1
|
Gao P, Wu F, Liu J, Li R, Jiang X, Pan W, Zhao F, Niu X, Xu W. Development of a dual-component biosensor for rapid and sensitive detection of influenza H7 and H5 subtypes. Talanta 2024; 280:126704. [PMID: 39151319 DOI: 10.1016/j.talanta.2024.126704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/17/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
The outbreak of highly pathogenic influenza virus subtypes, such as H7 and H5, presents a significant global health challenge, necessitating the development of rapid and sensitive diagnostic methods. In this study, we have developed a novel dual-component biosensor assembly, each component of which incorporates an antibody fused with a nano-luciferase subunit. Our results demonstrate the effectiveness of this biosensor in enabling the rapid and sensitive detection of influenza H7 and other subtypes. Additionally, we successfully applied the biosensor in paper-based assay and lateral flow assay formats, expanding its versatility and potential for field-deployable applications. Notably, we achieved effective detection of the H7N9 virus using this biosensor. Furthermore, we designed and optimized a dedicated biosensor to the sensitive detection of the influenza H5 subtype. Collectively, our findings underscore the significant potential of this dual-component biosensor assembly as a valuable and versatile tool for accurate and timely diagnosis of influenza virus infections, promising to advance the field of influenza diagnostics and contribute to outbreak management and surveillance efforts.
Collapse
Affiliation(s)
- Peixuan Gao
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Fang Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, 528000, China
| | - Jun Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Rui Li
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiwen Jiang
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Weiqi Pan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Fang Zhao
- National Clinical Research Centre for Infectious Diseases, the Third People's Hospital of Shenzhen and the Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, 518112, China
| | - Xuefeng Niu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Wei Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Liu X, Li Z, Li X, Zhang X, Zheng Y, Su W, Feng Y, Liu Y, Wu W, Sun X, Wang N, Ye X, Zhou Z, Liu W, He J, Wang W, Qu L, Zhou R, Chen L, Feng L. Neutralizing monoclonal antibodies protect against human adenovirus type 55 infection in transgenic mice and tree shrews. Emerg Microbes Infect 2024; 13:2307513. [PMID: 38240267 PMCID: PMC10836490 DOI: 10.1080/22221751.2024.2307513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/16/2024] [Indexed: 02/03/2024]
Abstract
Re-emerging human adenovirus type 55 (HAdV55) has become a significant threat to public health due to its widespread circulation and the association with severe pneumonia, but an effective anti-HAdV55 agent remains unavailable. Herein, we report the generation of macaque-derived, human-like monoclonal antibodies (mAbs) protecting against HAdV55 infection with high potency. Using fluorophore-labelled HAdV55 virions as probes, we isolated specific memory B cells from rhesus macaques (Macaca mulatta) that were immunized twice with an experimental vaccine based on E1-, E3-deleted, replication-incompetent HAdV55. We cloned a total of 19 neutralizing mAbs, nine of which showed half-maximal inhibitory concentrations below 1.0 ng/ml. These mAbs recognized the hyper-variable-region (HVR) 1, 2, or 7 of viral hexon protein, or the fibre knob. In transgenic mice expressing human desmoglein-2, the major cellular receptor for HAdV55, a single intraperitoneal injection with hexon-targeting mAbs efficiently prevented HAdV55 infection, and mAb 29C12 showed protection at a dose as low as 0.004 mg/kg. Fibre-targeting mAb 28E8, however, showed protection only at a dose up to 12.5 mg/kg. In tree shrews that are permissive for HAdV55 infection and disease, mAb 29C12 effectively prevented HAdV55-caused pneumonia. Further analysis revealed that fibre-targeting mAbs blocked the attachment of HAdV55 to host cells, whereas hexon-targeting mAbs, regardless of their targeting HVRs, mainly functioned at post-attachment stage via inhibiting viral endosomal escape. Our results indicate that hexon-targeting mAbs have great anti-HAdV55 activities and warrant pre-clinical and clinical evaluation.
Collapse
Affiliation(s)
- Xinglong Liu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Zhengfeng Li
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Xiao Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiaoyan Zhang
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Yali Zheng
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Wan Su
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Ying Feng
- Guangzhou Laboratory & Bioland Laboratory, Guangzhou, People's Republic of China
| | - Yutong Liu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Weixuan Wu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Xikui Sun
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Nana Wang
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Xianmiao Ye
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Zhichao Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Wenkuan Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jun He
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Wei Wang
- Guangzhou Laboratory & Bioland Laboratory, Guangzhou, People's Republic of China
| | - Linbing Qu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Rong Zhou
- Guangzhou Laboratory & Bioland Laboratory, Guangzhou, People's Republic of China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
- Guangzhou nBiomed Ltd., Guangzhou, People's Republic of China
| | - Liqiang Feng
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
3
|
Chen X, Huang Y, Gao P, Wu F, Han Y, Zhang C, Hu Z, Zhao F, Shcherbakov DN, Pan W, Niu X, Li X, Liu S, Xu W. Engineering of novel hemagglutinin biosensors for rapid detection and drug screening of Influenza A H7N9 virus. Int J Biol Macromol 2024; 258:129126. [PMID: 38163504 DOI: 10.1016/j.ijbiomac.2023.129126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
New pathogenic influenza virus strains are constantly emerging, posing a serious risk to both human health and economic growth. To effectively control the spread of this virus, there is an urgent need for early, rapid, sensitive, simple, and cost-effective detection technologies, as well as new and effective antiviral drugs. In this study, we have successfully achieved a significant milestone by successfully fusing the H7N9 influenza virus hemagglutinin (HA) protein with the nano-luciferase component, resulting in the development of a novel set of biosensors. This remarkable achievement marks the first instance of utilizing this biosensor technology for influenza antibody detection. Our biosensor technology also has the potential to facilitate the development of antiviral drugs targeting specific epitopes of the HA protein, providing a promising avenue for the treatment of H7N9 influenza virus infections. Furthermore, our biosensors have broad applications beyond H7N9 influenza virus detection, as they can be expanded for the detection of other pathogens and drug screening applications in the future. By providing a novel and effective solution to the detection and treatment of influenza viruses, our biosensors have the potential to revolutionize the field of infectious disease control.
Collapse
Affiliation(s)
- Xin Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yuan Huang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Peixuan Gao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Fang Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yongyue Han
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Chuwen Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhuowen Hu
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510070, China
| | - Fang Zhao
- National Clinical Research Centre for Infectious Diseases, the Third People's Hospital of Shenzhen and the Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Dmitry N Shcherbakov
- State Research Center of Virology and Biotechnology VECTOR, Koltsovo 630559, Russia; Department of Physical-Chemistry Biology and Biotechnology, Altai State University, Barnaul 656049, Russia
| | - Weiqi Pan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Xuefeng Niu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Xiaoyan Li
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510070, China.
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China; State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou 510515, China.
| | - Wei Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
4
|
Li Y, Xu M, Li Y, Gu W, Halimu G, Li Y, Zhang Z, Zhou L, Liao H, Yao S, Zhang H, Zhang C. A recombinant protein containing influenza viral conserved epitopes and superantigen induces broad-spectrum protection. eLife 2021; 10:e71725. [PMID: 34783655 PMCID: PMC8635977 DOI: 10.7554/elife.71725] [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: 06/28/2021] [Accepted: 11/13/2021] [Indexed: 01/22/2023] Open
Abstract
Influenza pandemics pose public health threats annually for lacking vaccine that provides cross-protection against novel and emerging influenza viruses. Combining conserved antigens that induce cross-protective antibody responses with epitopes that activate cross-protective T cell responses might be an attractive strategy for developing a universal vaccine. In this study, we constructed a recombinant protein named NMHC that consists of influenza viral conserved epitopes and a superantigen fragment. NMHC promoted the maturation of bone marrow-derived dendritic cells and induced CD4+ T cells to differentiate into Th1, Th2, and Th17 subtypes. Mice vaccinated with NMHC produced high levels of immunoglobulins that cross-bound to HA fragments from six influenza virus subtypes with high antibody titers. Anti-NMHC serum showed potent hemagglutinin inhibition effects to highly divergent group 1 (H1 subtype) and group 2 (H3 subtype) influenza virus strains. Furthermore, purified anti-NMHC antibodies bound to multiple HAs with high affinities. NMHC vaccination effectively protected mice from infection and lung damage when exposed to two subtypes of H1N1 influenza virus. Moreover, NMHC vaccination elicited CD4+ and CD8+ T cell responses that cleared the virus from infected tissues and prevented virus spread. In conclusion, this study provides proof of concept that NMHC vaccination triggers B and T cell immune responses against multiple influenza virus infections. Therefore, NMHC might be a candidate universal broad-spectrum vaccine for the prevention and treatment of multiple influenza viruses.
Collapse
Affiliation(s)
- Yansheng Li
- Institute of Applied Ecology, Chinese Academy of SciencesShenyangChina
- University of Chinese Academy of SciencesBeijingChina
- Key Laboratory of Superantigen Research, Shenyang Bureau of Science and TechnologyShenyangChina
| | - Mingkai Xu
- Institute of Applied Ecology, Chinese Academy of SciencesShenyangChina
- Key Laboratory of Superantigen Research, Shenyang Bureau of Science and TechnologyShenyangChina
| | - Yongqiang Li
- Institute of Applied Ecology, Chinese Academy of SciencesShenyangChina
- University of Chinese Academy of SciencesBeijingChina
- Key Laboratory of Superantigen Research, Shenyang Bureau of Science and TechnologyShenyangChina
| | - Wu Gu
- Institute of Applied Ecology, Chinese Academy of SciencesShenyangChina
- Key Laboratory of Superantigen Research, Shenyang Bureau of Science and TechnologyShenyangChina
| | - Gulinare Halimu
- Institute of Applied Ecology, Chinese Academy of SciencesShenyangChina
- University of Chinese Academy of SciencesBeijingChina
- Key Laboratory of Superantigen Research, Shenyang Bureau of Science and TechnologyShenyangChina
| | - Yuqi Li
- Institute of Applied Ecology, Chinese Academy of SciencesShenyangChina
- University of Chinese Academy of SciencesBeijingChina
- Key Laboratory of Superantigen Research, Shenyang Bureau of Science and TechnologyShenyangChina
| | - Zhichun Zhang
- Institute of Applied Ecology, Chinese Academy of SciencesShenyangChina
- University of Chinese Academy of SciencesBeijingChina
- Key Laboratory of Superantigen Research, Shenyang Bureau of Science and TechnologyShenyangChina
| | - Libao Zhou
- Chengda Biotechnology Co. LtdLiaoningChina
| | - Hui Liao
- Chengda Biotechnology Co. LtdLiaoningChina
| | | | - Huiwen Zhang
- Institute of Applied Ecology, Chinese Academy of SciencesShenyangChina
- Key Laboratory of Superantigen Research, Shenyang Bureau of Science and TechnologyShenyangChina
| | - Chenggang Zhang
- Institute of Applied Ecology, Chinese Academy of SciencesShenyangChina
- Key Laboratory of Superantigen Research, Shenyang Bureau of Science and TechnologyShenyangChina
| |
Collapse
|
5
|
Ali MG, Zhang Z, Gao Q, Pan M, Rowan EG, Zhang J. Recent advances in therapeutic applications of neutralizing antibodies for virus infections: an overview. Immunol Res 2020; 68:325-339. [PMID: 33161557 PMCID: PMC7648849 DOI: 10.1007/s12026-020-09159-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/21/2020] [Indexed: 12/14/2022]
Abstract
Antibodies are considered as an excellent foundation to neutralize pathogens and as highly specific therapeutic agents. Antibodies are generated in response to a vaccine but little use as immunotherapy to combat virus infections. A new generation of broadly cross-reactive and highly potent antibodies has led to a unique chance for them to be used as a medical intervention. Neutralizing antibodies (monoclonal and polyclonal antibodies) are desirable for pharmaceutical products because of their ability to target specific epitopes with their variable domains by precise neutralization mechanisms. The isolation of neutralizing antiviral antibodies has been achieved by Phage displayed antibody libraries, transgenic mice, B cell approaches, and hybridoma technology. Antibody engineering technologies have led to efficacy improvements, to further boost antibody in vivo activities. "Although neutralizing antiviral antibodies have some limitations that hinder their full development as therapeutic agents, the potential for prevention and treatment of infections, including a range of viruses (HIV, Ebola, MERS-COV, CHIKV, SARS-CoV, and SARS-CoV2), are being actively pursued in human clinical trials."
Collapse
Affiliation(s)
- Manasik Gumah Ali
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Zhening Zhang
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Qi Gao
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Mingzhu Pan
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Edward G Rowan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University Strathclyde, Glasgow, UK
| | - Juan Zhang
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
6
|
Niu X, Yan Q, Yao Z, Zhang F, Qu L, Wang C, Wang C, Lei H, Chen C, Liang R, Luo J, Wang Q, Zhao L, Zhang Y, Luo K, Wang L, Wu H, Liu T, Li P, Zheng Z, Tan YJ, Feng L, Zhang Z, Han J, Zhang F, Chen L. Longitudinal analysis of the antibody repertoire of a Zika virus-infected patient revealed dynamic changes in antibody response. Emerg Microbes Infect 2020; 9:111-123. [PMID: 31906823 PMCID: PMC6968589 DOI: 10.1080/22221751.2019.1701953] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Zika virus (ZIKV) is a mosquito-borne flavivirus that causes neonatal abnormalities and other disorders. Antibodies to the ZIKV envelope (E) protein can block infection. In this study, next-generation sequencing (NGS) of immunoglobulin heavy chain (IgH) mRNA transcripts was combined with single-cell PCR cloning of E-binding monoclonal antibodies for analysing antibody response in a patient from the early stages of infection to more than one year after the clearance of the virus. The patient's IgH repertoire 14 and 64 days after symptom onset showed dramatic dominant clonal expansion but low clonal diversity. IgH repertoire 6 months after disease-free status had few dominant clones but increased diversity. E-binding antibodies appeared abundantly in the repertoire during the early stages of infection but quickly declined after clearance of the virus. Certain VH genes such as VH5-10-1 and VH4-39 appeared to be preferentially enlisted for a rapid antibody response to ZIKV infection. Most of these antibodies require relatively few somatic hypermutations to acquire the ability to bind to the E protein, pointing to a possible mechanism for rapid defence against ZIKV infection. This study provides a unique and holistic view of the dynamic changes and characteristics of the antibody response to ZIKV infection.
Collapse
Affiliation(s)
- Xuefeng Niu
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Qihong Yan
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China.,University of Chinese Academy of Science, Beijing, People's Republic of China
| | - Zhipeng Yao
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China.,Institute of Physical Science and Information Technology, Anhui University, Hefei, People's Republic of China
| | - Fan Zhang
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China.,Institute of Physical Science and Information Technology, Anhui University, Hefei, People's Republic of China
| | - Linbing Qu
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Chunlin Wang
- HudsonAlpha Institute of Biotechnology, Huntsville, AL, USA
| | - Chengrui Wang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Hui Lei
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Chaoming Chen
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Renshan Liang
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jia Luo
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Qian Wang
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China.,University of Chinese Academy of Science, Beijing, People's Republic of China
| | - Lingzhai Zhao
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yudi Zhang
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China.,University of Chinese Academy of Science, Beijing, People's Republic of China
| | - Kun Luo
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China.,University of Chinese Academy of Science, Beijing, People's Republic of China
| | - Longyu Wang
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China.,Institute of Physical Science and Information Technology, Anhui University, Hefei, People's Republic of China
| | - Hongkai Wu
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Tingting Liu
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Pingchao Li
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Zhiqiang Zheng
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore, Singapore
| | - Yee Joo Tan
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore, Singapore
| | - Liqiang Feng
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Zhenhai Zhang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jian Han
- HudsonAlpha Institute of Biotechnology, Huntsville, AL, USA
| | - Fuchun Zhang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China.,Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| |
Collapse
|
7
|
Zhang F, Wang L, Niu X, Li J, Luo J, Feng Y, Yang Y, He P, Fan W, Liang R, Zheng Z, Pan W, Li C, Tan YJ, Yu H, Chen L, Li P. Phenotypic Characterization of Chinese Rhesus Macaque Plasmablasts for Cloning Antigen-Specific Monoclonal Antibodies. Front Immunol 2019; 10:2426. [PMID: 31681312 PMCID: PMC6798180 DOI: 10.3389/fimmu.2019.02426] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 09/27/2019] [Indexed: 11/14/2022] Open
Abstract
Rhesus macaques (Macaca mulatta) are used as a human-relevant animal species for the evaluation of vaccines and as a source for cloning monoclonal antibodies (mAbs) that are highly similar to human-derived antibodies. Although antibody-secreting plasmablasts in humans are well-defined and can be easily isolated for mAb cloning, it remains unclear whether the same phenotypic markers could be applied for isolating antibody-secreting plasmablasts from Chinese rhesus macaques. In this study, we evaluated a series of cell surface and intracellular markers and identified the phenotypic markers of plasmablasts in Chinese rhesus macaques as CD3−CD14−CD56−CD19−CD27−CD20−/lowCD80+HLA-DR+CD95+. After influenza virus vaccination, the plasmablasts in peripheral blood mononuclear cells (PBMCs) increased transiently, peaked at day 4–7 after booster vaccination and returned to nearly undetectable levels by day 14. Antigen-specific enzyme-linked immunosorbent spot (ELISPOT) assays confirmed that the majority of the plasmablasts could produce influenza virus-specific antibodies. These plasmablasts showed transcriptional characteristics similar to those of human plasmablasts. Using single-cell PCR for immunoglobulin heavy and light chains, most mAbs cloned from the CD3−CD14−CD56−CD19−CD27−CD20−/lowCD80+HLA-DR+CD95+ plasmablasts after vaccination exhibited specific binding to influenza virus. This study defined the phenotypic markers for isolating antibody-secreting plasmablasts from Chinese rhesus macaques, which has implications for efficient cloning of mAbs and for the evaluation of plasmablast response after vaccination or infection in Chinese rhesus macaques.
Collapse
Affiliation(s)
- Fan Zhang
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Longyu Wang
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Xuefeng Niu
- State Key Laboratory of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiashun Li
- Department of Respiratory Medicine, Huadu People's Hospital, Guangzhou, China
| | - Jia Luo
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yupeng Feng
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yanjia Yang
- State Key Laboratory of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ping He
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Wenxia Fan
- State Key Laboratory of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Renshan Liang
- State Key Laboratory of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiqiang Zheng
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore, Singapore.,Institute of Molecular and Cell Biology, ASTAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Weiqi Pan
- State Key Laboratory of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chufang Li
- State Key Laboratory of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yee Joo Tan
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore, Singapore.,Institute of Molecular and Cell Biology, ASTAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Haijian Yu
- Department of Respiratory Medicine, Huadu People's Hospital, Guangzhou, China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,State Key Laboratory of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pingchao Li
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
8
|
Dibo M, Battocchio EC, dos Santos Souza LM, da Silva MDV, Banin-Hirata BK, Sapla MM, Marinello P, Rocha SP, Faccin-Galhardi LC. Antibody Therapy for the Control of Viral Diseases: An Update. Curr Pharm Biotechnol 2019; 20:1108-1121. [DOI: 10.2174/1389201020666190809112704] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 04/22/2019] [Accepted: 08/01/2019] [Indexed: 12/29/2022]
Abstract
The epidemiological impact of viral diseases, combined with the emergence and reemergence of some viruses, and the difficulties in identifying effective therapies, have encouraged several studies to develop new therapeutic strategies for viral infections. In this context, the use of immunotherapy for the treatment of viral diseases is increasing. One of the strategies of immunotherapy is the use of antibodies, particularly the monoclonal antibodies (mAbs) and multi-specific antibodies, which bind directly to the viral antigen and bring about activation of the immune system. With current advancements in science and technology, several such antibodies are being tested, and some are already approved and are undergoing clinical trials. The present work aims to review the status of mAb development for the treatment of viral diseases.
Collapse
Affiliation(s)
- Miriam Dibo
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Eduardo C. Battocchio
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Lucas M. dos Santos Souza
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | | | - Bruna K. Banin-Hirata
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Milena M.M. Sapla
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Poliana Marinello
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Sérgio P.D. Rocha
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Lígia C. Faccin-Galhardi
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| |
Collapse
|
9
|
Niu X, Zhao L, Qu L, Yao Z, Zhang F, Yan Q, Zhang S, Liang R, Chen P, Luo J, Xu W, Lv H, Liu X, Lei H, Yi C, Li P, Wang Q, Wang Y, Yu L, Zhang X, Bryan LA, Davidson E, Doranz JB, Feng L, Pan W, Zhang F, Chen L. Convalescent patient-derived monoclonal antibodies targeting different epitopes of E protein confer protection against Zika virus in a neonatal mouse model. Emerg Microbes Infect 2019; 8:749-759. [PMID: 31130109 PMCID: PMC6542155 DOI: 10.1080/22221751.2019.1614885] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The Zika virus (ZIKV) outbreak and its link to microcephaly triggered a public health concern. To examine antibody response in a patient infected with ZIKV, we used single-cell PCR to clone 31 heavy and light chain-paired monoclonal antibodies (mAbs) that bind to ZIKV envelope (E) proteins isolated from memory B cells of a ZIKV-infected patient. Three mAbs (7B3, 1C11, and 6A6) that showed the most potent and broad neutralization activities against the African, Asian, and American strains were selected for further analysis. mAb 7B3 showed an IC50 value of 11.6 ng/mL against the circulating American strain GZ02. Epitope mapping revealed that mAbs 7B3 and 1C11 targeted residue K394 of the lateral ridge (LR) epitope of the EDIII domain, but 7B3 has a broader LR epitope footprint and recognizes residues T335, G337, E370, and N371 as well. mAb 6A6 recognized residues D67, K118, and K251 of the EDII domain. Interestingly, although the patient was seronegative for DENV infection, mAb 1C11, originating from the VH3-23 and VK1-5 germline pair, neutralized both ZIKV and DENV1. Administration of the mAbs 7B3, 1C11, and 6A6 protected neonatal SCID mice infected with a lethal dose of ZIKV. This study provides potential therapeutic antibody candidates and insights into the antibody response after ZIKV infection.
Collapse
Affiliation(s)
- Xuefeng Niu
- a State Key Lab of Respiratory Disease , the First Affiliated Hospital of Guangzhou Medical University , Guangzhou , People's Republic of China
| | - Lingzhai Zhao
- b Guangzhou 8th People's Hospital of Guangzhou Medical University , Guangzhou , People's Republic of China
| | - Linbing Qu
- c Guangdong Laboratory of Computational Biomedicine, Chinese Academy of Sciences , Guangzhou Institutes of Biomedicine and Health , Guangzhou , People's Republic of China
| | - Zhipeng Yao
- c Guangdong Laboratory of Computational Biomedicine, Chinese Academy of Sciences , Guangzhou Institutes of Biomedicine and Health , Guangzhou , People's Republic of China.,d Institute of Physical Science and Information Technology , Anhui University , Hefei , People's Republic of China
| | - Fan Zhang
- c Guangdong Laboratory of Computational Biomedicine, Chinese Academy of Sciences , Guangzhou Institutes of Biomedicine and Health , Guangzhou , People's Republic of China.,d Institute of Physical Science and Information Technology , Anhui University , Hefei , People's Republic of China
| | - Qihong Yan
- c Guangdong Laboratory of Computational Biomedicine, Chinese Academy of Sciences , Guangzhou Institutes of Biomedicine and Health , Guangzhou , People's Republic of China.,e University of Chinese Academy of Science , Beijing , People's Republic of China
| | - Shengnan Zhang
- c Guangdong Laboratory of Computational Biomedicine, Chinese Academy of Sciences , Guangzhou Institutes of Biomedicine and Health , Guangzhou , People's Republic of China
| | - Renshan Liang
- a State Key Lab of Respiratory Disease , the First Affiliated Hospital of Guangzhou Medical University , Guangzhou , People's Republic of China
| | - Peihai Chen
- d Institute of Physical Science and Information Technology , Anhui University , Hefei , People's Republic of China
| | - Jia Luo
- c Guangdong Laboratory of Computational Biomedicine, Chinese Academy of Sciences , Guangzhou Institutes of Biomedicine and Health , Guangzhou , People's Republic of China
| | - Wei Xu
- b Guangzhou 8th People's Hospital of Guangzhou Medical University , Guangzhou , People's Republic of China
| | - Huibin Lv
- a State Key Lab of Respiratory Disease , the First Affiliated Hospital of Guangzhou Medical University , Guangzhou , People's Republic of China
| | - Xinglong Liu
- c Guangdong Laboratory of Computational Biomedicine, Chinese Academy of Sciences , Guangzhou Institutes of Biomedicine and Health , Guangzhou , People's Republic of China
| | - Hui Lei
- a State Key Lab of Respiratory Disease , the First Affiliated Hospital of Guangzhou Medical University , Guangzhou , People's Republic of China
| | - Changhua Yi
- c Guangdong Laboratory of Computational Biomedicine, Chinese Academy of Sciences , Guangzhou Institutes of Biomedicine and Health , Guangzhou , People's Republic of China
| | - Pingchao Li
- c Guangdong Laboratory of Computational Biomedicine, Chinese Academy of Sciences , Guangzhou Institutes of Biomedicine and Health , Guangzhou , People's Republic of China
| | - Qian Wang
- c Guangdong Laboratory of Computational Biomedicine, Chinese Academy of Sciences , Guangzhou Institutes of Biomedicine and Health , Guangzhou , People's Republic of China
| | - Yang Wang
- a State Key Lab of Respiratory Disease , the First Affiliated Hospital of Guangzhou Medical University , Guangzhou , People's Republic of China
| | - Lei Yu
- b Guangzhou 8th People's Hospital of Guangzhou Medical University , Guangzhou , People's Republic of China
| | - Xiaoyan Zhang
- c Guangdong Laboratory of Computational Biomedicine, Chinese Academy of Sciences , Guangzhou Institutes of Biomedicine and Health , Guangzhou , People's Republic of China.,e University of Chinese Academy of Science , Beijing , People's Republic of China
| | | | | | | | - Liqiang Feng
- c Guangdong Laboratory of Computational Biomedicine, Chinese Academy of Sciences , Guangzhou Institutes of Biomedicine and Health , Guangzhou , People's Republic of China
| | - Weiqi Pan
- a State Key Lab of Respiratory Disease , the First Affiliated Hospital of Guangzhou Medical University , Guangzhou , People's Republic of China
| | - Fuchun Zhang
- b Guangzhou 8th People's Hospital of Guangzhou Medical University , Guangzhou , People's Republic of China
| | - Ling Chen
- a State Key Lab of Respiratory Disease , the First Affiliated Hospital of Guangzhou Medical University , Guangzhou , People's Republic of China.,b Guangzhou 8th People's Hospital of Guangzhou Medical University , Guangzhou , People's Republic of China.,c Guangdong Laboratory of Computational Biomedicine, Chinese Academy of Sciences , Guangzhou Institutes of Biomedicine and Health , Guangzhou , People's Republic of China
| |
Collapse
|
10
|
Pan W, Xie H, Li X, Guan W, Chen P, Zhang B, Zhang M, Dong J, Wang Q, Li Z, Li S, Yang Z, Li C, Zhong N, Huang J, Chen L. Patient-derived avian influenza A (H5N6) virus is highly pathogenic in mice but can be effectively treated by anti-influenza polyclonal antibodies. Emerg Microbes Infect 2018; 7:107. [PMID: 29899428 PMCID: PMC6000000 DOI: 10.1038/s41426-018-0113-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 05/04/2018] [Accepted: 05/13/2018] [Indexed: 11/30/2022]
Abstract
Highly pathogenic avian influenza A (H5N6) virus has been circulating in poultry since 2013 and causes sporadic infections and fatalities in humans. Due to the re-occurrence and continuous evolution of this virus subtype, there is an urgent need to better understand the pathogenicity of the H5N6 virus and to identify effective preventative and therapeutic strategies. We established a mouse model to evaluate the virulence of H5N6 A/Guangzhou/39715/2014 (H5N6/GZ14), which was isolated from an infected patient. BALB/c mice were inoculated intranasally with H5N6/GZ14 and monitored for morbidity, mortality, cytokine production, lung injury, viral replication, and viral dissemination to other organs. H5N6/GZ14 is highly pathogenic and can kill 50% of mice at a very low infectious dose of 5 plaque-forming units (pfu). Infection with H5N6/GZ14 showed rapid disease progression, viral replication to high titers in the lung, a strongly induced pro-inflammatory cytokine response, and severe lung injury. Moreover, infectious H5N6/GZ14 could be detected in the heart and brain of the infected mice. We also demonstrated that anti-influenza polyclonal antibodies generated by immunizing rhesus macaques could protect mice from lethal infection. Our results provide insights into the pathogenicity of the H5N6 human isolate.
Collapse
Affiliation(s)
- Weiqi Pan
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haojun Xie
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaobo Li
- Health Quarantine Laboratory, Guangdong Inspection and Quarantine Technology Center, Guangzhou, China
| | - Wenda Guan
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Peihai Chen
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Beiwu Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Mincong Zhang
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ji Dong
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qian Wang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhixia Li
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Shufen Li
- Health Quarantine Laboratory, Guangdong Inspection and Quarantine Technology Center, Guangzhou, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chufang Li
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jicheng Huang
- Health Quarantine Laboratory, Guangdong Inspection and Quarantine Technology Center, Guangzhou, China.
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China. .,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
| |
Collapse
|
11
|
Antibody therapies for the prevention and treatment of viral infections. NPJ Vaccines 2017; 2:19. [PMID: 29263875 PMCID: PMC5627241 DOI: 10.1038/s41541-017-0019-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/08/2017] [Accepted: 05/16/2017] [Indexed: 12/18/2022] Open
Abstract
Antibodies are an important component in host immune responses to viral pathogens. Because of their unique maturation process, antibodies can evolve to be highly specific to viral antigens. Physicians and researchers have been relying on such high specificity in their quest to understand host–viral interaction and viral pathogenesis mechanisms and to find potential cures for viral infection and disease. With more than 60 recombinant monoclonal antibodies developed for human use in the last 20 years, monoclonal antibodies are now considered a viable therapeutic modality for infectious disease targets, including newly emerging viral pathogens such as Ebola representing heightened public health concerns, as well as pathogens that have long been known, such as human cytomegalovirus. Here, we summarize some recent advances in identification and characterization of monoclonal antibodies suitable as drug candidates for clinical evaluation, and review some promising candidates in the development pipeline.
Collapse
|
12
|
Li JL, Zheng YT, Zhao XD, Hu XT. Meeting report: the 4 th symposium on animal models of non-human primates in Kunming, Yunnan, China. Zool Res 2016; 37:361-365. [PMID: 28105801 PMCID: PMC5359324 DOI: 10.13918/j.issn.2095-8137.2016.6.361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jia-Li Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China.
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China.
| | - Xu-Dong Zhao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Xin-Tian Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| |
Collapse
|
13
|
Mettler Izquierdo S, Varela S, Park M, Collarini EJ, Lu D, Pramanick S, Rucker J, Lopalco L, Etches R, Harriman W. High-efficiency antibody discovery achieved with multiplexed microscopy. Microscopy (Oxf) 2016; 65:341-52. [PMID: 27107009 PMCID: PMC5895110 DOI: 10.1093/jmicro/dfw014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/09/2016] [Indexed: 12/16/2022] Open
Abstract
The analysis of secreted antibody from large and diverse populations of B cells in parallel at the clonal level can reveal desirable antibodies for diagnostic or therapeutic applications. By immobilizing B cells in microdroplets with particulate reporters, decoding and isolating them in a microscopy environment, we have recovered panels of antibodies with rare attributes to therapeutically relevant targets. The ability to screen up to 100 million cells in a single experiment can be fully leveraged by accessing primary B-cell populations from evolutionarily divergent species such as chickens.
Collapse
Affiliation(s)
| | | | | | | | - Daniel Lu
- Crystal Bioscience, Emeryville, CA, USA
| | | | | | - Lucia Lopalco
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | | | | |
Collapse
|
14
|
Meng W, Li L, Xiong W, Fan X, Deng H, Bett AJ, Chen Z, Tang A, Cox KS, Joyce JG, Freed DC, Thoryk E, Fu TM, Casimiro DR, Zhang N, A Vora K, An Z. Efficient generation of monoclonal antibodies from single rhesus macaque antibody secreting cells. MAbs 2016; 7:707-18. [PMID: 25996084 PMCID: PMC4622687 DOI: 10.1080/19420862.2015.1051440] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Nonhuman primates (NHPs) are used as a preclinical model for vaccine development, and the antibody profiles to experimental vaccines in NHPs can provide critical information for both vaccine design and translation to clinical efficacy. However, an efficient protocol for generating monoclonal antibodies from single antibody secreting cells of NHPs is currently lacking. In this study we established a robust protocol for cloning immunoglobulin (IG) variable domain genes from single rhesus macaque (Macaca mulatta) antibody secreting cells. A sorting strategy was developed using a panel of molecular markers (CD3, CD19, CD20, surface IgG, intracellular IgG, CD27, Ki67 and CD38) to identify the kinetics of B cell response after vaccination. Specific primers for the rhesus macaque IG genes were designed and validated using cDNA isolated from macaque peripheral blood mononuclear cells. Cloning efficiency was averaged at 90% for variable heavy (VH) and light (VL) domains, and 78.5% of the clones (n = 335) were matched VH and VL pairs. Sequence analysis revealed that diverse IGHV subgroups (for VH) and IGKV and IGLV subgroups (for VL) were represented in the cloned antibodies. The protocol was tested in a study using an experimental dengue vaccine candidate. About 26.6% of the monoclonal antibodies cloned from the vaccinated rhesus macaques react with the dengue vaccine antigens. These results validate the protocol for cloning monoclonal antibodies in response to vaccination from single macaque antibody secreting cells, which have general applicability for determining monoclonal antibody profiles in response to other immunogens or vaccine studies of interest in NHPs.
Collapse
Affiliation(s)
- Weixu Meng
- a Texas Therapeutics Institute; Brown Foundation Institute of Molecular Medicine; University of Texas Health Science Center at Houston ; Houston , TX , USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
A novel humanized antibody neutralizes H5N1 influenza virus via two different mechanisms. J Virol 2015; 89:3712-22. [PMID: 25609802 DOI: 10.1128/jvi.03014-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Highly pathogenic avian influenza virus subtype H5N1 continues to be a severe threat to public health, as well as the poultry industry, because of its high lethality and antigenic drift rate. Neutralizing monoclonal antibodies (MAbs) can serve as a useful tool for preventing, treating, and detecting H5N1. In the present study, humanized H5 antibody 8A8 was developed from a murine H5 MAb. Both the humanized and mouse MAbs presented positive activity in hemagglutination inhibition (HI), virus neutralization, and immunofluorescence assays against a wide range of H5N1 strains. Interestingly, both human and murine 8A8 antibodies were able to detect H5 in Western blot assays under reducing conditions. Further, by sequencing of escape mutants, the conformational epitope of 8A8 was found to be located within the receptor binding domain (RBD) of H5. The linear epitope of 8A8 was identified by Western blotting of overlapping fragments and substitution mutant forms of HA1. Reverse genetic H5N1 strains with individual mutations in either the conformational or the linear epitope were generated and characterized in a series of assays, including HI, postattachment, and cell-cell fusion inhibition assays. The results indicate that for 8A8, virus neutralization mediated by RBD blocking relies on the conformational epitope while binding to the linear epitope contributes to the neutralization by inhibiting membrane fusion. Taken together, the results of this study show that a novel humanized H5 MAb binds to two types of epitopes on HA, leading to virus neutralization via two mechanisms. IMPORTANCE Recurrence of the highly pathogenic avian influenza virus subtype H5N1 in humans and poultry continues to be a serious public health concern. Preventive and therapeutic measures against influenza A viruses have received much interest in the context of global efforts to combat the current and future pandemics. Passive immune therapy is considered to be the most effective and economically prudent preventive strategy against influenza virus besides vaccination. It is important to develop a humanized neutralizing monoclonal antibody (MAb) against all of the clades of H5N1. For the first time, we report in this study that a novel humanized H5 MAb binds to two types of epitopes on HA, leading to virus neutralization via two mechanisms. These findings further deepen our understanding of influenza virus neutralization.
Collapse
|
16
|
Mak TM, Hanson BJ, Tan YJ. Chimerization and characterization of a monoclonal antibody with potent neutralizing activity across multiple influenza A H5N1 clades. Antiviral Res 2014; 107:76-83. [PMID: 24797696 DOI: 10.1016/j.antiviral.2014.04.011] [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: 12/17/2013] [Revised: 04/21/2014] [Accepted: 04/22/2014] [Indexed: 01/06/2023]
Abstract
The persistent evolution and circulation of highly pathogenic avian influenza H5N1 viruses pose a serious threat to global heath and hamper pandemic preparedness through conventional vaccine strategies. Combination passive immunotherapy using non-competing neutralizing antibodies has been proposed as a viable alternative to provide broad protection against drift variants. This necessitates the pre-pandemic production and characterization of potently neutralizing monoclonal antibodies (MAbs). One such antibody, MAb 9F4 was shown to provide heterologous protection against multiple H5N1 clade viruses, including one of the recently designated subclades, namely 2.3.4, through binding to a novel epitope, warranting its further development and characterization as a therapeutic candidate. In this study, the conversion of MAb 9F4 from mouse IgG2b to mouse-human chimeric (xi) IgG1 and IgA1 was achieved. These chimeric MAb versions were found to retain high degrees of binding and neutralizing activity against H5N1. The demonstration that xi-IgA1-9F4 retains a fairly high level of neutralizing activity, which is ∼10-fold lower than the corresponding xi-IgG1 isotype, suggests that this MAb could be further developed and engineered for intranasal administration.
Collapse
Affiliation(s)
- Tze-Minn Mak
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Brendon J Hanson
- Defence Medical and Environmental Research Institute, DSO National Laboratories, Singapore
| | - Yee-Joo Tan
- Infrastructure, Technology and Translational Division, Institute of Molecular and Cell Biology, A∗STAR, Singapore; Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore.
| |
Collapse
|
17
|
Rinaldi C, Penhale WJ, Stumbles PA, Tay G, Berry CM. Modulation of innate immune responses by influenza-specific ovine polyclonal antibodies used for prophylaxis. PLoS One 2014; 9:e89674. [PMID: 24586955 PMCID: PMC3938480 DOI: 10.1371/journal.pone.0089674] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 01/21/2014] [Indexed: 11/18/2022] Open
Abstract
In the event of a novel influenza A virus pandemic, prophylaxis mediated by antibodies provides an adjunct control option to vaccines and antivirals. This strategy is particularly pertinent to unvaccinated populations at risk during the lag time to produce and distribute an effective vaccine. Therefore, development of effective prophylactic therapies is of high importance. Although previous approaches have used systemic delivery of monoclonal antibodies or convalescent sera, available supply is a serious limitation. Here, we have investigated intranasal delivery of influenza-specific ovine polyclonal IgG antibodies for their efficacy against homologous influenza virus challenge in a mouse model. Both influenza-specific IgG and F(ab')2 reduced clinical scores, body weight loss and lung viral loads in mice treated 1 hour before virus exposure. Full protection from disease was also observed when antibody was delivered up to 3 days prior to virus infection. Furthermore, effective prophylaxis was independent of a strong innate immune response. This strategy presents a further option for prophylactic intervention against influenza A virus using ruminants to generate a bulk supply that could potentially be used in a pandemic setting, to slow virus transmission and reduce morbidity associated with a high cytokine phenotype.
Collapse
Affiliation(s)
- Catherine Rinaldi
- Centre for Forensic Science, The University of Western Australia, Nedlands, Western Australia, Australia
| | - William J. Penhale
- Molecular and Biomedical Sciences, School of Veterinary and Life Sciences, Murdoch University, Murdoch, Western Australia, Australia
| | - Philip A. Stumbles
- Molecular and Biomedical Sciences, School of Veterinary and Life Sciences, Murdoch University, Murdoch, Western Australia, Australia
| | - Guan Tay
- Centre for Forensic Science, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Cassandra M. Berry
- Molecular and Biomedical Sciences, School of Veterinary and Life Sciences, Murdoch University, Murdoch, Western Australia, Australia
- * E-mail:
| |
Collapse
|
18
|
Xiao L, Wang D, Sun C, Li P, Jin Y, Feng L, Chen L. Enhancement of SIV-specific cell mediated immune responses by co-administration of soluble PD-1 and Tim-3 as molecular adjuvants in mice. Hum Vaccin Immunother 2013; 10:724-33. [PMID: 24326266 DOI: 10.4161/hv.27340] [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] [Indexed: 11/19/2022] Open
Abstract
The development of an effective T cell based HIV vaccine would need to elicit cell mediated immune responses with superior magnitude, breadth, and quality. Since blocking the interactions between inhibitory receptors with their associated ligands using soluble PD-1 (sPD-1) and soluble Tim-3 (sTim-3) have been shown to reverse T cell exhaustion and enhance cell mediated immune responses, we tested if co-administration of sPD-1 and sTim-3 with an adenovirus vectored SIV vaccine (rAd5-SIV) can enhance cell mediated immune responses. The frequency of SIV antigen specific IFN-γ spot-forming cells and the secretion of IFN-γ and TNF-α by splenocytes from rAd5-SIV immunized mice was significantly increased when stimulated ex vivo with SIV peptides in the presence of sPD-1 or sTim-3 or both sPD-1 and sTim-3. The magnitude of cell mediated immune responses elicited by rAd5-SIV was enhanced by co-administration of sPD-1 and sTim-3. Co-administration of both sPD-1 and sTim-3 induced higher frequency of SIV antigen specific IFN-γ(+) spot-forming cells to poorly immunogenic Vif and Tat. The percentage of cell mediated responses for each SIV antigen became more balanced, with reduction to Gag but induction to non-structural proteins. Furthermore, co-injection of rAd5-sPD1 and rAd5-sTim3 with rAd5-SIV in mice enhanced T cell proliferation capability and generated more antigen specific IFN-γ(+) CD4(+) and CD8(+) T cells. Our study provided a new approach to enhance vaccine induced cell mediated immune responses, which may be applicable to improve the efficacy of vaccines against SIV/HIV.
Collapse
Affiliation(s)
- Lijun Xiao
- State Key Laboratory of Respiratory Diseases; Guangzhou Institutes of Biomedicine and Health (GIBH); Chinese Academy of Sciences; Guangzhou, PR China; University of Chinese Academy of Sciences; Beijing, PR China
| | | | | | | | | | | | | |
Collapse
|