritux are enhanced, its further development requires methods to improve its circulation lifetime.
Collapse
Affiliation(s)
- Jesse Popov
- Department of Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Ave., Vancouver BC, V5Z 1L3, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Improvement of the trivalent inactivated flu vaccine using PapMV nanoparticles. PLoS One 2011; 6:e21522. [PMID: 21747909 PMCID: PMC3126827 DOI: 10.1371/journal.pone.0021522] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 05/30/2011] [Indexed: 02/06/2023] Open
Abstract
Commercial seasonal flu vaccines induce production of antibodies directed mostly towards hemaglutinin (HA). Because HA changes rapidly in the circulating virus, the protection remains partial. Several conserved viral proteins, e.g., nucleocapsid (NP) and matrix proteins (M1), are present in the vaccine, but are not immunogenic. To improve the protection provided by these vaccines, we used nanoparticles made of the coat protein of a plant virus (papaya mosaic virus; PapMV) as an adjuvant. Immunization of mice and ferrets with the adjuvanted formulation increased the magnitude and breadth of the humoral response to NP and to highly conserved regions of HA. They also triggered a cellular mediated immune response to NP and M1, and long-lasting protection in animals challenged with a heterosubtypic influenza strain (WSN/33). Thus, seasonal flu vaccine adjuvanted with PapMV nanoparticles can induce universal protection to influenza, which is a major advancement when facing a pandemic.
Collapse
|
39
|
Interactions of human complement with virus particles containing the Nipah virus glycoproteins. J Virol 2011; 85:5940-8. [PMID: 21450814 DOI: 10.1128/jvi.00193-11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Complement is an innate immune response system that most animal viruses encounter during natural infections. We have tested the role of human complement in the neutralization of virus particles harboring the Nipah virus (NiV) glycoproteins. A luciferase-expressing vesicular stomatitis virus (VSV) pseudotype that contained the NiV fusion (F) and attachment (G) glycoproteins (NiVpp) showed dose- and time-dependent activation of human complement through the alternative pathway. In contrast to our findings with other paramyxoviruses, normal human serum (NHS) alone did not neutralize NiVpp infectivity in vitro, and electron microscopy demonstrated no significant deposition of complement component C3 on particles. This lack of NiVpp neutralization by NHS was not due to a global inhibition of complement pathways, since complement was found to significantly enhance neutralization by antibodies specific for the NiV F and G glycoproteins. Complement components C4 and C1q were necessary but not sufficient by themselves for the enhancement of antibody neutralization. Human complement also enhanced NiVpp neutralization by a soluble version of the NiV receptor EphrinB2, and this depended on components in the classical pathway. The ability of complement to enhance neutralization fell into one of two profiles: (i) anti-F monoclonal antibodies showed enhancement only at high and not low antibody concentrations, and (ii) anti-G monoclonal antibodies and EphrinB2 showed enhancement at both high and very low levels of antibody (e.g., 3.1 ng) or EphrinB2 (e.g., 2.5 ng). Together, these data establish the importance of human complement in the neutralization of particles containing the NiV glycoproteins and will help guide the design of more effective therapeutics that harness the potency of complement pathways.
Collapse
|
40
|
Dowd KA, Pierson TC. Antibody-mediated neutralization of flaviviruses: a reductionist view. Virology 2011; 411:306-15. [PMID: 21255816 PMCID: PMC3100196 DOI: 10.1016/j.virol.2010.12.020] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 12/11/2010] [Indexed: 12/28/2022]
Abstract
Flaviviruses are a group of ~70 small RNA viruses responsible for significant morbidity and mortality across the globe. Efforts to develop effective vaccines for several clinically important flaviviruses are underway. Antibodies are a significant component of the host's protective response against flavivirus infection with the potential to contribute to immunity via several distinct mechanisms, including an ability to directly neutralize virus infection. Conversely, virus-reactive antibodies have been implicated in the increased risk of severe clinical manifestations following secondary dengue virus infection. In this review, we will discuss recent progress toward understanding the molecular basis of antibody-mediated neutralization of flaviviruses. Neutralization requires engagement of the virion with a stoichiometry that exceeds a required threshold. From this perspective, we will discuss viral and host factors that impact the number of antibody molecules bound to the virus particle and significantly modulate the potency of neutralizing antibodies.
Collapse
Affiliation(s)
- Kimberly A. Dowd
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Theodore C. Pierson
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
41
|
Abstract
The complement system functions as an immune surveillance system that rapidly responds to infection. Activation of the complement system by specific recognition pathways triggers a protease cascade, generating cleavage products that function to eliminate pathogens, regulate inflammatory responses, and shape adaptive immune responses. However, when dysregulated, these powerful functions can become destructive and the complement system has been implicated as a pathogenic effector in numerous diseases, including infectious diseases. This review highlights recent discoveries that have identified critical roles for the complement system in the pathogenesis of viral infection.
Collapse
|
42
|
Staneková Z, Varečková E. Conserved epitopes of influenza A virus inducing protective immunity and their prospects for universal vaccine development. Virol J 2010; 7:351. [PMID: 21118546 PMCID: PMC3009981 DOI: 10.1186/1743-422x-7-351] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Accepted: 11/30/2010] [Indexed: 01/08/2023] Open
Abstract
Influenza A viruses belong to the best studied viruses, however no effective prevention against influenza infection has been developed. The emerging of still new escape variants of influenza A viruses causing epidemics and periodic worldwide pandemics represents a threat for human population. Therefore, current, hot task of influenza virus research is to look for a way how to get us closer to a universal vaccine. Combination of chosen conserved antigens inducing cross-protective antibody response with epitopes activating also cross-protective cytotoxic T-cells would offer an attractive strategy for improving protection against drift variants of seasonal influenza viruses and reduces the impact of future pandemic strains. Antigenically conserved fusion-active subunit of hemagglutinin (HA2 gp) and ectodomain of matrix protein 2 (eM2) are promising candidates for preparation of broadly protective HA2- or eM2-based vaccine that may aid in pandemic preparedness. Overall protective effect could be achieved by contribution of epitopes recognized by cytotoxic T-lymphocytes (CTL) that have been studied extensively to reach much broader control of influenza infection. In this review we present the state-of-art in this field. We describe known adaptive immune mechanisms mediated by influenza specific B- and T-cells involved in the anti-influenza immune defense together with the contribution of innate immunity. We discuss the mechanisms of neutralization of influenza infection mediated by antibodies, the role of CTL in viral elimination and new approaches to develop epitope based vaccine inducing cross-protective influenza virus-specific immune response.
Collapse
Affiliation(s)
- Zuzana Staneková
- Institute of Virology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovak Republic.
| | | |
Collapse
|
43
|
Pyaram K, Yadav VN, Reza MJ, Sahu A. Virus–complement interactions: an assiduous struggle for dominance. Future Virol 2010. [DOI: 10.2217/fvl.10.60] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The complement system is a major component of the innate immune system that recognizes invading pathogens and eliminates them by means of an array of effector mechanisms, in addition to using direct lytic destruction. Viruses, in spite of their small size and simple composition, are also deftly recognized and neutralized by the complement system. In turn, as a result of years of coevolution with the host, viruses have developed multiple mechanisms to evade the host complement. These complex interactions between the complement system and viruses have been an area of focus for over three decades. In this article, we provide a broad overview of the field using key examples and up-to-date information on the complement-evasion strategies of viruses.
Collapse
Affiliation(s)
- Kalyani Pyaram
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune 411007, India
| | - Viveka Nand Yadav
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune 411007, India
| | - Malik Johid Reza
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune 411007, India
| | | |
Collapse
|
44
|
Kanai Y, Boonsathorn N, Chittaganpitch M, Bai G, Li Y, Kase T, Takahashi K, Okuno Y, Jampangern W, Ikuta K, Sawanpanyalert P. The impact of antigenic drift of influenza A virus on human herd immunity: Sero-epidemiological study of H1N1 in healthy Thai population in 2009. Vaccine 2010; 28:5437-44. [PMID: 20558249 DOI: 10.1016/j.vaccine.2010.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 05/18/2010] [Accepted: 06/01/2010] [Indexed: 02/03/2023]
Abstract
To examine the effect of the antigenic drift of H1N1 influenza viruses on herd immunity, neutralization antibodies from 744 sera from Thai healthy volunteers in 2008-2009, who had not been vaccinated for at least the last 5 years, were investigated by microneutralization (MN) and hemagglutination inhibition (HI) assays. Significantly higher MN titers were observed for the H1N1 Thai isolate in 2006 than in 2008. The results indicate that the antigenically drifted virus effectively escaped herd immunity. Since the low neutralization activity of herd immunity against drifted viruses is an important factor for viruses to spread efficiently, continuous sero-epidemiological study is required for public health.
Collapse
Affiliation(s)
- Yuta Kanai
- Section of Viral Infections, Thailand-Japan Research Collaboration Center on Emerging and Re-emerging Infections (RCC-ERI), Nonthaburi, Thailand.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Cross-reactive neutralizing antibodies directed against pandemic H1N1 2009 virus are protective in a highly sensitive DBA/2 mouse influenza model. J Virol 2010; 84:7662-7. [PMID: 20484500 DOI: 10.1128/jvi.02444-09] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Our ability to rapidly respond to an emerging influenza pandemic is hampered somewhat by the lack of a susceptible small-animal model. To develop a more sensitive model, we pathotyped 18 low-pathogenic non-mouse-adapted influenza A viruses of human and avian origin in DBA/2 and C57BL/6 mice. The majority of the isolates (13/18) induced severe morbidity and mortality in DBA/2 mice upon intranasal challenge with 1 million infectious doses. Also, at a 100-fold-lower dose, more than 50% of the viruses induced severe weight loss, and mice succumbed to the infection. In contrast, only two virus strains were pathogenic for C57BL/6 mice upon high-dose inoculation. Therefore, DBA/2 mice are a suitable model to validate influenza A virus vaccines and antiviral therapies without the need for extensive viral adaptation. Correspondingly, we used the DBA/2 model to assess the level of protection afforded by preexisting pandemic H1N1 2009 virus (H1N1pdm) cross-reactive human antibodies detected by a hemagglutination inhibition assay. Passive transfer of these antibodies prior to infection protected mice from H1N1pdm-induced pathogenicity, demonstrating the effectiveness of these cross-reactive neutralizing antibodies in vivo.
Collapse
|
46
|
Corti D, Suguitan AL, Pinna D, Silacci C, Fernandez-Rodriguez BM, Vanzetta F, Santos C, Luke CJ, Torres-Velez FJ, Temperton NJ, Weiss RA, Sallusto F, Subbarao K, Lanzavecchia A. Heterosubtypic neutralizing antibodies are produced by individuals immunized with a seasonal influenza vaccine. J Clin Invest 2010; 120:1663-73. [PMID: 20389023 DOI: 10.1172/jci41902] [Citation(s) in RCA: 371] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 02/17/2010] [Indexed: 11/17/2022] Open
Abstract
The target of neutralizing antibodies that protect against influenza virus infection is the viral protein HA. Genetic and antigenic variation in HA has been used to classify influenza viruses into subtypes (H1-H16). The neutralizing antibody response to influenza virus is thought to be specific for a few antigenically related isolates within a given subtype. However, while heterosubtypic antibodies capable of neutralizing multiple influenza virus subtypes have been recently isolated from phage display libraries, it is not known whether such antibodies are produced in the course of an immune response to influenza virus infection or vaccine. Here we report that, following vaccination with seasonal influenza vaccine containing H1 and H3 influenza virus subtypes, some individuals produce antibodies that cross-react with H5 HA. By immortalizing IgG-expressing B cells from 4 individuals, we isolated 20 heterosubtypic mAbs that bound and neutralized viruses belonging to several HA subtypes (H1, H2, H5, H6, and H9), including the pandemic A/California/07/09 H1N1 isolate. The mAbs used different VH genes and carried a high frequency of somatic mutations. With the exception of a mAb that bound to the HA globular head, all heterosubtypic mAbs bound to acid-sensitive epitopes in the HA stem region. Four mAbs were evaluated in vivo and protected mice from challenge with influenza viruses representative of different subtypes. These findings reveal that seasonal influenza vaccination can induce polyclonal heterosubtypic neutralizing antibodies that cross-react with the swine-origin pandemic H1N1 influenza virus and with the highly pathogenic H5N1 virus.
Collapse
Affiliation(s)
- Davide Corti
- Institute for Research in Biomedicine, Bellinzona, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Much remains to be learned about the pathogenesis of the different manifestations of dengue virus (DENV) infections in humans. They may range from subclinical infection to dengue fever, dengue hemorrhagic fever (DHF), and eventually dengue shock syndrome (DSS). As both cell tropism and tissue tropism of DENV are considered major determinants in the pathogenesis of dengue, there is a critical need for adequate tropism assays, animal models, and human autopsy data. More than 50 years of research on dengue has resulted in a host of literature, which strongly suggests that the pathogenesis of DHF and DSS involves viral virulence factors and detrimental host responses, collectively resulting in abnormal hemostasis and increased vascular permeability. Differential targeting of specific vascular beds is likely to trigger the localized vascular hyperpermeability underlying DSS. A personalized approach to the study of pathogenesis will elucidate the basis of individual risk for development of DHF and DSS as well as identify the genetic and environmental bases for differences in risk for development of severe disease.
Collapse
|
48
|
Mehlhop E, Nelson S, Jost CA, Gorlatov S, Johnson S, Fremont DH, Diamond MS, Pierson TC. Complement protein C1q reduces the stoichiometric threshold for antibody-mediated neutralization of West Nile virus. Cell Host Microbe 2009; 6:381-91. [PMID: 19837377 DOI: 10.1016/j.chom.2009.09.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2009] [Revised: 07/23/2009] [Accepted: 09/15/2009] [Indexed: 01/16/2023]
Abstract
Virus neutralization is governed by the number of antibodies that bind a virion during the cellular entry process. Cellular and serum factors that interact with antibodies have the potential to modulate neutralization potency. Although the addition of serum complement can increase the neutralizing activity of antiviral antibodies in vitro, the mechanism and significance of this augmented potency in vivo remain uncertain. Herein, we show that the complement component C1q increases the potency of antibodies against West Nile virus by modulating the stoichiometric requirements for neutralization. The addition of C1q does not result in virolysis but instead reduces the number of antibodies that must bind the virion to neutralize infectivity. For IgG subclasses that bind C1q avidly, this reduced stoichiometric threshold falls below the minimal number of antibodies required for antibody-dependent enhancement (ADE) of infection of cells expressing Fc-gamma receptors (CD32) and explains how C1q restricts the ADE of flavivirus infection.
Collapse
Affiliation(s)
- Erin Mehlhop
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Hartikka J, Bozoukova V, Yang CK, Ye M, Rusalov D, Shlapobersky M, Vilalta A, Wei Q, Rolland A, Smith LR. Vaxfectin®, a cationic lipid-based adjuvant for protein-based influenza vaccines. Vaccine 2009; 27:6399-403. [DOI: 10.1016/j.vaccine.2009.06.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
50
|
Complement modulates pathogenesis and antibody-dependent neutralization of West Nile virus infection through a C5-independent mechanism. Virology 2009; 393:11-5. [PMID: 19744691 DOI: 10.1016/j.virol.2009.08.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Revised: 07/28/2009] [Accepted: 08/08/2009] [Indexed: 11/23/2022]
Abstract
Although the interactions of complement and viruses have been widely studied, the function of C5 and the membrane attack complex in the context of viral infection or antibody-mediated neutralization remains controversial. Using C5-depleted or -deficient human or mouse sera, we show that C5 does not contribute to the antibody-dependent or -independent neutralization of West Nile virus (WNV) in cell culture. Consistent with this, C5 neither contributed to protection against WNV pathogenesis nor augmented the neutralizing efficacy of complement-fixing anti-WNV neutralizing antibodies in mice. Although previous studies established that activation of the classical, lectin, and alternative complement pathways restricts WNV infection, our results show little effect of C5 and by inference the terminal lytic complement components. Overall, these results enhance our mechanistic understanding of how complement controls flavivirus infections.
Collapse
|